1
|
Darley R, Illing PT, Duriez P, Bailey A, Purcell AW, van Hateren A, Elliott T. Evidence of focusing the MHC class I immunopeptidome by tapasin. Front Immunol 2025; 16:1563789. [PMID: 40406141 PMCID: PMC12094946 DOI: 10.3389/fimmu.2025.1563789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Major Histocompatibility Complex class I (MHC-I) molecules bind and present peptides to cytotoxic T cells, protecting against pathogens and cancer. MHC-I is highly polymorphic and each allotype is promiscuous, and capable of binding a unique and diverse repertoire of peptide ligands. The peptide editing chaperone tapasin optimizes this allotype specific repertoire of peptides, resulting in the selection of high affinity peptides. MHC-I allotypes differ in the extent they engage tapasin. This suggests that tapasin-dependent MHC-I allotypes should present a less diverse repertoire that is enriched in higher-affinity peptides, and which are present in higher abundance, than tapasin independent MHC-I allotypes, which should present a broader repertoire containing peptides with a lower average affinity. Experimental verification of this hypothesis has been confounded by the different peptide binding specificities of MHC-I allotypes. Here, we independently investigated the peptide focusing function of tapasin by introducing a point mutation into a tapasin independent MHC-I allotype that dramatically increased its tapasin dependence without substantially altering its peptide binding specificity. This allowed us to demonstrate ligand focusing by tapasin at both the repertoire level in cellulo, and by using an in vitro system in which tapasin was artificially tethered to MHC-I, at the individual peptide level. We found that tapasin had a greater influence on tapasin dependent MHC-I molecules, and that tapasin modulated peptide selection according to peptide-MHC-I complex stability, disfavoring short-lived peptide-MHC-I complexes. Thus, tapasin dependent MHC-I molecules experience greater tapasin filtering, resulting in less diverse MHC-I immunopeptidomes that are enriched in high affinity peptide-MHC-I complexes.
Collapse
Affiliation(s)
- Rachel Darley
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Patricia T. Illing
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Patrick Duriez
- Cancer Research UK Protein Core Facility, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Alistair Bailey
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology and Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Andy van Hateren
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Tim Elliott
- Centre for Immuno-Oncology and Chinese Academy of Medical Sciences (CAMS)-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
2
|
Coppola J, Parren M, Bastidas R, Saye-Francisco K, Malvin J, Jardine JG, Gilbride RM, Ojha S, Feltham S, Morrow D, Barber-Axthelm A, Bochart R, Fast R, Oswald K, Shoemaker R, Lifson JD, Picker LJ, Burton DR, Hansen SG. Combining a rhesus cytomegalovirus/SIV vaccine with neutralizing antibody to protect against SIV challenge in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644395. [PMID: 40196580 PMCID: PMC11974736 DOI: 10.1101/2025.03.20.644395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
A vaccine is widely regarded as necessary for the control of the HIV pandemic and eventual eradication of AIDS. Neutralizing antibodies and MHC-E-restricted CD8+ T cells have both been shown capable of vaccine protection against the simian counterpart of HIV, SIV, in rhesus macaques. Here we provide preliminary evidence that combining these orthogonal antiviral mechanisms can provide increased protection against SIV challenge such that replication arrest observed following vaccination with a rhesus cytomegalovirus (RhCMV/SIV)-based vaccine was enhanced in the presence of passively administered incompletely protective levels of neutralizing antibody. The report invites studies involving larger cohorts of macaques and alternate routes of providing neutralizing antibody.
Collapse
|
3
|
Zhang H, Xia M, Li H, Zeng X, Jia H, Zhang W, Zhou J. Implication of Immunobiological Function of Melanocytes in Dermatology. Clin Rev Allergy Immunol 2025; 68:30. [PMID: 40097884 DOI: 10.1007/s12016-025-09040-7] [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] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Melanocytes are essential for regulating pigmentation and providing photoprotection in human skin. Originating from neural crest cells, these cells migrate to the basal layer of the epidermis and hair follicles during embryogenesis. Melanosomes, the specialized, membrane-bound organelles are essential for melanin synthesis. Beyond their role in pigmentation, melanocytes exhibit complex immune functions, expressing a variety of immune-related markers and receptors, such as pattern recognition receptors (PRRs), major histocompatibility complex class II (MHC-II) molecules, CD40, intercellular adhesion molecule 1 (ICAM-1), and programmed death-ligand 1 (PD-L1). These receptors allow melanocytes to detect environmental signals and engage in the innate immune response. Furthermore, melanocytes release various immunomodulatory substances, including proinflammatory cytokines, chemokines, and damage-associated molecular patterns (DAMPs), contributing to immune regulation. The immune functions of melanocytes are significantly influenced by external factors such as ultraviolet radiation (UVR), the microbiome, and oxidative stress. In different skin diseases, these immune functions may vary. For example, vitiligo, a common hypopigmentary disorder, is primarily driven by an autoimmune response targeting melanocytes, giving rise to depigmentation and the appearance of white patches. In contrast, melanoma, a form of skin cancer that arises from melanocytes, is closely linked to UV exposure. This review highlights the diverse immunobiological functions of melanocytes and their implications in dermatology.
Collapse
Affiliation(s)
- Hejuan Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Maomei Xia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hongyang Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Xuesi Zeng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Hong Jia
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Wei Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.
| | - Jia Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
4
|
Hansen SG, Schell JB, Marshall EE, Ojha S, Feltham S, Morrow D, Hughes CM, Gilbride RM, Ford JC, Cleveland-Rubeor HC, McArdle MR, Whitmer T, Barber-Axthelm A, Bochart R, Smedley J, Oswald K, Fast R, Shoemaker R, Kosmider E, Edlefsen PT, Lifson JD, Malouli D, Früh K, Picker LJ. Glycoprotein L-deleted single-cycle rhesus cytomegalovirus vectors elicit MHC-E-restricted CD8+ T cells that protect against SIV. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf104. [PMID: 40420384 DOI: 10.1093/jimmun/vkaf104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/28/2025]
Abstract
Strain 68-1 rhesus CMV (RhCMV) vectors induce immune responses that mediate early, complete replication arrest of SIV infection in ∼60% of vaccinated rhesus macaques (RMs). This unique efficacy depends on the ability of these vectors to elicit effector memory (EM)-biased CD8+ T cells recognizing SIV peptides presented by MHC-E, rather than MHC-Ia. These efficacious responses still occurred when spread of the 68-1 vector was impaired by deletion of the viral anti-host intrinsic immunity factor phosphoprotein 71 (pp71), but efficacy was lost with a more stringent attenuation strategy based on destabilization of Rh108, the ortholog of the essential human CMV (HCMV) transcription factor UL79 that is required for late viral gene expression. Although unable to produce infectious progeny (ie single-cycle infection), Rh108-deficient vectors elicited durable, high frequency, EM-biased, SIV-specific CD8+ T-cell responses in RMs, but these responses were MHC-Ia-restricted and therefore non-efficacious. Here, we tested a different single-cycle attenuation strategy based on deletion (Δ) of the glycoprotein L (gL) that is essential for viral entry but allows for late gene expression and viral assembly. ΔgL 68-1 RhCMV/SIV vectors, grown on gL-complementing fibroblasts, were robustly immunogenic at doses above 105 PFU, generating high frequency, EM-biased, SIV-specific CD8+ T-cell responses that were also unconventionally restricted, including the MHC-E restriction associated with efficacy. Indeed, these single-cycle vectors manifested replication arrest efficacy in 70% of vaccinated RMs, further linking MHC-E restriction with efficacy, and demonstrating that 68-1 RhCMV/SIV efficacy does not require vector dissemination within the host.
Collapse
Affiliation(s)
- Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - John B Schell
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Emily E Marshall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Sohita Ojha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Shana Feltham
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Hilary C Cleveland-Rubeor
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Travis Whitmer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Rachelle Bochart
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Jeremy Smedley
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Kelli Oswald
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Randy Fast
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ewelina Kosmider
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, SAIC Frederick, Inc., Frederick National Laboratory, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| |
Collapse
|
5
|
Bruton J, Hanke T. Exploitation of Unconventional CD8 T-Cell Responses Induced by Engineered Cytomegaloviruses for the Development of an HIV-1 Vaccine. Vaccines (Basel) 2025; 13:72. [PMID: 39852851 PMCID: PMC11769474 DOI: 10.3390/vaccines13010072] [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: 11/27/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025] Open
Abstract
After four decades of intensive research, traditional vaccination strategies for HIV-1 remain ineffective due to HIV-1's extraordinary genetic diversity and complex immune evasion mechanisms. Cytomegaloviruses (CMV) have emerged as a novel type of vaccine vector with unique advantages due to CMV persistence and immunogenicity. Rhesus macaques vaccinated with molecular clone 68-1 of RhCMV (RhCMV68-1) engineered to express simian immunodeficiency virus (SIV) immunogens elicited an unconventional major histocompatibility complex class Ib allele E (MHC-E)-restricted CD8+ T-cell response, which consistently protected over half of the animals against a highly pathogenic SIV challenge. The RhCMV68-1.SIV-induced responses mediated a post-infection replication arrest of the challenge virus and eventually cleared it from the body. These observations in rhesus macaques opened a possibility that MHC-E-restricted CD8+ T-cells could achieve similar control of HIV-1 in humans. The potentially game-changing advantage of the human CMV (HCMV)-based vaccines is that they would induce protective CD8+ T-cells persisting at the sites of entry that would be insensitive to HIV-1 evasion. In the RhCMV68-1-protected rhesus macaques, MHC-E molecules and their peptide cargo utilise complex regulatory mechanisms and unique transport patterns, and researchers study these to guide human vaccine development. However, CMVs are highly species-adapted viruses and it is yet to be shown whether the success of RhCMV68-1 can be translated into an HCMV ortholog for humans. Despite some safety concerns regarding using HCMV as a vaccine vector in humans, there is a vision of immune programming of HCMV to induce pathogen-tailored CD8+ T-cells effective against HIV-1 and other life-threatening diseases.
Collapse
Affiliation(s)
- Joseph Bruton
- Hertford College, University of Oxford, Oxford OX1 3BW, UK;
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
6
|
Gillespie GM, Quastel MN, McMichael AJ. HLA-E: Immune Receptor Functional Mechanisms Revealed by Structural Studies. Immunol Rev 2025; 329:e13434. [PMID: 39753525 PMCID: PMC11698700 DOI: 10.1111/imr.13434] [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: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025]
Abstract
HLA-E is a nonclassical, nonpolymorphic, class Ib HLA molecule. Its primary function is to present a conserved nonamer peptide, termed VL9, derived from the signal sequence of classical MHC molecules to the NKG2x-CD94 receptors on NK cells and a subset of T lymphocytes. These receptors regulate the function of NK cells, and the importance of this role, which is conserved across mammalian species, probably accounts for the lack of genetic polymorphism. A second minor function is to present other, weaker binding, pathogen-derived peptides to T lymphocytes. Most of these peptides bind suboptimally to HLA-E, but this binding appears to be enabled by the relative stability of peptide-free, but receptive, HLA-E-β2m complexes. This, in turn, may favor nonclassical antigen processing that may be associated with bacteria infected cells. This review explores how the structure of HLA-E, bound to different peptides and then to NKG2-CD94 or T-cell receptors, relates to HLA-E cell biology and immunology. A detailed understanding of this molecule could open up opportunities for development of universal T-cell and NK-cell-based immunotherapies.
Collapse
MESH Headings
- Humans
- Histocompatibility Antigens Class I/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/chemistry
- Animals
- HLA-E Antigens
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Protein Binding
- Antigen Presentation
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/chemistry
- NK Cell Lectin-Like Receptor Subfamily C/metabolism
- Structure-Activity Relationship
- Peptides/chemistry
- Peptides/immunology
- Peptides/metabolism
- NK Cell Lectin-Like Receptor Subfamily D/metabolism
- NK Cell Lectin-Like Receptor Subfamily D/chemistry
- NK Cell Lectin-Like Receptor Subfamily D/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Protein Conformation
Collapse
Affiliation(s)
| | - Max N. Quastel
- Nuffield Department of Medicine, Center for Immuno‐OncologyUniversity of OxfordOxfordUK
| | - Andrew J. McMichael
- Nuffield Department of Medicine, Center for Immuno‐OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
7
|
Tsao HW, Anderson S, Finn KJ, Perera JJ, Pass LF, Schneider EM, Jiang A, Fetterman R, Chuong CL, Kozuma K, Stickler MM, Creixell M, Klaeger S, Phulphagar KM, Rachimi S, Verzani EK, Olsson N, Dubrot J, Pech MF, Silkworth W, Lane-Reticker SK, Allen PM, Ibrahim K, Knudsen NH, Cheng AY, Long AH, Ebrahimi-Nik H, Kim SY, Du PP, Iracheta-Vellve A, Robitschek EJ, Suermondt JSMT, Davis TGR, Wolfe CH, Atluri T, Olander KE, Rush JS, Sundberg TB, McAllister FE, Abelin JG, Firestone A, Stokoe D, Carr SA, Harding FA, Yates KB, Manguso RT. Targeting the aminopeptidase ERAP enhances antitumor immunity by disrupting the NKG2A-HLA-E inhibitory checkpoint. Immunity 2024; 57:2863-2878.e12. [PMID: 39561763 DOI: 10.1016/j.immuni.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/12/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024]
Abstract
The aminopeptidase, endoplasmic reticulum aminopeptidase 1 (ERAP1), trims peptides for loading into major histocompatibility complex class I (MHC class I), and loss of this activity has broad effects on the MHC class I peptidome. Here, we investigated the impact of targeting ERAP1 in immune checkpoint blockade (ICB), as MHC class I interactions mediate both activating and inhibitory functions in antitumor immunity. Loss of ERAP sensitized mouse tumor models to ICB, and this sensitivity depended on CD8+ T cells and natural killer (NK) cells. In vivo suppression screens revealed that Erap1 deletion inactivated the inhibitory NKG2A-HLA-E checkpoint, which requires presentation of a restricted set of invariant epitopes (VL9) on HLA-E. Loss of ERAP altered the HLA-E peptidome, preventing NKG2A engagement. In humans, ERAP1 and ERAP2 showed functional redundancy for the processing and presentation of VL9, and loss of both inactivated the NKG2A checkpoint in cancer cells. Thus, loss of ERAP phenocopies the inhibition of the NKG2A-HLA-E pathway and represents an attractive approach to inhibit this critical checkpoint.
Collapse
Affiliation(s)
- Hsiao-Wei Tsao
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Seth Anderson
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | | | - Jonathan J Perera
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Lomax F Pass
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Emily M Schneider
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Aiping Jiang
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Rachel Fetterman
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Cun Lan Chuong
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Kaiya Kozuma
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - Suzanna Rachimi
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Eva K Verzani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - Juan Dubrot
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Sarah Kate Lane-Reticker
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Peter M Allen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Kyrellos Ibrahim
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Nelson H Knudsen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew Y Cheng
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Adrienne H Long
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hakimeh Ebrahimi-Nik
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Sarah Y Kim
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Peter P Du
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Arvin Iracheta-Vellve
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Emily J Robitschek
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Juliette S M T Suermondt
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas G R Davis
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Clara H Wolfe
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Trisha Atluri
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Kira E Olander
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Jason S Rush
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Thomas B Sundberg
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - Jennifer G Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - David Stokoe
- Calico Life Sciences, South San Francisco, CA, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | | | - Kathleen B Yates
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Robert T Manguso
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA; Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
Malouli D, Taher H, Mansouri M, Iyer RF, Reed J, Papen C, Schell JB, Beechwood T, Martinson T, Morrow D, Hughes CM, Gilbride RM, Randall K, Ford JC, Belica K, Ojha S, Sacha JB, Bimber BN, Hansen SG, Picker LJ, Früh K. Human cytomegalovirus UL18 prevents priming of MHC-E- and MHC-II-restricted CD8 + T cells. Sci Immunol 2024; 9:eadp5216. [PMID: 39392895 PMCID: PMC11797217 DOI: 10.1126/sciimmunol.adp5216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024]
Abstract
Rhesus cytomegalovirus (RhCMV) vectors elicit major histocompatibility complex (MHC)-E-restricted CD8+ T cells that stringently control simian immunodeficiency virus (SIV) in rhesus macaques. These responses require deletion of eight RhCMV chemokine-like open reading frames (ORFs) that are conserved in human cytomegalovirus (HCMV). To determine whether HCMV encodes additional, nonconserved inhibitors of unconventional T cell priming, we inserted 41 HCMV-specific ORFs into a chemokine-deficient strain (68-1 RhCMV). Monitoring of epitope recognition revealed that HCMV UL18 prevented unconventional T cell priming, resulting in MHC-Ia-targeted responses. UL18 is homologous to MHC-I but does not engage T cell receptors and, instead, binds with high affinity to inhibitory leukocyte immunoglobulin-like receptor-1 (LIR-1). UL18 lacking LIR-1 binding no longer interfered with MHC-E-restricted T cell stimulation by RhCMV-infected cells or the induction of unconventionally restricted T cells. Thus, LIR-1 binding needs to be deleted from UL18 of HCMV/HIV vaccines to allow for the induction of protective MHC-E-restricted T cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
9
|
Voogd L, van Wolfswinkel M, Satti I, White AD, Dijkman K, Gela A, van Meijgaarden KE, Franken KLMC, Marshall JL, Ottenhoff THM, Scriba TJ, McShane H, Sharpe SA, Verreck FAW, Joosten SA. Mtb-Specific HLA-E-Restricted T Cells Are Induced during Mtb Infection but Not after BCG Administration in Non-Human Primates and Humans. Vaccines (Basel) 2024; 12:1129. [PMID: 39460296 PMCID: PMC11511431 DOI: 10.3390/vaccines12101129] [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: 08/23/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Novel vaccines targeting the world's deadliest pathogen Mycobacterium tuberculosis (Mtb) are urgently needed as the efficacy of the Bacillus Calmette-Guérin (BCG) vaccine in its current use is limited. HLA-E is a virtually monomorphic unconventional antigen presentation molecule, and HLA-E-restricted Mtb-specific CD8+ T cells can control intracellular Mtb growth, making HLA-E a promising vaccine target for Mtb. Methods: In this study, we evaluated the frequency and phenotype of HLA-E-restricted Mtb-specific CD4+/CD8+ T cells in the circulation and bronchoalveolar lavage fluid of two independent non-human primate (NHP) studies and from humans receiving BCG either intradermally or mucosally. Results: BCG vaccination followed by Mtb challenge in NHPs did not affect the frequency of circulating and local HLA-E-Mtb CD4+ and CD8+ T cells, and we saw the same in humans receiving BCG. HLA-E-Mtb T cell frequencies were significantly increased after Mtb challenge in unvaccinated NHPs, which was correlated with higher TB pathology. Conclusions: Together, HLA-E-Mtb-restricted T cells are minimally induced by BCG in humans and rhesus macaques (RMs) but can be elicited after Mtb infection in unvaccinated RMs. These results give new insights into targeting HLA-E as a potential immune mechanism against TB.
Collapse
Affiliation(s)
- Linda Voogd
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marjolein van Wolfswinkel
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Andrew D. White
- UK Health Security Agency, Porton Down, Wiltshire SP4 0JG, UK
| | - Karin Dijkman
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Anele Gela
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Krista E. van Meijgaarden
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Kees L. M. C. Franken
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Tom H. M. Ottenhoff
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town 7925, South Africa
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Sally A. Sharpe
- UK Health Security Agency, Porton Down, Wiltshire SP4 0JG, UK
| | | | - Simone A. Joosten
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
10
|
Brackenridge S, John N, He W, Früh K, Borrow P, McMichael A. Regulation of the cell surface expression of classical and non-classical MHC proteins by the human cytomegalovirus UL40 and rhesus cytomegalovirus Rh67 proteins. J Virol 2024; 98:e0120624. [PMID: 39207137 PMCID: PMC11406984 DOI: 10.1128/jvi.01206-24] [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: 07/12/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The signal sequences of the human cytomegalovirus (CMV) UL40 protein and its rhesus CMV (RhCMV) counterpart, Rh67, contain a peptide (VMAPRT[L/V][F/I/L/V]L, VL9) that is presented by major histocompatibility complex (MHC) antigen E (MHC-E). The CMV VL9 peptides replace VL9 peptides derived from classical MHC (Ia) signal sequences, which are lost when CMV disrupts antigen processing and presentation and MHC Ia expression. This allows infected cells to maintain MHC-E surface expression and escape killing by Natural Killer cells. We demonstrate that processing of the Rh67 VL9 peptide mirrors that of UL40, despite the lack of sequence conservation between the two proteins. Processing of both VL9 peptides is dependent on cleavage of their signal sequences by the host protease signal peptide peptidase. As previously shown for UL40, up-regulation of MHC-E expression by Rh67 requires only its signal sequence, with sequences upstream of VL9 critical for conferring independence from TAP, the transporter associated with antigen processing. Our results also suggest that the mature UL40 and Rh67 proteins contribute to CMV immune evasion by decreasing surface expression of MHC Ia. Unexpectedly, while the Rh67 VL9 peptide is resistant to the effects of Rh67, UL40 can partially counteract the up-regulation of MHC-E expression mediated by its own VL9 peptide. This suggests differences in the mechanisms by which the two VL9 peptides up-regulate MHC-E, and further work will be required to determine if any such differences have implications for translating a RhCMV-vectored simian immunodeficiency virus (SIV) vaccine to HIV-1 using human CMV as a vector. IMPORTANCE The protective immune response induced by a rhesus cytomegalovirus (RhCMV)-vectored simian immunodeficiency virus (SIV) vaccine in rhesus macaques depends on the presence of the viral Rh67 gene in the vaccine. The Rh67 protein contains a peptide that allows the RhCMV-infected cells to maintain expression of major histocompatibility complex (MHC) antigen E at the cell surface. We show that production of this peptide, referred to as "VL9," mirrors that of the equivalent peptide present in the human cytomegalovirus (CMV) protein UL40, despite the little sequence similarity between the two CMV proteins. We also show that the mature UL40 and Rh67 proteins, which have no previously described function, also contribute to CMV immune evasion by reducing cell surface expression of MHC proteins important for the immune system to detect infected cells. Despite these similarities, our work also reveals possible differences between Rh67 and UL40, and these may have implications for the use of human CMV as the vector for a potential HIV-1 vaccine.
Collapse
Affiliation(s)
- Simon Brackenridge
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nessy John
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Wanlin He
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Klaus Früh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Persephone Borrow
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew McMichael
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Ahmed S, Herschhorn A. mRNA-based HIV-1 vaccines. Clin Microbiol Rev 2024; 37:e0004124. [PMID: 39016564 PMCID: PMC11391700 DOI: 10.1128/cmr.00041-24] [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] [Indexed: 07/18/2024] Open
Abstract
SUMMARYThe success of the Severe Acute Respiratory Syndrome Coronavirus 2 mRNA vaccines to lessen/prevent severe COVID-19 opened new opportunities to develop RNA vaccines to fight other infectious agents. HIV-1 is a lentivirus that integrates into the host cell genome and persists for the lifetime of infected cells. Multiple mechanisms of immune evasion have posed significant obstacles to the development of an effective HIV-1 vaccine over the last four decades since the identification of HIV-1. Recently, attempts to address some of these challenges have led to multiple studies that manufactured, optimized, and tested, in different animal models, mRNA-based HIV-1 vaccines. Several clinical trials have also been initiated or are planned to start soon. Here, we review the current strategies applied to HIV-1 mRNA vaccines, discuss different targeting approaches, summarize the latest findings, and offer insights into the challenges and future of HIV-1 mRNA vaccines.
Collapse
Affiliation(s)
- Shamim Ahmed
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alon Herschhorn
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Genome Engineering, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
- Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, Minnesota, USA
- The College of Veterinary Medicine Graduate Program, University of Minnesota, Minneapolis, Minnesota, USA
- Molecular Pharmacology and Therapeutics Graduate Program, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
12
|
Al-Talib M, Dimonte S, Humphreys IR. Mucosal T-cell responses to chronic viral infections: Implications for vaccine design. Cell Mol Immunol 2024; 21:982-998. [PMID: 38459243 PMCID: PMC11364786 DOI: 10.1038/s41423-024-01140-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/31/2024] [Indexed: 03/10/2024] Open
Abstract
Mucosal surfaces that line the respiratory, gastrointestinal and genitourinary tracts are the major interfaces between the immune system and the environment. Their unique immunological landscape is characterized by the necessity of balancing tolerance to commensal microorganisms and other innocuous exposures against protection from pathogenic threats such as viruses. Numerous pathogenic viruses, including herpesviruses and retroviruses, exploit this environment to establish chronic infection. Effector and regulatory T-cell populations, including effector and resident memory T cells, play instrumental roles in mediating the transition from acute to chronic infection, where a degree of viral replication is tolerated to minimize immunopathology. Persistent antigen exposure during chronic viral infection leads to the evolution and divergence of these responses. In this review, we discuss advances in the understanding of mucosal T-cell immunity during chronic viral infections and how features of T-cell responses develop in different chronic viral infections of the mucosa. We consider how insights into T-cell immunity at mucosal surfaces could inform vaccine strategies: not only to protect hosts from chronic viral infections but also to exploit viruses that can persist within mucosal surfaces as vaccine vectors.
Collapse
Affiliation(s)
- Mohammed Al-Talib
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
- Bristol Medical School, University of Bristol, 5 Tyndall Avenue, Bristol, BS8 1UD, UK
| | - Sandra Dimonte
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Ian R Humphreys
- Systems Immunity University Research Institute/Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK.
| |
Collapse
|
13
|
Voogd L, van Wolfswinkel M, Satti I, White AD, Dijkman K, Gela A, van Meijgaarden KE, Franken KL, Marshall JL, Ottenhoff TH, Scriba TJ, McShane H, Sharpe SA, Verreck FA, Joosten SA. Mtb specific HLA-E restricted T cells are induced during Mtb infection but not after BCG administration in non-human primates and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609630. [PMID: 39253433 PMCID: PMC11383316 DOI: 10.1101/2024.08.26.609630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Novel vaccines targeting the world's deadliest pathogen Mycobacterium tuberculosis (Mtb) are urgently needed as the efficacy of the Bacillus Calmette-Guérin (BCG) vaccine in its current use is limited. HLA-E is a virtually monomorphic unconventional antigen presentation molecule and HLA-E restricted Mtb specific CD8+ T cells can control intracellular Mtb growth, making HLA-E a promising vaccine target for Mtb. In this study, we evaluated the frequency and phenotype of HLA-E restricted Mtb specific CD4+/CD8+ T cells in the circulation and bronchoalveolar lavage fluid of two independent non-human primate (NHP) studies and from humans receiving BCG either intradermally or mucosally. BCG vaccination followed by Mtb challenge in NHPs did not affect the frequency of circulating and local HLA-E/Mtb CD4+ and CD8+ T cells, and we saw the same in humans receiving BCG. HLA-E/Mtb T cell frequencies were significantly increased after Mtb challenge in unvaccinated NHPs, which was correlated with higher TB pathology. Together, HLA-E/Mtb restricted T cells are minimally induced by BCG in humans and rhesus macaques (RMs) but can be elicited after Mtb infection in unvaccinated RMs. These results give new insights into targeting HLA-E as a potential immune mechanism against TB.
Collapse
Affiliation(s)
- Linda Voogd
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein van Wolfswinkel
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford, United Kingdom (UK)
| | | | - Karin Dijkman
- Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Anele Gela
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Krista E. van Meijgaarden
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees L.M.C. Franken
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Julia L. Marshall
- The Jenner Institute, University of Oxford, Oxford, United Kingdom (UK)
| | - Tom H.M. Ottenhoff
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Diseases and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, United Kingdom (UK)
| | | | | | - Simone A. Joosten
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Bimber BN, Sunshine J, McElfresh GW, Reed JS, Pathak R, Bateman KB, Hughes CM, Gilbride RM, Ford JC, Morrow D, Lifson JD, Sacha JB, Hansen SG, Picker LJ. Viral escape mutations do not account for non-protection from SIVmac239 challenge in RhCMV/SIV vaccinated rhesus macaques. Front Immunol 2024; 15:1444621. [PMID: 39170621 PMCID: PMC11336698 DOI: 10.3389/fimmu.2024.1444621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Simian immunodeficiency virus (SIV) vaccines based upon 68-1 Rhesus Cytomegalovirus (RhCMV) vectors show remarkable protection against pathogenic SIVmac239 challenge. Across multiple independent rhesus macaque (RM) challenge studies, nearly 60% of vaccinated RM show early, complete arrest of SIVmac239 replication after effective challenge, whereas the remainder show progressive infection similar to controls. Here, we performed viral sequencing to determine whether the failure to control viral replication in non-protected RMs is associated with the acquisition of viral escape mutations. While low level viral mutations accumulated in all animals by 28 days-post-challenge, which is after the establishment of viral control in protected animals, the dominant circulating virus in virtually all unprotected RMs was nearly identical to the challenge stock, and there was no difference in mutation patterns between this cohort and unvaccinated controls. These data definitively demonstrate that viral mutation does not explain lack of viral control in RMs not protected by RhCMV/SIV vaccination. We further demonstrate that during chronic infection RhCMV/SIV vaccinated RMs do not acquire escape mutation in epitopes targeted by RhCMV/SIV, but instead display mutation in canonical MHC-Ia epitopes similar to unvaccinated RMs. This suggests that after the initial failure of viral control, unconventional T cell responses induced by 68-1 RhCMV/SIV vaccination do not exert strong selective pressure on systemically replicating SIV.
Collapse
Affiliation(s)
- Benjamin N. Bimber
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Justine Sunshine
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - G. W. McElfresh
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Jason S. Reed
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Reese Pathak
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Katherine B. Bateman
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
| | - Colette M. Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Roxanne M. Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Julia C. Ford
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - David Morrow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD, United States
| | - Jonah B. Sacha
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Scott G. Hansen
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Louis J. Picker
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, United States
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| |
Collapse
|
15
|
Malouli D, Tiwary M, Gilbride RM, Morrow DW, Hughes CM, Selseth A, Penney T, Castanha P, Wallace M, Yeung Y, Midgett M, Williams C, Reed J, Yu Y, Gao L, Yun G, Treaster L, Laughlin A, Lundy J, Tisoncik-Go J, Whitmore LS, Aye PP, Schiro F, Dufour JP, Papen CR, Taher H, Picker LJ, Früh K, Gale M, Maness NJ, Hansen SG, Barratt-Boyes S, Reed DS, Sacha JB. Cytomegalovirus vaccine vector-induced effector memory CD4 + T cells protect cynomolgus macaques from lethal aerosolized heterologous avian influenza challenge. Nat Commun 2024; 15:6007. [PMID: 39030218 PMCID: PMC11272155 DOI: 10.1038/s41467-024-50345-6] [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: 02/19/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
An influenza vaccine approach that overcomes the problem of viral sequence diversity and provides long-lived heterosubtypic protection is urgently needed to protect against pandemic influenza viruses. Here, to determine if lung-resident effector memory T cells induced by cytomegalovirus (CMV)-vectored vaccines expressing conserved internal influenza antigens could protect against lethal influenza challenge, we immunize Mauritian cynomolgus macaques (MCM) with cynomolgus CMV (CyCMV) vaccines expressing H1N1 1918 influenza M1, NP, and PB1 antigens (CyCMV/Flu), and challenge with heterologous, aerosolized avian H5N1 influenza. All six unvaccinated MCM died by seven days post infection with acute respiratory distress, while 54.5% (6/11) CyCMV/Flu-vaccinated MCM survived. Survival correlates with the magnitude of lung-resident influenza-specific CD4 + T cells prior to challenge. These data demonstrate that CD4 + T cells targeting conserved internal influenza proteins can protect against highly pathogenic heterologous influenza challenge and support further exploration of effector memory T cell-based vaccines for universal influenza vaccine development.
Collapse
Affiliation(s)
- Daniel Malouli
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Meenakshi Tiwary
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Roxanne M Gilbride
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - David W Morrow
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Colette M Hughes
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Andrea Selseth
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Toni Penney
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Priscila Castanha
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Megan Wallace
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Yulia Yeung
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | | | - Connor Williams
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Jason Reed
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Yun Yu
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Lina Gao
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Gabin Yun
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luke Treaster
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Leanne S Whitmore
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Pyone P Aye
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Courtney R Papen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Husam Taher
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J Picker
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Scott G Hansen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | | | | | - Jonah B Sacha
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA.
| |
Collapse
|
16
|
Hwang JK, Marston DJ, Wrapp D, Li D, Tuyishime M, Brackenridge S, Rhodes B, Quastel M, Kapingidza AB, Gater J, Harner A, Wang Y, Rountree W, Ferrari G, Borrow P, McMichael AJ, Gillespie GM, Haynes BF, Azoitei ML. A high affinity monoclonal antibody against HLA-E-VL9 enhances natural killer cell anti-tumor killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602401. [PMID: 39026709 PMCID: PMC11257447 DOI: 10.1101/2024.07.08.602401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Natural killer (NK) cells kill target cells following triggering via germline-encoded receptors interacting with target cell-expressed ligands (direct killing), or via antibody-dependent cellular cytotoxicity (ADCC) mediated by FcγRIIIa. NK cytotoxicity is modulated by signaling through activating or inhibitory receptors. A major checkpoint is mediated by the NK inhibitory receptor NKG2A/CD94 and its target cell ligand, HLA-E, which is complexed with HLA signal sequence-derived peptides termed VL9 (HLA-E-VL9). We have previously reported the isolation of a murine HLA-E-VL9-specific IgM antibody 3H4 and the generation of a higher affinity IgG version (3H4v3). Here we have used phage display library selection to generate a high affinity version of 3H4v3, called 3H4v31, with an ∼700 fold increase in binding affinity. We show using an HLA-E-VL9+ K562 tumor model that, in vitro, the addition of 3H4v31 to target cells increased direct killing of targets by CD16-negative NK cell line NK-92 and also mediated ADCC by NK-92 cells transfected with CD16. Moreover, ADCC by primary NK cells was also enhanced in vitro by 3H4v31. 3H4v31 was also able to bind and enhance target cell lysis of endogenously expressed HLA-E-VL9 on human cervical cancer and human pancreatic cancer cell lines. In vivo, 3H4v31 slowed the growth rate of HLA-E-VL9+ K562 tumors implanted into NOD/SCID/IL2rγ null mice compared to isotype control when injected with NK-92 cells intratumorally. Together, these data demonstrate that mAb 3H4v31 can enhance NK cell killing of HLA-E-VL9-expressing tumor cells in vitro by both direct killing activity and by ADCC. Moreover, mAb 3H4v31 can enhance NK cell control of tumor growth in vivo. We thus identify HLA-E-VL9 monoclonal antibodies as a promising novel anti-tumor immunotherapy. One Sentence Summary A high affinity monoclonal antibody against HLA-E-VL9 enhances natural killer cell anti-tumor killing by checkpoint inhibition and antibody dependent cellular cytotoxicity.
Collapse
|
17
|
Iyer RF, Verweij MC, Nair SS, Morrow D, Mansouri M, Chakravarty D, Beechwood T, Meyer C, Uebelhoer L, Lauron EJ, Selseth A, John N, Thin TH, Dzedzik S, Havenar-Daughton C, Axthelm MK, Douglas J, Korman A, Bhardwaj N, Tewari AK, Hansen S, Malouli D, Picker LJ, Früh K. CD8 + T cell targeting of tumor antigens presented by HLA-E. SCIENCE ADVANCES 2024; 10:eadm7515. [PMID: 38728394 PMCID: PMC11086602 DOI: 10.1126/sciadv.adm7515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024]
Abstract
The nonpolymorphic major histocompatibility complex E (MHC-E) molecule is up-regulated on many cancer cells, thus contributing to immune evasion by engaging inhibitory NKG2A/CD94 receptors on NK cells and tumor-infiltrating T cells. To investigate whether MHC-E expression by cancer cells can be targeted for MHC-E-restricted T cell control, we immunized rhesus macaques (RM) with rhesus cytomegalovirus (RhCMV) vectors genetically programmed to elicit MHC-E-restricted CD8+ T cells and to express established tumor-associated antigens (TAAs) including prostatic acidic phosphatase (PAP), Wilms tumor-1 protein, or Mesothelin. T cell responses to all three tumor antigens were comparable to viral antigen-specific responses with respect to frequency, duration, phenotype, epitope density, and MHC restriction. Thus, CMV-vectored cancer vaccines can bypass central tolerance by eliciting T cells to noncanonical epitopes. We further demonstrate that PAP-specific, MHC-E-restricted CD8+ T cells from RhCMV/PAP-immunized RM respond to PAP-expressing HLA-E+ prostate cancer cells, suggesting that the HLA-E/NKG2A immune checkpoint can be exploited for CD8+ T cell-based immunotherapies.
Collapse
Affiliation(s)
- Ravi F. Iyer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Marieke C. Verweij
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sujit S. Nair
- Department of Urology and Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, NY 10029, USA
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Mandana Mansouri
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Dimple Chakravarty
- Department of Urology and Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, NY 10029, USA
| | - Teresa Beechwood
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | - Luke Uebelhoer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | - Andrea Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Tin Htwe Thin
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siarhei Dzedzik
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | - Alan Korman
- Vir Biotechnology, San Francisco, CA 14158, USA
| | - Nina Bhardwaj
- Department of Urology and Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, NY 10029, USA
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashutosh K. Tewari
- Department of Urology and Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, NY 10029, USA
| | - Scott Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| |
Collapse
|
18
|
Rückert T, Romagnani C. Extrinsic and intrinsic drivers of natural killer cell clonality. Immunol Rev 2024; 323:80-106. [PMID: 38506411 DOI: 10.1111/imr.13324] [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] [Indexed: 03/21/2024]
Abstract
Clonal expansion of antigen-specific lymphocytes is the fundamental mechanism enabling potent adaptive immune responses and the generation of immune memory. Accompanied by pronounced epigenetic remodeling, the massive proliferation of individual cells generates a critical mass of effectors for the control of acute infections, as well as a pool of memory cells protecting against future pathogen encounters. Classically associated with the adaptive immune system, recent work has demonstrated that innate immune memory to human cytomegalovirus (CMV) infection is stably maintained as large clonal expansions of natural killer (NK) cells, raising questions on the mechanisms for clonal selection and expansion in the absence of re-arranged antigen receptors. Here, we discuss clonal NK cell memory in the context of the mechanisms underlying clonal competition of adaptive lymphocytes and propose alternative selection mechanisms that might decide on the clonal success of their innate counterparts. We propose that the integration of external cues with cell-intrinsic sources of heterogeneity, such as variegated receptor expression, transcriptional states, and somatic variants, compose a bottleneck for clonal selection, contributing to the large size of memory NK cell clones.
Collapse
Affiliation(s)
- Timo Rückert
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| | - Chiara Romagnani
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Medical Immunology, Berlin, Germany
| |
Collapse
|
19
|
MacLachlan BJ, Sullivan LC, Brooks AG, Rossjohn J, Vivian JP. Structure of the murine CD94-NKG2A receptor in complex with Qa-1 b presenting an MHC-I leader peptide. FEBS J 2024; 291:1530-1544. [PMID: 38158698 DOI: 10.1111/febs.17050] [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: 10/23/2023] [Revised: 11/26/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
The heterodimeric natural killer cells antigen CD94 (CD94)-NKG2-A/NKG2-B type II integral membrane protein (NKG2A) receptor family expressed on human and mouse natural killer (NK) cells monitors global major histocompatibility complex (MHC) class I cell surface expression levels through binding to MHC class Ia-derived leader sequence peptides presented by HLA class I histocompatibility antigen, alpha chain E (HLA-E; in humans) or H-2 class I histocompatibility antigen, D-37 (Qa-1b; in mice). Although the molecular basis underpinning human CD94-NKG2A recognition of HLA-E is known, the equivalent interaction in the murine setting is not. By determining the high-resolution crystal structure of murine CD94-NKG2A in complex with Qa-1b presenting the Qa-1 determinant modifier peptide (QDM), we resolved the mode of binding. Compared to the human homologue, the murine CD94-NKG2A-Qa-1b-QDM displayed alterations in the distribution of interactions across CD94 and NKG2A subunits that coincide with differences in electrostatic complementarity of the ternary complex and the lack of cross-species reactivity. Nevertheless, we show that Qa-1b could be modified through W65R + N73I mutations to mimic HLA-E, facilitating binding with both human and murine CD94-NKG2A. These data underscore human and murine CD94-NKG2A cross-species heterogeneity and provide a foundation for humanising Qa-1b in immune system models.
Collapse
Affiliation(s)
- Bruce J MacLachlan
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Lucy C Sullivan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
- Institute of Infection and Immunity, School of Medicine, Cardiff University, UK
| | - Julian P Vivian
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
20
|
Brackenridge S, John N, Früh K, Borrow P, McMichael AJ. The antibodies 3D12 and 4D12 recognise distinct epitopes and conformations of HLA-E. Front Immunol 2024; 15:1329032. [PMID: 38571959 PMCID: PMC10987726 DOI: 10.3389/fimmu.2024.1329032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
The commonly used antibodies 3D12 and 4D12 recognise the human leukocyte antigen E (HLA-E) protein. These antibodies bind distinct epitopes on HLA-E and differ in their ability to bind alleles of the major histocompatibility complex E (MHC-E) proteins of rhesus and cynomolgus macaques. We confirmed that neither antibody cross-reacts with classical HLA alleles, and used hybrids of different MHC-E alleles to map the regions that are critical for their binding. 3D12 recognises a region on the alpha 3 domain, with its specificity for HLA-E resulting from the amino acids present at three key positions (219, 223 and 224) that are unique to HLA-E, while 4D12 binds to the start of the alpha 2 domain, adjacent to the C terminus of the presented peptide. 3D12 staining is increased by incubation of cells at 27°C, and by addition of the canonical signal sequence peptide presented by HLA-E peptide (VL9, VMAPRTLVL). This suggests that 3D12 may bind peptide-free forms of HLA-E, which would be expected to accumulate at the cell surface when cells are incubated at lower temperatures, as well as HLA-E with peptide. Therefore, additional studies are required to determine exactly what forms of HLA-E can be recognised by 3D12. In contrast, while staining with 4D12 was also increased when cells were incubated at 27°C, it was decreased when the VL9 peptide was added. We conclude that 4D12 preferentially binds to peptide-free HLA-E, and, although not suitable for measuring the total cell surface levels of MHC-E, may putatively identify peptide-receptive forms.
Collapse
Affiliation(s)
- Simon Brackenridge
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nessy John
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Klaus Früh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, United States
| | - Persephone Borrow
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew J. McMichael
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Voogd L, Drittij AM, Dingenouts CK, Franken KL, Unen VV, van Meijgaarden KE, Ruibal P, Hagedoorn RS, Leitner JA, Steinberger P, Heemskerk MH, Davis MM, Scriba TJ, Ottenhoff TH, Joosten SA. Mtb HLA-E-tetramer-sorted CD8 + T cells have a diverse TCR repertoire. iScience 2024; 27:109233. [PMID: 38439958 PMCID: PMC10909886 DOI: 10.1016/j.isci.2024.109233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/05/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
HLA-E molecules can present self- and pathogen-derived peptides to both natural killer (NK) cells and T cells. T cells that recognize HLA-E peptides via their T cell receptor (TCR) are termed donor-unrestricted T cells due to restricted allelic variation of HLA-E. The composition and repertoire of HLA-E TCRs is not known so far. We performed TCR sequencing on CD8+ T cells from 21 individuals recognizing HLA-E tetramers (TMs) folded with two Mtb-HLA-E-restricted peptides. We sorted HLA-E Mtb TM+ and TM- CD8+ T cells directly ex vivo and performed bulk RNA-sequencing and single-cell TCR sequencing. The identified TCR repertoire was diverse and showed no conservation between and within individuals. TCRs selected from our single-cell TCR sequencing data could be activated upon HLA-E/peptide stimulation, although not robust, reflecting potentially weak interactions between HLA-E peptide complexes and TCRs. Thus, HLA-E-Mtb-specific T cells have a highly diverse TCR repertoire.
Collapse
Affiliation(s)
- Linda Voogd
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne M.H.F. Drittij
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Calinda K.E. Dingenouts
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Kees L.M.C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | - Paula Ruibal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith A. Leitner
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Mark M. Davis
- Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
22
|
Wallace Z, Heunis T, Paterson RL, Suckling RJ, Grant T, Dembek M, Donoso J, Brener J, Long J, Bunjobpol W, Gibbs-Howe D, Kay DP, Leneghan DB, Godinho LF, Walker A, Singh PK, Knox A, Leonard S, Dorrell L. Instability of the HLA-E peptidome of HIV presents a major barrier to therapeutic targeting. Mol Ther 2024; 32:678-688. [PMID: 38219014 PMCID: PMC10928138 DOI: 10.1016/j.ymthe.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/14/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024] Open
Abstract
Naturally occurring T cells that recognize microbial peptides via HLA-E, a nonpolymorphic HLA class Ib molecule, could provide the foundation for new universal immunotherapeutics. However, confidence in the biological relevance of putative ligands is crucial, given that the mechanisms by which pathogen-derived peptides can access the HLA-E presentation pathway are poorly understood. We systematically interrogated the HIV proteome using immunopeptidomic and bioinformatic approaches, coupled with biochemical and cellular assays. No HIV HLA-E peptides were identified by tandem mass spectrometry analysis of HIV-infected cells. In addition, all bioinformatically predicted HIV peptide ligands (>80) were characterized by poor complex stability. Furthermore, infected cell elimination assays using an affinity-enhanced T cell receptor bispecific targeted to a previously reported HIV Gag HLA-E epitope demonstrated inconsistent presentation of the peptide, despite normal HLA-E expression on HIV-infected cells. This work highlights the instability of the HIV HLA-E peptidome as a major challenge for drug development.
Collapse
Affiliation(s)
- Zoë Wallace
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK.
| | - Tiaan Heunis
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| | | | | | | | | | - Jose Donoso
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| | | | - Joshua Long
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| | | | | | - Daniel P Kay
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| | | | | | | | | | - Andrew Knox
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| | | | - Lucy Dorrell
- Immunocore Ltd., Abingdon, Oxfordshire OX14 4RY, UK
| |
Collapse
|
23
|
Maciel M, Amara RR, Bar KJ, Crotty S, Deeks SG, Duplessis C, Gaiha G, McElrath MJ, McMichael A, Palin A, Rutishauser R, Shapiro S, Smiley ST, D'Souza MP. Exploring synergies between B- and T-cell vaccine approaches to optimize immune responses against HIV-workshop report. NPJ Vaccines 2024; 9:39. [PMID: 38383616 PMCID: PMC10881492 DOI: 10.1038/s41541-024-00818-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Affiliation(s)
- Milton Maciel
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rama R Amara
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Katharine J Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
- Division of HIV, Infectious Diseases, and Global Medicine, San Francisco, CA, USA
| | - Steven G Deeks
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Christopher Duplessis
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Gaurav Gaiha
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew McMichael
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Amy Palin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Rachel Rutishauser
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Stuart Shapiro
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Stephen T Smiley
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - M Patricia D'Souza
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
24
|
Kim HJ, Nakagawa H, Choi JY, Che X, Divris A, Liu Q, Wight AE, Zhang H, Saad A, Solhjou Z, Deban C, Azzi JR, Cantor H. A narrow T cell receptor repertoire instructs thymic differentiation of MHC class Ib-restricted CD8+ regulatory T cells. J Clin Invest 2024; 134:e170512. [PMID: 37934601 PMCID: PMC10760956 DOI: 10.1172/jci170512] [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: 03/14/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
Although most CD8+ T cells are equipped to kill infected or transformed cells, a subset may regulate immune responses and preserve self-tolerance. Here, we describe a CD8 lineage that is instructed to differentiate into CD8 T regulatory cells (Tregs) by a surprisingly restricted set of T cell receptors (TCRs) that recognize MHC-E (mouse Qa-1) and several dominant self-peptides. Recognition and elimination of pathogenic target cells that express these Qa-1-self-peptide complexes selectively inhibits pathogenic antibody responses without generalized immune suppression. Immunization with synthetic agonist peptides that mobilize CD8 Tregs in vivo efficiently inhibit antigraft antibody responses and markedly prolong heart and kidney organ graft survival. Definition of TCR-dependent differentiation and target recognition by this lineage of CD8 Tregs may open the way to new therapeutic approaches to inhibit pathogenic antibody responses.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology and
| | - Hidetoshi Nakagawa
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology and
| | - John Y. Choi
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Xuchun Che
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Andrew Divris
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Qingshi Liu
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Andrew E. Wight
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology and
| | - Hengcheng Zhang
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Anis Saad
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Zhabiz Solhjou
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Christa Deban
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Jamil R. Azzi
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Transplant Research Center, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Harvey Cantor
- Department of Cancer Immunology & Virology, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Immunology and
| |
Collapse
|
25
|
Middelburg J, Ghaffari S, Schoufour TAW, Sluijter M, Schaap G, Göynük B, Sala BM, Al-Tamimi L, Scheeren F, Franken KLMC, Akkermans JJLL, Cabukusta B, Joosten SA, Derksen I, Neefjes J, van der Burg SH, Achour A, Wijdeven RHM, Weidanz J, van Hall T. The MHC-E peptide ligands for checkpoint CD94/NKG2A are governed by inflammatory signals, whereas LILRB1/2 receptors are peptide indifferent. Cell Rep 2023; 42:113516. [PMID: 38048225 DOI: 10.1016/j.celrep.2023.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/23/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023] Open
Abstract
The immune checkpoint NKG2A/CD94 is a promising target for cancer immunotherapy, and its ligand major histocompatibility complex E (MHC-E) is frequently upregulated in cancer. NKG2A/CD94-mediated inhibition of lymphocytes depends on the presence of specific leader peptides in MHC-E, but when and where they are presented in situ is unknown. We apply a nanobody specific for the Qdm/Qa-1b complex, the NKG2A/CD94 ligand in mouse, and find that presentation of Qdm peptide depends on every member of the endoplasmic reticulum-resident peptide loading complex. With a turnover rate of 30 min, the Qdm peptide reflects antigen processing capacity in real time. Remarkably, Qdm/Qa-1b complexes require inflammatory signals for surface expression in situ, despite the broad presence of Qa-1b molecules in homeostasis. Furthermore, we identify LILRB1 as a functional inhibition receptor for MHC-E in steady state. These data provide a molecular understanding of NKG2A blockade in immunotherapy and assign MHC-E as a convergent ligand for multiple immune checkpoints.
Collapse
Affiliation(s)
- Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Soroush Ghaffari
- Department of Biology, College of Science, The University of Texas at Arlington, Arlington, TX, USA
| | - Tom A W Schoufour
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Gaby Schaap
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Büsra Göynük
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Benedetta M Sala
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Lejla Al-Tamimi
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Ferenc Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Jimmy J L L Akkermans
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Birol Cabukusta
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ian Derksen
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine, Karolinska Institute & Division of Infectious Diseases, Karolinska University Hospital, 171 65 Solna, Sweden
| | - Ruud H M Wijdeven
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Jon Weidanz
- Abexxa Biologics, Inc., Arlington, TX, USA; College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, USA
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
26
|
Guan J, Peske JD, Manoharan Valerio M, Park C, Robey EA, Sadegh-Nasseri S. Commensal bacteria maintain a Qa-1 b-restricted unconventional CD8 + T population in gut epithelium. eLife 2023; 12:RP90466. [PMID: 38127067 PMCID: PMC10735220 DOI: 10.7554/elife.90466] [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] [Indexed: 12/23/2023] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs) are characterized by an unusual phenotype and developmental pathway, yet their specific ligands and functions remain largely unknown. Here by analysis of QFL T cells, a population of CD8+ T cells critical for monitoring the MHC I antigen processing pathway, we established that unconventional Qa-1b-restricted CD8+ T cells are abundant in intestinal epithelium. We found that QFL T cells showed a Qa-1b-dependent unconventional phenotype in the spleen and small intestine of naïve wild-type mice. The splenic QFL T cells showed innate-like functionality exemplified by rapid response to cytokines or antigens, while the gut population was refractory to stimuli. Microbiota was required for the maintenance, but not the initial gut homing of QFL T cells. Moreover, monocolonization with Pediococcus pentosaceus, which expresses a peptide that cross-activated QFL T cells, was sufficient to maintain QFL T cells in the intestine. Thus, microbiota is critical for shaping the Qa-1b-restricted IEL landscape.
Collapse
Affiliation(s)
- Jian Guan
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - J David Peske
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Michael Manoharan Valerio
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Chansu Park
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ellen A Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | | |
Collapse
|
27
|
Affiliation(s)
- Jaroslav Holly
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
| |
Collapse
|
28
|
Manoharan Valerio M, Arana K, Guan J, Chan SW, Yang X, Kurd N, Lee A, Shastri N, Coscoy L, Robey EA. The promiscuous development of an unconventional Qa1b-restricted T cell population. Front Immunol 2023; 14:1250316. [PMID: 38022509 PMCID: PMC10644506 DOI: 10.3389/fimmu.2023.1250316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
MHC-E restricted CD8 T cells show promise in vaccine settings, but their development and specificity remain poorly understood. Here we focus on a CD8 T cell population reactive to a self-peptide (FL9) bound to mouse MHC-E (Qa-1b) that is presented in response to loss of the MHC I processing enzyme ERAAP, termed QFL T cells. We find that mature QFL thymocytes are predominantly CD8αβ+CD4-, show signs of agonist selection, and give rise to both CD8αα and CD8αβ intraepithelial lymphocytes (IEL), as well as memory phenotype CD8αβ T cells. QFL T cells require the MHC I subunit β-2 microglobulin (β2m), but do not require Qa1b or classical MHC I for positive selection. However, QFL thymocytes do require Qa1b for agonist selection and full functionality. Our data highlight the relaxed requirements for positive selection of an MHC-E restricted T cell population and suggest a CD8αβ+CD4- pathway for development of CD8αα IELs.
Collapse
Affiliation(s)
- Michael Manoharan Valerio
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Kathya Arana
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Jian Guan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shiao Wei Chan
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Xiaokun Yang
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Nadia Kurd
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Angus Lee
- Gene Targeting Facility Cancer Research Laboratory, University of California Berkeley, Berkeley, CA, United States
| | - Nilabh Shastri
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laurent Coscoy
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Ellen A. Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
29
|
Li Q, Lin L, Shou P, Liu K, Xue Y, Hu M, Ling W, Huang Y, Du L, Zheng C, Wang X, Zheng F, Zhang T, Wang Y, Shao C, Melino G, Shi Y, Wang Y. MHC class Ib-restricted CD8 + T cells possess strong tumoricidal activities. Proc Natl Acad Sci U S A 2023; 120:e2304689120. [PMID: 37856544 PMCID: PMC10614629 DOI: 10.1073/pnas.2304689120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/28/2023] [Indexed: 10/21/2023] Open
Abstract
The importance of classical CD8+ T cells in tumor eradication is well acknowledged. However, the anti-tumor activity of MHC (major histocompatibility complex) Ib-restricted CD8+ T (Ib-CD8+ T) cells remains obscure. Here, we show that CX3CR1-expressing Ib-CD8+ T cells (Ib-restricted CD8+ T cells) highly express cytotoxic factors, austerely resist exhaustion, and effectively eliminate various tumors. These Ib-CD8+ T cells can be primed by MHC Ia (MHC class Ia molecules) expressed on various cell types for optimal activation in a Tbet-dependent manner. Importantly, MHC Ia does not allogeneically activate Ib-CD8+ T cells, rather, sensitizes these cells for T cell receptor activation. Such effects were observed when MHC Ia+ cells were administered to tumor-bearing Kb-/-Db-/-mice. A similar population of tumoricidal CX3CR1+CD8+ T cells was identified in wild-type mice and melanoma patients. Adoptive transfer of Ib-CD8+ T cells to wild-type mice inhibited tumor progression without damaging normal tissues. Taken together, we demonstrate that MHC class Ia can prime Ib-CD8+ T cells for robust tumoricidal activities.
Collapse
Affiliation(s)
- Qing Li
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Liangyu Lin
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Peishun Shou
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Keli Liu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yueqing Xue
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Mingyuan Hu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Weifang Ling
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yin Huang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Liming Du
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Chunxing Zheng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Xuefeng Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Fanjun Zheng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Tao Zhang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yu Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu215123, China
| | - Gerry Melino
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome Tor Vergata, Rome00133, Italy
| | - Yufang Shi
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu215123, China
| | - Ying Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| |
Collapse
|
30
|
Zeng J, Jaijyan DK, Yang S, Pei S, Tang Q, Zhu H. Exploring the Potential of Cytomegalovirus-Based Vectors: A Review. Viruses 2023; 15:2043. [PMID: 37896820 PMCID: PMC10612100 DOI: 10.3390/v15102043] [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: 09/03/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/29/2023] Open
Abstract
Viral vectors have emerged as powerful tools for delivering and expressing foreign genes, playing a pivotal role in gene therapy. Among these vectors, cytomegalovirus (CMV) stands out as a promising viral vector due to its distinctive attributes including large packaging capacity, ability to achieve superinfection, broad host range, capacity to induce CD8+ T cell responses, lack of integration into the host genome, and other qualities that make it an appealing vector candidate. Engineered attenuated CMV strains such as Towne and AD169 that have a ~15 kb genomic DNA deletion caused by virus passage guarantee human safety. CMV's large genome enables the efficient incorporation of substantial foreign genes as demonstrated by CMV vector-based therapies for SIV, tuberculosis, cancer, malaria, aging, COVID-19, and more. CMV is capable of reinfecting hosts regardless of prior infection or immunity, making it highly suitable for multiple vector administrations. In addition to its broad cellular tropism and sustained high-level gene expression, CMV triggers robust, virus-specific CD8+ T cell responses, offering a significant advantage as a vaccine vector. To date, successful development and testing of murine CMV (MCMV) and rhesus CMV (RhCMV) vectors in animal models have demonstrated the efficacy of CMV-based vectors. These investigations have explored the potential of CMV vectors for vaccines against HIV, cancer, tuberculosis, malaria, and other infectious pathogens, as well as for other gene therapy applications. Moreover, the generation of single-cycle replication CMV vectors, produced by deleting essential genes, ensures robust safety in an immunocompromised population. The results of these studies emphasize CMV's effectiveness as a gene delivery vehicle and shed light on the future applications of a CMV vector. While challenges such as production complexities and storage limitations need to be addressed, ongoing efforts to bridge the gap between animal models and human translation continue to fuel the optimism surrounding CMV-based vectors. This review will outline the properties of CMV vectors and discuss their future applications as well as possible limitations.
Collapse
Affiliation(s)
- Janine Zeng
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA; (J.Z.); (D.K.J.); (S.P.)
| | - Dabbu Kumar Jaijyan
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA; (J.Z.); (D.K.J.); (S.P.)
| | - Shaomin Yang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518060, China;
| | - Shaokai Pei
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA; (J.Z.); (D.K.J.); (S.P.)
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 070101, USA; (J.Z.); (D.K.J.); (S.P.)
| |
Collapse
|
31
|
Huisman BD, Guan N, Rückert T, Garner L, Singh NK, McMichael AJ, Gillespie GM, Romagnani C, Birnbaum ME. High-throughput characterization of HLA-E-presented CD94/NKG2x ligands reveals peptides which modulate NK cell activation. Nat Commun 2023; 14:4809. [PMID: 37558657 PMCID: PMC10412585 DOI: 10.1038/s41467-023-40220-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 07/13/2023] [Indexed: 08/11/2023] Open
Abstract
HLA-E is a non-classical class I MHC protein involved in innate and adaptive immune recognition. While recent studies have shown HLA-E can present diverse peptides to NK cells and T cells, the HLA-E repertoire recognized by CD94/NKG2x has remained poorly defined, with only a limited number of peptide ligands identified. Here we screen a yeast-displayed peptide library in the context of HLA-E to identify 500 high-confidence unique peptides that bind both HLA-E and CD94/NKG2A or CD94/NKG2C. Utilizing the sequences identified via yeast display selections, we train prediction algorithms and identify human and cytomegalovirus (CMV) proteome-derived, HLA-E-presented peptides capable of binding and signaling through both CD94/NKG2A and CD94/NKG2C. In addition, we identify peptides which selectively activate NKG2C+ NK cells. Taken together, characterization of the HLA-E-binding peptide repertoire and identification of NK activity-modulating peptides present opportunities for studies of NK cell regulation in health and disease, in addition to vaccine and therapeutic design.
Collapse
Affiliation(s)
- Brooke D Huisman
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Ning Guan
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
| | - Timo Rückert
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany
| | - Lee Garner
- Centre for Immuno-Oncology, Old Road Campus Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nishant K Singh
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Andrew J McMichael
- Centre for Immuno-Oncology, Old Road Campus Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Geraldine M Gillespie
- Centre for Immuno-Oncology, Old Road Campus Research Building, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Chiara Romagnani
- Innate Immunity, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Leibniz Institut, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
- Department of Biological Engineering, MIT, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
32
|
He W, Gea-Mallorquí E, Colin-York H, Fritzsche M, Gillespie GM, Brackenridge S, Borrow P, McMichael AJ. Intracellular trafficking of HLA-E and its regulation. J Exp Med 2023; 220:214089. [PMID: 37140910 PMCID: PMC10165540 DOI: 10.1084/jem.20221941] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/13/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
Interest in MHC-E-restricted CD8+ T cell responses has been aroused by the discovery of their efficacy in controlling simian immunodeficiency virus (SIV) infection in a vaccine model. The development of vaccines and immunotherapies utilizing human MHC-E (HLA-E)-restricted CD8+ T cell response requires an understanding of the pathway(s) of HLA-E transport and antigen presentation, which have not been clearly defined previously. We show here that, unlike classical HLA class I, which rapidly exits the endoplasmic reticulum (ER) after synthesis, HLA-E is largely retained because of a limited supply of high-affinity peptides, with further fine-tuning by its cytoplasmic tail. Once at the cell surface, HLA-E is unstable and is rapidly internalized. The cytoplasmic tail plays a crucial role in facilitating HLA-E internalization, which results in its enrichment in late and recycling endosomes. Our data reveal distinctive transport patterns and delicate regulatory mechanisms of HLA-E, which help to explain its unusual immunological functions.
Collapse
Affiliation(s)
- Wanlin He
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Ester Gea-Mallorquí
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Huw Colin-York
- Kennedy Institute of Rheumatology, University of Oxford , Oxford, UK
| | - Marco Fritzsche
- Kennedy Institute of Rheumatology, University of Oxford , Oxford, UK
| | - Geraldine M Gillespie
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Simon Brackenridge
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Persephone Borrow
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Andrew J McMichael
- Nuffield Department of Medicine, Center for Immuno-Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Lin Z, Bashirova AA, Viard M, Garner L, Quastel M, Beiersdorfer M, Kasprzak WK, Akdag M, Yuki Y, Ojeda P, Das S, Andresson T, Naranbhai V, Horowitz A, McMichael AJ, Hoelzemer A, Gillespie GM, Garcia-Beltran WF, Carrington M. HLA class I signal peptide polymorphism determines the level of CD94/NKG2-HLA-E-mediated regulation of effector cell responses. Nat Immunol 2023; 24:1087-1097. [PMID: 37264229 PMCID: PMC10690437 DOI: 10.1038/s41590-023-01523-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 04/27/2023] [Indexed: 06/03/2023]
Abstract
Human leukocyte antigen (HLA)-E binds epitopes derived from HLA-A, HLA-B, HLA-C and HLA-G signal peptides (SPs) and serves as a ligand for CD94/NKG2A and CD94/NKG2C receptors expressed on natural killer and T cell subsets. We show that among 16 common classical HLA class I SP variants, only 6 can be efficiently processed to generate epitopes that enable CD94/NKG2 engagement, which we term 'functional SPs'. The single functional HLA-B SP, known as HLA-B/-21M, induced high HLA-E expression, but conferred the lowest receptor recognition. Consequently, HLA-B/-21M SP competes with other SPs for providing epitope to HLA-E and reduces overall recognition of target cells by CD94/NKG2A, calling for reassessment of previous disease models involving HLA-B/-21M. Genetic population data indicate a positive correlation between frequencies of functional SPs in humans and corresponding cytomegalovirus mimics, suggesting a means for viral escape from host responses. The systematic, quantitative approach described herein will facilitate development of prediction algorithms for accurately measuring the impact of CD94/NKG2-HLA-E interactions in disease resistance/susceptibility.
Collapse
Affiliation(s)
- Zhansong Lin
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Arman A Bashirova
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mathias Viard
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Lee Garner
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Max Quastel
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Maya Beiersdorfer
- Leibniz Institute of Virology, Hamburg, Germany
- 1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Wojciech K Kasprzak
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Marjan Akdag
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yuko Yuki
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Pedro Ojeda
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Sudipto Das
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Thorkell Andresson
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Vivek Naranbhai
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Center for the AIDS Programme of Research in South Africa, Durban, South Africa
| | - Amir Horowitz
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Angelique Hoelzemer
- Leibniz Institute of Virology, Hamburg, Germany
- 1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | | | | | - Mary Carrington
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
34
|
Yang H, Sun H, Brackenridge S, Zhuang X, Wing PAC, Quastel M, Walters L, Garner L, Wang B, Yao X, Felce SL, Peng Y, Moore S, Peeters BWA, Rei M, Canto Gomes J, Tomas A, Davidson A, Semple MG, Turtle LCW, Openshaw PJM, Baillie JK, Mentzer AJ, Klenerman P, Borrow P, Dong T, McKeating JA, Gillespie GM, McMichael AJ. HLA-E-restricted SARS-CoV-2-specific T cells from convalescent COVID-19 patients suppress virus replication despite HLA class Ia down-regulation. Sci Immunol 2023; 8:eabl8881. [PMID: 37390223 DOI: 10.1126/sciimmunol.abl8881] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 06/07/2023] [Indexed: 07/02/2023]
Abstract
Pathogen-specific CD8+ T cell responses restricted by the nonpolymorphic nonclassical class Ib molecule human leukocyte antigen E (HLA-E) are rarely reported in viral infections. The natural HLA-E ligand is a signal peptide derived from classical class Ia HLA molecules that interact with the NKG2/CD94 receptors to regulate natural killer cell functions, but pathogen-derived peptides can also be presented by HLA-E. Here, we describe five peptides from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that elicited HLA-E-restricted CD8+ T cell responses in convalescent patients with coronavirus disease 2019. These T cell responses were identified in the blood at frequencies similar to those reported for classical HLA-Ia-restricted anti-SARS-CoV-2 CD8+ T cells. HLA-E peptide-specific CD8+ T cell clones, which expressed diverse T cell receptors, suppressed SARS-CoV-2 replication in Calu-3 human lung epithelial cells. SARS-CoV-2 infection markedly down-regulated classical HLA class I expression in Calu-3 cells and primary reconstituted human airway epithelial cells, whereas HLA-E expression was not affected, enabling T cell recognition. Thus, HLA-E-restricted T cells could contribute to the control of SARS-CoV-2 infection alongside classical T cells.
Collapse
Affiliation(s)
- Hongbing Yang
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
| | - Hong Sun
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Key Laboratory of AIDS Immunology, Department of Laboratory Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Simon Brackenridge
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Xiaodong Zhuang
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Peter A C Wing
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Max Quastel
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Lucy Walters
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Lee Garner
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Beibei Wang
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Xuan Yao
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Suet Ling Felce
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
| | - Yanchun Peng
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Shona Moore
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Bas W A Peeters
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Margarida Rei
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus, Oxford, UK
| | - Joao Canto Gomes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, Braga, Portugal
| | - Ana Tomas
- Unidada de Investigacao em Patobiologia Molecular, Instituto Portugues de Oncologia de Lisboa Francisco Gentil, EPE Lisbon, Portugal
- Chronic Diseases Research Centre, NOVA Medical School, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Andrew Davidson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Malcolm G Semple
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Respiratory Unit, Alder Hey Children's Hospital, Eaton Road, Liverpool L12 2AP, UK
| | - Lance C W Turtle
- Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Tropical and Infectious Disease Unit, Liverpool University Hospitals NHS Foundation Trust (member of Liverpool Health Partners), Liverpool, UK
| | | | | | - Alexander J Mentzer
- Welcome Centre for Human Genetics, University of Oxford, Old Road Campus, Oxford, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Persephone Borrow
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Tao Dong
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jane A McKeating
- Chinese Academy of Medical Sciences Oxford Institute, Old Road Campus, Oxford, UK
- Nuffield Depertment of Clinical Medicine, NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Geraldine M Gillespie
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| | - Andrew J McMichael
- Centre for Immuno-Oncology, Nuffield Department of Clinical Medicine, University of Oxford, Old Road Campus, Oxford, UK
| |
Collapse
|
35
|
Arenas VR, Rugeles MT, Perdomo-Celis F, Taborda N. Recent advances in CD8 + T cell-based immune therapies for HIV cure. Heliyon 2023; 9:e17481. [PMID: 37441388 PMCID: PMC10333625 DOI: 10.1016/j.heliyon.2023.e17481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Achieving a cure for HIV infection is a global priority. There is substantial evidence supporting a central role for CD8+ T cells in the natural control of HIV, suggesting the rationale that these cells may be exploited to achieve remission or cure of this infection. In this work, we review the major challenges for achieving an HIV cure, the models of HIV remission, and the mechanisms of HIV control mediated by CD8+ T cells. In addition, we discuss strategies based on this cell population that could be used in the search for an HIV cure. Finally, we analyze the current challenges and perspectives to translate this basic knowledge toward scalable HIV cure strategies.
Collapse
Affiliation(s)
| | - María T. Rugeles
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | | | - Natalia Taborda
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
- Grupo de Investigaciones Biomédicas Uniremington, Programa de Medicina, Facultad de Ciencias de la Salud, Corporación Universitaria Remington, Medellin, Colombia
| |
Collapse
|
36
|
Geiger KM, Manoharan M, Coombs R, Arana K, Park CS, Lee AY, Shastri N, Robey EA, Coscoy L. Murine cytomegalovirus downregulates ERAAP and induces an unconventional T cell response to self. Cell Rep 2023; 42:112317. [PMID: 36995940 PMCID: PMC10539480 DOI: 10.1016/j.celrep.2023.112317] [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: 03/16/2022] [Revised: 01/02/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The endoplasmic reticulum aminopeptidase associated with antigen processing (ERAAP) plays a crucial role in shaping the peptide-major histocompatibility complex (MHC) class I repertoire and maintaining immune surveillance. While murine cytomegalovirus (MCMV) has multiple strategies for manipulating the antigen processing pathway to evade immune responses, the host has also developed ways to counter viral immune evasion. In this study, we find that MCMV modulates ERAAP and induces an interferon γ (IFN-γ)-producing CD8+ T cell effector response that targets uninfected ERAAP-deficient cells. We observe that ERAAP downregulation during infection leads to the presentation of the self-peptide FL9 on non-classical Qa-1b, thereby eliciting Qa-1b-restricted QFL T cells to proliferate in the liver and spleen of infected mice. QFL T cells upregulate effector markers upon MCMV infection and are sufficient to reduce viral load after transfer to immunodeficient mice. Our study highlights the consequences of ERAAP dysfunction during viral infection and provides potential targets for anti-viral therapies.
Collapse
Affiliation(s)
- Kristina M Geiger
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA; Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Manoharan
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rachel Coombs
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kathya Arana
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chan-Su Park
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Angus Y Lee
- Cancer Research Lab, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nilabh Shastri
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ellen A Robey
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA; Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Laurent Coscoy
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA; Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
37
|
Picker LJ, Lifson JD, Gale M, Hansen SG, Früh K. Programming cytomegalovirus as an HIV vaccine. Trends Immunol 2023; 44:287-304. [PMID: 36894436 PMCID: PMC10089689 DOI: 10.1016/j.it.2023.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 03/09/2023]
Abstract
The initial development of cytomegalovirus (CMV) as a vaccine vector for HIV/simian immunodeficiency virus (SIV) was predicated on its potential to pre-position high-frequency, effector-differentiated, CD8+ T cells in tissues for immediate immune interception of nascent primary infection. This goal was achieved and also led to the unexpected discoveries that non-human primate (NHP) CMVs can be programmed to differentially elicit CD8+ T cell responses that recognize viral peptides via classical MHC-Ia, and/or MHC-II, and/or MHC-E, and that MHC-E-restricted CD8+ T cell responses can uniquely mediate stringent arrest and subsequent clearance of highly pathogenic SIV, an unprecedented type of vaccine-mediated protection. These discoveries delineate CMV vector-elicited MHC-E-restricted CD8+ T cells as a functionally distinct T cell response with the potential for superior efficacy against HIV-1, and possibly other infectious agents or cancers.
Collapse
Affiliation(s)
- Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
38
|
Hansen SG, Womack JL, Perez W, Schmidt KA, Marshall E, Iyer RF, Cleveland Rubeor H, Otero CE, Taher H, Vande Burgt NH, Barfield R, Randall KT, Morrow D, Hughes CM, Selseth AN, Gilbride RM, Ford JC, Caposio P, Tarantal AF, Chan C, Malouli D, Barry PA, Permar SR, Picker LJ, Früh K. Late gene expression-deficient cytomegalovirus vectors elicit conventional T cells that do not protect against SIV. JCI Insight 2023; 8:e164692. [PMID: 36749635 PMCID: PMC10070102 DOI: 10.1172/jci.insight.164692] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Rhesus cytomegalovirus-based (RhCMV-based) vaccine vectors induce immune responses that protect ~60% of rhesus macaques (RMs) from SIVmac239 challenge. This efficacy depends on induction of effector memory-based (EM-biased) CD8+ T cells recognizing SIV peptides presented by major histocompatibility complex-E (MHC-E) instead of MHC-Ia. The phenotype, durability, and efficacy of RhCMV/SIV-elicited cellular immune responses were maintained when vector spread was severely reduced by deleting the antihost intrinsic immunity factor phosphoprotein 71 (pp71). Here, we examined the impact of an even more stringent attenuation strategy on vector-induced immune protection against SIV. Fusion of the FK506-binding protein (FKBP) degradation domain to Rh108, the orthologue of the essential human CMV (HCMV) late gene transcription factor UL79, generated RhCMV/SIV vectors that conditionally replicate only when the FK506 analog Shield-1 is present. Despite lacking in vivo dissemination and reduced innate and B cell responses to vaccination, Rh108-deficient 68-1 RhCMV/SIV vectors elicited high-frequency, durable, EM-biased, SIV-specific T cell responses in RhCMV-seropositive RMs at doses of ≥ 1 × 106 PFU. Strikingly, elicited CD8+ T cells exclusively targeted MHC-Ia-restricted epitopes and failed to protect against SIVmac239 challenge. Thus, Rh108-dependent late gene expression is required for both induction of MHC-E-restricted T cells and protection against SIV.
Collapse
Affiliation(s)
- Scott G. Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Jennie L. Womack
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Wilma Perez
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | | | - Emily Marshall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Ravi F. Iyer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Hillary Cleveland Rubeor
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Claire E. Otero
- Duke Human Vaccine Institute, Duke University Medical School, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Nathan H. Vande Burgt
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
- Center for Human Systems Immunology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Kurt T. Randall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Colette M. Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Andrea N. Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Roxanne M. Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Julia C. Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Alice F. Tarantal
- California National Primate Research Center, UCD, Davis, California, USA
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, UCD, Davis, California, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
- Center for Human Systems Immunology, School of Medicine, Duke University, Durham, North Carolina, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Peter A. Barry
- California National Primate Research Center, UCD, Davis, California, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical School, Durham, North Carolina, USA
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
39
|
Ruibal P, Derksen I, van Wolfswinkel M, Voogd L, Franken KLMC, El Hebieshy AF, van Hall T, Schoufour TAW, Wijdeven RH, Ottenhoff THM, Scheeren FA, Joosten SA. Thermal-exchange HLA-E multimers reveal specificity in HLA-E and NKG2A/CD94 complex interactions. Immunology 2023; 168:526-537. [PMID: 36217755 DOI: 10.1111/imm.13591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022] Open
Abstract
There is growing interest in HLA-E-restricted T-cell responses as a possible novel, highly conserved, vaccination targets in the context of infectious and malignant diseases. The developing field of HLA multimers for the detection and study of peptide-specific T cells has allowed the in-depth study of TCR repertoires and molecular requirements for efficient antigen presentation and T-cell activation. In this study, we developed a method for efficient peptide thermal exchange on HLA-E monomers and multimers allowing the high-throughput production of HLA-E multimers. We optimized the thermal-mediated peptide exchange, and flow cytometry staining conditions for the detection of TCR and NKG2A/CD94 receptors, showing that this novel approach can be used for high-throughput identification and analysis of HLA-E-binding peptides which could be involved in T-cell and NK cell-mediated immune responses. Importantly, our analysis of NKG2A/CD94 interaction in the presence of modified peptides led to new molecular insights governing the interaction of HLA-E with this receptor. In particular, our results reveal that interactions of HLA-E with NKG2A/CD94 and the TCR involve different residues. Altogether, we present a novel HLA-E multimer technology based on thermal-mediated peptide exchange allowing us to investigate the molecular requirements for HLA-E/peptide interaction with its receptors.
Collapse
Affiliation(s)
- Paula Ruibal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Ian Derksen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Linda Voogd
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Angela F El Hebieshy
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom A W Schoufour
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruud H Wijdeven
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
Wang L, Song Y, Wang H, Zhang X, Wang M, He J, Li S, Zhang L, Li K, Cao L. Advances of Artificial Intelligence in Anti-Cancer Drug Design: A Review of the Past Decade. Pharmaceuticals (Basel) 2023; 16:253. [PMID: 37259400 PMCID: PMC9963982 DOI: 10.3390/ph16020253] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 10/13/2023] Open
Abstract
Anti-cancer drug design has been acknowledged as a complicated, expensive, time-consuming, and challenging task. How to reduce the research costs and speed up the development process of anti-cancer drug designs has become a challenging and urgent question for the pharmaceutical industry. Computer-aided drug design methods have played a major role in the development of cancer treatments for over three decades. Recently, artificial intelligence has emerged as a powerful and promising technology for faster, cheaper, and more effective anti-cancer drug designs. This study is a narrative review that reviews a wide range of applications of artificial intelligence-based methods in anti-cancer drug design. We further clarify the fundamental principles of these methods, along with their advantages and disadvantages. Furthermore, we collate a large number of databases, including the omics database, the epigenomics database, the chemical compound database, and drug databases. Other researchers can consider them and adapt them to their own requirements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kang Li
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin 150081, China
| | - Lei Cao
- Department of Biostatistics, School of Public Health, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
41
|
Yee JL, Strelow LI, White JA, Rosenthal AN, Barry PA. Horizontal transmission of endemic viruses among rhesus macaques (Macaca mulatta): Implications for human cytomegalovirus vaccine/challenge design. J Med Primatol 2023; 52:53-63. [PMID: 36151734 PMCID: PMC9825633 DOI: 10.1111/jmp.12621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Rhesus macaques are natural hosts to multiple viruses including rhesus cytomegalovirus (RhCMV), rhesus rhadinovirus (RRV), and Simian Foamy Virus (SFV). While viral infections are ubiquitous, viral transmissions to uninfected animals are incompletely defined. Management procedures of macaque colonies include cohorts that are Specific Pathogen Free (SPF). Greater understanding of viral transmission would augment SPF protocols. Moreover, vaccine/challenge studies of human viruses would be enhanced by leveraging transmission of macaque viruses to recapitulate expected challenges of human vaccine trials. MATERIALS AND METHODS This study characterizes viral transmissions to uninfected animals following inadvertent introduction of RhCMV/RRV/SFV-infected adults to a cohort of uninfected juveniles. Following co-housing with virus-positive adults, juveniles were serially evaluated for viral infection. RESULTS Horizontal viral transmission was rapid and absolute, reaching 100% penetrance between 19 and 78 weeks. CONCLUSIONS This study provides insights into viral natural histories with implications for colony management and modeling vaccine-mediated immune protection studies.
Collapse
Affiliation(s)
- JoAnn L Yee
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Lisa I Strelow
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| | - Jessica A White
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Ann N Rosenthal
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Peter A Barry
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| |
Collapse
|
42
|
Rich RR, Cron RQ. The Human Immune Response. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
43
|
Brochu H, Wang R, Tollison T, Pyo CW, Thomas A, Tseng E, Law L, Picker LJ, Gale M, Geraghty DE, Peng X. Alternative splicing and genetic variation of mhc-e: implications for rhesus cytomegalovirus-based vaccines. Commun Biol 2022; 5:1387. [PMID: 36536032 PMCID: PMC9762870 DOI: 10.1038/s42003-022-04344-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Rhesus cytomegalovirus (RhCMV)-based vaccination against Simian Immunodeficiency virus (SIV) elicits MHC-E-restricted CD8+ T cells that stringently control SIV infection in ~55% of vaccinated rhesus macaques (RM). However, it is unclear how accurately the RM model reflects HLA-E immunobiology in humans. Using long-read sequencing, we identified 16 Mamu-E isoforms and all Mamu-E splicing junctions were detected among HLA-E isoforms in humans. We also obtained the complete Mamu-E genomic sequences covering the full coding regions of 59 RM from a RhCMV/SIV vaccine study. The Mamu-E gene was duplicated in 32 (54%) of 59 RM. Among four groups of Mamu-E alleles: three ~5% divergent full-length allele groups (G1, G2, G2_LTR) and a fourth monomorphic group (G3) with a deletion encompassing the canonical Mamu-E exon 6, the presence of G2_LTR alleles was significantly (p = 0.02) associated with the lack of RhCMV/SIV vaccine protection. These genomic resources will facilitate additional MHC-E targeted translational research.
Collapse
Affiliation(s)
- Hayden Brochu
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, NC, 27695, USA
| | - Ruihan Wang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Tammy Tollison
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA
| | - Chul-Woo Pyo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Alexander Thomas
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | | | - Lynn Law
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Diseases, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Daniel E Geraghty
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Xinxia Peng
- Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, NC, 27607, USA.
- Bioinformatics Graduate Program, North Carolina State University, Raleigh, NC, 27695, USA.
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
44
|
Fisher JG, Doyle ADP, Graham LV, Khakoo SI, Blunt MD. Disruption of the NKG2A:HLA-E Immune Checkpoint Axis to Enhance NK Cell Activation against Cancer. Vaccines (Basel) 2022; 10:1993. [PMID: 36560403 PMCID: PMC9783329 DOI: 10.3390/vaccines10121993] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Ligation of the inhibitory receptor NKG2A by its ligand HLA-E negatively regulates the activation of natural killer (NK) cells, as well as subsets of CD8+ T cells and innate T cell populations. NKG2A has recently become a novel immune checkpoint target for the treatment of cancer and direct antibody mediated blockade of NKG2A function is currently under assessment in two phase 3 clinical trials. In addition to direct targeting, the NKG2A:HLA-E axis can also be disrupted indirectly via multiple different targeted cancer agents that were not previously recognised to possess immunomodulatory properties. Increased understanding of immune cell modulation by targeted cancer therapies will allow for the design of rational and more efficacious drug combination strategies to improve cancer patient outcomes. In this review, we summarise and discuss the various strategies currently in development which either directly or indirectly disrupt the NKG2A:HLA-E interaction to enhance NK cell activation against cancer.
Collapse
Affiliation(s)
| | | | | | | | - Matthew D. Blunt
- School of Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
45
|
Romero-Martín L, Duran-Castells C, Olivella M, Rosás-Umbert M, Ruiz-Riol M, Sanchez J, Hartigan-O Connor D, Mothe B, Olvera À, Brander C. Disruption of the HLA-E/NKG2X axis is associated with uncontrolled HIV infections. Front Immunol 2022; 13:1027855. [PMID: 36466823 PMCID: PMC9716355 DOI: 10.3389/fimmu.2022.1027855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/18/2022] [Indexed: 09/28/2023] Open
Abstract
The contribution of the HLA-E/NKG2X axis in NK-mediated control of HIV infection remains unclear. We have studied the relationship between HLA-E expression and phenotypical as well as functional characteristics of NK cells, in the context of chronic HIV infection and in an in vitro model of acute infection. High viremia in HIV+ individuals was related to increased HLA-E expression, and changes in NK subpopulations, especially a reduction of the CD56bright as well as an increase in adaptive NK subpopulation. Uncontrolled HIV infection was also characterized by a reversion of the NKG2A/NKG2C expression ratio and a loss of positive and negative regulation of NK mediated by HLA-E. This was reflected in a lower cytotoxic, degranulation and cytokine production capacity, especially in CD56bright and adaptive NK. In line with these results, HLA-E expression showed a positive correlation with viral growth inhibition in an in vitro model of acute infection at day 7, which was lost after 14 days of culture. Using HLA-E expressing K562 cells, we determined that only one out of 11 described HIV-derived HLA-E epitopes increased HLA-E surface stability. In spite of that, eight of the 11 epitopes were capable of increasing degranulation and three drove differences in NK-cell mediated cell lysis or cytokine secretion. In conclusion, our results indicate that HLA-E molecules presenting HIV-derived epitopes may sensitize target cells for NK lysis in early HIV infection. However, prolonged exposure to elevated HLA-E expression levels in vivo may lead to NK cell dysfunction and reduced viral control In chronic infection.
Collapse
Affiliation(s)
- Luis Romero-Martín
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Clara Duran-Castells
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- Universitat Autonoma de Barcelona (UAB), Barcelona, Spain
| | - Mireia Olivella
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Míriam Rosás-Umbert
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Marta Ruiz-Riol
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Dennis Hartigan-O Connor
- California National Primate Research Center and Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- Fundació Lluita contra la Sida, Infectious Disease Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Àlex Olvera
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Institute for Health Science Research Germans Trias i Pujol (IGTP), Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
- CIBERINFEC, Centro de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
- ICREA, Barcelona, Spain
| |
Collapse
|
46
|
Alrubayyi A, Rowland-Jones S, Peppa D. Natural killer cells during acute HIV-1 infection: clues for HIV-1 prevention and therapy. AIDS 2022; 36:1903-1915. [PMID: 35851334 PMCID: PMC9612724 DOI: 10.1097/qad.0000000000003319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022]
Abstract
Despite progress in preexposure prophylaxis, the number of newly diagnosed cases with HIV-1 remains high, highlighting the urgent need for preventive and therapeutic strategies to reduce HIV-1 acquisition and limit disease progression. Early immunological events, occurring during acute infection, are key determinants of the outcome and course of disease. Understanding early immune responses occurring before viral set-point is established, is critical to identify potential targets for prophylactic and therapeutic approaches. Natural killer (NK) cells represent a key cellular component of innate immunity and contribute to the early host defence against HIV-1 infection, modulating the pathogenesis of acute HIV-1 infection (AHI). Emerging studies have identified tools for harnessing NK cell responses and expanding specialized NK subpopulations with adaptive/memory features, paving the way for development of novel HIV-1 therapeutics. This review highlights the knowns and unknowns regarding the role of NK cell subsets in the containment of acute HIV-1 infection, and summarizes recent advances in selectively augmenting NK cell functions through prophylactic and therapeutic interventions.
Collapse
Affiliation(s)
- Aljawharah Alrubayyi
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford
- Division of Infection and Immunity, University College London
| | | | - Dimitra Peppa
- Division of Infection and Immunity, University College London
- Mortimer Market Centre, Department of HIV, CNWL NHS Trust, London, UK
| |
Collapse
|
47
|
Ruibal P, Franken KLMC, van Meijgaarden KE, van Wolfswinkel M, Derksen I, Scheeren FA, Janssen GMC, van Veelen PA, Sarfas C, White AD, Sharpe SA, Palmieri F, Petrone L, Goletti D, Abeel T, Ottenhoff THM, Joosten SA. Identification of HLA-E Binding Mycobacterium tuberculosis-Derived Epitopes through Improved Prediction Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1555-1565. [PMID: 36096642 PMCID: PMC9536328 DOI: 10.4049/jimmunol.2200122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/03/2022] [Indexed: 01/04/2023]
Abstract
Tuberculosis (TB) remains one of the deadliest infectious diseases worldwide, posing great social and economic burden to affected countries. Novel vaccine approaches are needed to increase protective immunity against the causative agent Mycobacterium tuberculosis (Mtb) and to reduce the development of active TB disease in latently infected individuals. Donor-unrestricted T cell responses represent such novel potential vaccine targets. HLA-E-restricted T cell responses have been shown to play an important role in protection against TB and other infections, and recent studies have demonstrated that these cells can be primed in vitro. However, the identification of novel pathogen-derived HLA-E binding peptides presented by infected target cells has been limited by the lack of accurate prediction algorithms for HLA-E binding. In this study, we developed an improved HLA-E binding peptide prediction algorithm and implemented it to identify (to our knowledge) novel Mtb-derived peptides with capacity to induce CD8+ T cell activation and that were recognized by specific HLA-E-restricted T cells in Mycobacterium-exposed humans. Altogether, we present a novel algorithm for the identification of pathogen- or self-derived HLA-E-presented peptides.
Collapse
Affiliation(s)
- Paula Ruibal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Ian Derksen
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Charlotte Sarfas
- Research and Development Department, UK Health Security Agency, Salisbury, United Kingdom
| | - Andrew D White
- Research and Development Department, UK Health Security Agency, Salisbury, United Kingdom
| | - Sally A Sharpe
- Research and Development Department, UK Health Security Agency, Salisbury, United Kingdom
| | - Fabrizio Palmieri
- National Institute for Infectious Diseases Lazzaro Spallanzani Scientific Institute for Research, Hospitalization and Healthcare, Rome, Italy
| | - Linda Petrone
- National Institute for Infectious Diseases Lazzaro Spallanzani Scientific Institute for Research, Hospitalization and Healthcare, Rome, Italy
| | - Delia Goletti
- National Institute for Infectious Diseases Lazzaro Spallanzani Scientific Institute for Research, Hospitalization and Healthcare, Rome, Italy
| | - Thomas Abeel
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands; and
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands;
| |
Collapse
|
48
|
Malouli D, Gilbride RM, Wu HL, Hwang JM, Maier N, Hughes CM, Newhouse D, Morrow D, Ventura AB, Law L, Tisoncik-Go J, Whitmore L, Smith E, Golez I, Chang J, Reed JS, Waytashek C, Weber W, Taher H, Uebelhoer LS, Womack JL, McArdle MR, Gao J, Papen CR, Lifson JD, Burwitz BJ, Axthelm MK, Smedley J, Früh K, Gale M, Picker LJ, Hansen SG, Sacha JB. Cytomegalovirus-vaccine-induced unconventional T cell priming and control of SIV replication is conserved between primate species. Cell Host Microbe 2022; 30:1207-1218.e7. [PMID: 35981532 PMCID: PMC9927879 DOI: 10.1016/j.chom.2022.07.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/01/2022] [Accepted: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Strain 68-1 rhesus cytomegalovirus expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) primes MHC-E-restricted CD8+ T cells that control SIV replication in 50%-60% of the vaccinated rhesus macaques. Whether this unconventional SIV-specific immunity and protection is unique to rhesus macaques or RhCMV or is intrinsic to CMV remains unknown. Here, using cynomolgus CMV vectors expressing SIV antigens (CyCMV/SIV) and Mauritian cynomolgus macaques, we demonstrate that the induction of MHC-E-restricted CD8+ T cells requires matching CMV to its host species. RhCMV does not elicit MHC-E-restricted CD8+ T cells in cynomolgus macaques. However, cynomolgus macaques vaccinated with species-matched 68-1-like CyCMV/SIV mounted MHC-E-restricted CD8+ T cells, and half of the vaccinees stringently controlled SIV post-challenge. Protected animals manifested a vaccine-induced IL-15 transcriptomic signature that is associated with efficacy in rhesus macaques. These findings demonstrate that the ability of species-matched CMV vectors to elicit MHC-E-restricted CD8+ T cells that are required for anti-SIV efficacy is conserved in nonhuman primates, and these data support the development of HCMV/HIV for a prophylactic HIV vaccine.
Collapse
Affiliation(s)
- Daniel Malouli
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Helen L Wu
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Joseph M Hwang
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Nicholas Maier
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - David Morrow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Lynn Law
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Leanne Whitmore
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Elise Smith
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Inah Golez
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jean Chang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jason S Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Courtney Waytashek
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Whitney Weber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Husam Taher
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Luke S Uebelhoer
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jennie L Womack
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Matthew R McArdle
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Junwei Gao
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Courtney R Papen
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Benjamin J Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jeremy Smedley
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Klaus Früh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Louis J Picker
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Jonah B Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
49
|
Wallace Z, Singh PK, Dorrell L. Combination strategies to durably suppress HIV-1: Soluble T cell receptors. J Virus Erad 2022; 8:100082. [PMID: 36065296 PMCID: PMC9440443 DOI: 10.1016/j.jve.2022.100082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/06/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Immunotherapeutic interventions to enhance natural HIV-specific CD8+ T cell responses, such as vaccination or adoptive T cell transfer, have been a major focus of HIV cure efforts. However, these approaches have not been effective in overcoming viral immune evasion mechanisms. Soluble T cell receptor (TCR) bispecifics are a new class of 'off-the-shelf' therapeutic designed to address these limitations. These biologics are built on the Immune mobilising monoclonal TCRs against X disease (ImmTAX) platform, which was pioneered in oncology and recently validated by the FDA's approval of tebentafusp for treatment of metastatic uveal melanoma. ImmTAV® are an application of this technology undergoing clinical development for the elimination of chronic viral infections. ImmTAV molecules comprise an affinity-enhanced virus-specific TCR fused to an anti-CD3 effector domain. Engineering of the TCR confers extraordinary specificity and affinity for cognate viral antigen and the anti-CD3 enables retargeting of non-exhausted cytolytic T cells, irrespective of their specificity. These features enable ImmTAV molecules to detect and kill infected cells, even when expressing very low levels of antigen, bypassing ineffective host immune responses. Furthermore, the modularity of the platform allows for engineering of TCRs that effectively target viral variants. In this review, we discuss the progress made in the development of ImmTAV molecules as therapeutics for functional cure of chronic hepatitis B and HIV, from concept to the clinic.
Collapse
|
50
|
Medetgul-Ernar K, Davis MM. Standing on the shoulders of mice. Immunity 2022; 55:1343-1353. [PMID: 35947979 PMCID: PMC10035762 DOI: 10.1016/j.immuni.2022.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/13/2023]
Abstract
While inbred mice have informed most of what we know about the immune system in the modern era, they have clear limitations with respect to their ability to be informative regarding genetic heterogeneity or microbial influences. They have also not been very predictive as models of human disease or vaccination results. Although there are concerted attempts to compensate for these flaws, the rapid rise of human studies, driven by both technical and conceptual advances, promises to fill in these gaps, as well as provide direct information about human diseases and vaccination responses. Work on human immunity has already provided important additional perspectives on basic immunology such as the importance of clonal deletion to self-tolerance, and while many challenges remain, it seems inevitable that "the human model" will continue to inform us about the immune system and even allow for the discovery of new mechanisms.
Collapse
Affiliation(s)
- Kwat Medetgul-Ernar
- Immunology Program, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Mark M Davis
- Howard Hughes Medical Institute, Institute for Immunity, Transplantation and Infection, Department of Microbiology and Immunology, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|