1
|
Li Y, Li Z, Tang Y, Zhuang X, Feng W, Boor PPC, Buschow S, Sprengers D, Zhou G. Unlocking the therapeutic potential of the NKG2A-HLA-E immune checkpoint pathway in T cells and NK cells for cancer immunotherapy. J Immunother Cancer 2024; 12:e009934. [PMID: 39486805 PMCID: PMC11529472 DOI: 10.1136/jitc-2024-009934] [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: 07/02/2024] [Accepted: 09/29/2024] [Indexed: 11/04/2024] Open
Abstract
Immune checkpoint blockade, which enhances the reactivity of T cells to eliminate cancer cells, has emerged as a potent strategy in cancer therapy. Besides T cells, natural killer (NK) cells also play an indispensable role in tumor surveillance and destruction. NK Group 2 family of receptor A (NKG2A), an emerging co-inhibitory immune checkpoint expressed on both NK cells and T cells, mediates inhibitory signal via interaction with its ligand human leukocyte antigen-E (HLA-E), thereby attenuating the effector and cytotoxic functions of NK cells and T cells. Developing antibodies to block NKG2A, holds promise in restoring the antitumor cytotoxicity of NK cells and T cells. In this review, we delve into the expression and functional significance of NKG2A and HLA-E, elucidating how the NKG2A-HLA-E axis contributes to tumor immune escape via signal transduction mechanisms. Furthermore, we provide an overview of clinical trials investigating NKG2A blockade, either as monotherapy or in combination with other therapeutic antibodies, highlighting the responses of the immune system and the clinical benefits for patients. We pay special attention to additional immune co-signaling molecules that serve as potential targets on both NK cells and T cells, aiming to evoke more robust immune responses against cancer. This review offers an in-depth exploration of the NKG2A-HLA-E pathway as a pivotal checkpoint in the anti-tumor responses, paving the way for new immunotherapeutic strategies to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Yan Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Zhu Li
- Department of Dermatology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yisen Tang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xiaomei Zhuang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wanhua Feng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sonja Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Guoying Zhou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Cheng W, Zhu N, Wang J, Yang R. A role of gut microbiota metabolites in HLA-E and NKG2 blockage immunotherapy against tumors: new insights for clinical application. Front Immunol 2024; 15:1331518. [PMID: 39229258 PMCID: PMC11368731 DOI: 10.3389/fimmu.2024.1331518] [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: 11/01/2023] [Accepted: 07/16/2024] [Indexed: 09/05/2024] Open
Abstract
One of major breakthroughs in immunotherapy against tumor is from blocking immune checkpoint molecules on tumor and reactive T cells. The development of CTLA-4 and PD-1 blockage antibodies has triggered to search for additional effective therapeutic strategies. This causes recent findings that blocking the interaction of checkpoint molecule NKG2A in NK and CD8 T cells with HLA-E in tumors is effective in defensing tumors. Interestingly, gut microbiota also affects this immune checkpoint immunotherapy against tumor. Gut microbiota such as bacteria can contribute to the regulation of host immune response and homeostasis. They not only promote the differentiation and function of immunosuppressive cells but also the inflammatory cells through the metabolites such as tryptophan (Trp) and bile acid (BA) metabolites as well as short chain fatty acids (SCFAs). These gut microbiota metabolites (GMMs) educated immune cells can affect the differentiation and function of effective CD8 and NK cells. Notably, these metabolites also directly affect the activity of CD8 and NK cells. Furthermore, the expression of CD94/NKG2A in the immune cells and/or their ligand HLA-E in the tumor cells is also regulated by gut microbiota associated immune factors. These findings offer new insights for the clinical application of gut microbiota in precise and/or personalized treatments of tumors. In this review, we will discuss the impacts of GMMs and GMM educated immune cells on the activity of effective CD8 and NK cells and the expression of CD94/NKG2A in immune cells and/or their ligand HLA-E in tumor cells.
Collapse
Affiliation(s)
- Wenyue Cheng
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Ningning Zhu
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Juanjuan Wang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rongcun Yang
- Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Nankai University, Tianjin, China
- Department of Immunology, Nankai University School of Medicine, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| |
Collapse
|
3
|
Patin EC, Nenclares P, Chan Wah Hak C, Dillon MT, Patrikeev A, McLaughlin M, Grove L, Foo S, Soliman H, Barata JP, Marsden J, Baldock H, Gkantalis J, Roulstone V, Kyula J, Burley A, Hubbard L, Pedersen M, Smith SA, Clancy-Thompson E, Melcher AA, Ono M, Rullan A, Harrington KJ. Sculpting the tumour microenvironment by combining radiotherapy and ATR inhibition for curative-intent adjuvant immunotherapy. Nat Commun 2024; 15:6923. [PMID: 39134540 PMCID: PMC11319479 DOI: 10.1038/s41467-024-51236-6] [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: 01/05/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
The combination of radiotherapy/chemoradiotherapy and immune checkpoint blockade can result in poor outcomes in patients with locally advanced head and neck squamous cell carcinoma (HNSCC). Here, we show that combining ATR inhibition (ATRi) with radiotherapy (RT) increases the frequency of activated NKG2A+PD-1+ T cells in animal models of HNSCC. Compared with the ATRi/RT treatment regimen alone, the addition of simultaneous NKG2A and PD-L1 blockade to ATRi/RT, in the adjuvant, post-radiotherapy setting induces a robust antitumour response driven by higher infiltration and activation of cytotoxic T cells in the tumour microenvironment. The efficacy of this combination relies on CD40/CD40L costimulation and infiltration of activated, proliferating memory CD8+ and CD4+ T cells with persistent or new T cell receptor (TCR) signalling, respectively. We also observe increased richness in the TCR repertoire and emergence of numerous and large TCR clonotypes that cluster based on antigen specificity in response to NKG2A/PD-L1/ATRi/RT. Collectively, our data point towards potential combination approaches for the treatment of HNSCC.
Collapse
Affiliation(s)
- Emmanuel C Patin
- Targeted Therapy Team, The Institute of Cancer Research, London, UK.
| | - Pablo Nenclares
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Charleen Chan Wah Hak
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Magnus T Dillon
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Anton Patrikeev
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Lorna Grove
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Shane Foo
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | | | - Holly Baldock
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Jim Gkantalis
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | - Joan Kyula
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Amy Burley
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Lisa Hubbard
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
| | | | | | - Alan A Melcher
- Translational Immunotherapy Team, The Institute of Cancer Research, London, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Antonio Rullan
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| | - Kevin J Harrington
- Targeted Therapy Team, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital, London, UK
| |
Collapse
|
4
|
Barlesi F, Cho BC, Goldberg SB, Yoh K, Zimmer Gelatti AC, Mann H, Gopinathan A, Bielecka ZF, Newton M, Aggarwal C. PACIFIC-9: Phase III trial of durvalumab + oleclumab or monalizumab in unresectable stage III non-small-cell lung cancer. Future Oncol 2024; 20:2137-2147. [PMID: 39023287 PMCID: PMC11508940 DOI: 10.1080/14796694.2024.2354160] [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: 02/09/2024] [Accepted: 05/08/2024] [Indexed: 07/20/2024] Open
Abstract
Evidence from the Phase III PACIFIC trial established durvalumab, a monoclonal antibody (mAb) targeting PD-L1, following concurrent chemoradiotherapy (cCRT) as a global standard of care for patients with unresectable, stage III non-small-cell lung cancer (NSCLC). There remains an unmet need to improve upon the outcomes achieved with the PACIFIC regimen. Combining durvalumab with other immunotherapies may improve outcomes further. Two such immunotherapies include oleclumab, an mAb targeting CD73, and monalizumab, an mAb targeting NKG2A. Both agents demonstrated antitumor activity in early-phase trials. PACIFIC-9 (NCT05221840) is an international, double-blind, randomized, placebo-controlled, Phase III trial comparing durvalumab plus either oleclumab or monalizumab with durvalumab plus placebo in patients with unresectable, stage III NSCLC and no disease progression following cCRT.Clinical Trial Registration: NCT05221840 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Fabrice Barlesi
- Gustave Roussy, Medical Oncology Department, Villejuif, France
- Paris Saclay University, Faculty of Medicine, Kremlin-Bicêtre, France
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Sarah B Goldberg
- Yale School of Medicine & Yale Cancer Center, New Haven, CT, USA
| | - Kiyotaka Yoh
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Ana Caroline Zimmer Gelatti
- Grupo Oncoclínicas, Porto Alegre, Brazil
- Grupo Brasileiro de Oncologia Torácica, Porto Alegre, Brazil
- Hospital São Lucas PUC/RS, Brazil
| | | | | | | | | | - Charu Aggarwal
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
5
|
Fert I, Douguet L, Vesin B, Moncoq F, Noirat A, Authié P, Ciret S, Le Chevalier F, Blanc C, Vitrenko Y, Charneau P, Majlessi L, Anna F. T-cell immunity induced and reshaped by an anti-HPV immuno-oncotherapeutic lentiviral vector. NPJ Vaccines 2024; 9:102. [PMID: 38858404 PMCID: PMC11164992 DOI: 10.1038/s41541-024-00894-0] [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: 11/30/2023] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
We recently developed an immuno-oncotherapy against human papillomavirus (HPV)-induced tumors based on a lentiviral vector encoding the Early E6 and E7 oncoproteins of HPV16 and HPV18 genotypes, namely "Lenti-HPV-07". The robust and long-lasting anti-tumor efficacy of Lenti-HPV-07 is dependent on CD8+ T-cell induction and remodeling of the tumor microenvironment. Here, we first established that anti-vector immunity induced by Lenti-HPV-07 prime has no impact on the efficacy of a homologous boost to amplify anti-HPV T-cell immunity. To longitudinally monitor the evolution of the T-cell repertoire generated after the prime, homologous or heterologous boost with Lenti-HPV-07, we tracked T-cell clonotypes by deep sequencing of T-Cell Receptor (TCR) variable β and α chain mRNA, applied to whole peripheral blood cells (PBL) and a T cell population specific of an immunodominant E7HPV16 epitope. We observed a hyper-expansion of clonotypes post prime, accompanied by increased frequencies of HPV-07-specific T cells. Additionally, there was a notable diversification of clonotypes post boost in whole PBL, but not in the E7HPV16-specific T cells. We then demonstrated that the effector functions of such Lenti-HPV-07-induced T cells synergize with anti-checkpoint inhibitory treatments by systemic administration of anti-TIM3 or anti-NKG2A monoclonal antibodies. While Lenti-HPV-07 is about to enter a Phase I/IIa clinical trial, these results will help better elucidate its mode of action in immunotherapy against established HPV-mediated malignancies.
Collapse
Affiliation(s)
- Ingrid Fert
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laëtitia Douguet
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Benjamin Vesin
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fanny Moncoq
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Amandine Noirat
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Sylvain Ciret
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Catherine Blanc
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Yakov Vitrenko
- Institut Pasteur, Université Paris Cité, Biomics Technology Platform, F-75015, Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| | - François Anna
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 Rue du Dr. Roux, F-75015, Paris, France.
| |
Collapse
|
6
|
Liang J, Liao L, Xie L, Tang W, Yu X, Lu Y, Chen H, Xu J, Sun L, Wu H, Cui C, Tan Y. PITPNC1 Suppress CD8 + T cell immune function and promote radioresistance in rectal cancer by modulating FASN/CD155. J Transl Med 2024; 22:117. [PMID: 38291470 PMCID: PMC10826121 DOI: 10.1186/s12967-024-04931-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Radioresistance is a primary factor contributing to the failure of rectal cancer treatment. Immune suppression plays a significant role in the development of radioresistance. We have investigated the potential role of phosphatidylinositol transfer protein cytoplasmic 1 (PITPNC1) in regulating immune suppression associated with radioresistance. METHODS To elucidate the mechanisms by which PITPNC1 influences radioresistance, we established HT29, SW480, and MC38 radioresistant cell lines. The relationship between radioresistance and changes in the proportion of immune cells was verified through subcutaneous tumor models and flow cytometry. Changes in the expression levels of PITPNC1, FASN, and CD155 were determined using immunohistochemistry and western blotting techniques. The interplay between these proteins was investigated using immunofluorescence co-localization and immunoprecipitation assays. Additionally, siRNA and lentivirus-mediated gene knockdown or overexpression, as well as co-culture of tumor cells with PBMCs or CD8+ T cells and establishment of stable transgenic cell lines in vivo, were employed to validate the impact of the PITPNC1/FASN/CD155 pathway on CD8+ T cell immune function. RESULTS Under irradiation, the apoptosis rate and expression of apoptosis-related proteins in radioresistant colorectal cancer cell lines were significantly decreased, while the cell proliferation rate increased. In radioresistant tumor-bearing mice, the proportion of CD8+ T cells and IFN-γ production within immune cells decreased. Immunohistochemical analysis of human and animal tissue specimens resistant to radiotherapy showed a significant increase in the expression levels of PITPNC1, FASN, and CD155. Gene knockdown and rescue experiments demonstrated that PITPNC1 can regulate the expression of CD155 on the surface of tumor cells through FASN. In addition, co-culture experiments and in vivo tumor-bearing experiments have shown that silencing PITPNC1 can inhibit FASN/CD155, enhance CD8+ T cell immune function, promote colorectal cancer cell death, and ultimately reduce radioresistance in tumor-bearing models. CONCLUSIONS PITPNC1 regulates the expression of CD155 through FASN, inhibits CD8+ T cell immune function, and promotes radioresistance in rectal cancer.
Collapse
Affiliation(s)
- Junxian Liang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Limin Liao
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lang Xie
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - WenWen Tang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yinghao Lu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongzhen Chen
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Juanli Xu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Sun
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huanmei Wu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yujing Tan
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
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
|
8
|
Melero I, Ochoa MC, Molina C, Sanchez‐Gregorio S, Garasa S, Luri‐Rey C, Hervas‐Stubbs S, Casares N, Elizalde E, Gomis G, Cirella A, Berraondo P, Teijeira A, Alvarez M. Intratumoral co-injection of NK cells and NKG2A-neutralizing monoclonal antibodies. EMBO Mol Med 2023; 15:e17804. [PMID: 37782273 PMCID: PMC10630884 DOI: 10.15252/emmm.202317804] [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: 04/04/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023] Open
Abstract
NK-cell reactivity against cancer is conceivably suppressed in the tumor microenvironment by the interaction of the inhibitory receptor NKG2A with the non-classical MHC-I molecules HLA-E in humans or Qa-1b in mice. We found that intratumoral delivery of NK cells attains significant therapeutic effects only if co-injected with anti-NKG2A and anti-Qa-1b blocking monoclonal antibodies against solid mouse tumor models. Such therapeutic activity was contingent on endogenous CD8 T cells and type-1 conventional dendritic cells (cDC1). Moreover, the anti-tumor effects were enhanced upon combination with systemic anti-PD-1 mAb treatment and achieved partial abscopal efficacy against distant non-injected tumors. In xenografted mice bearing HLA-E-expressing human cancer cells, intratumoral co-injection of activated allogeneic human NK cells and clinical-grade anti-NKG2A mAb (monalizumab) synergistically achieved therapeutic effects. In conclusion, these studies provide evidence for the clinical potential of intratumoral NK cell-based immunotherapies that exert their anti-tumor efficacy as a result of eliciting endogenous T-cell responses.
Collapse
Affiliation(s)
- Ignacio Melero
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
- Departments of Immunology and OncologyClínica Universidad de NavarraPamplonaSpain
| | - Maria C Ochoa
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Carmen Molina
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Sandra Sanchez‐Gregorio
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Saray Garasa
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Carlos Luri‐Rey
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Sandra Hervas‐Stubbs
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Noelia Casares
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Edurne Elizalde
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Gabriel Gomis
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Assunta Cirella
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
| | - Pedro Berraondo
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Alvaro Teijeira
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Maite Alvarez
- Program for Immunology and Immunotherapy, CIMAUniversidad de NavarraPamplonaSpain
- Navarra Institute for Health Research (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
- Cell Therapy, Stem Cells and Tissue GroupBiocruces Bizkaia Health Research InstituteBarakaldoSpain
- Research Unit, Basque Center for Blood Transfusion and Human TissuesOsakidetzaGaldakaoSpain
| |
Collapse
|
9
|
Stieg DC, Parris JLD, Yang THL, Mirji G, Reiser SK, Murali N, Werts M, Barnoud T, Lu DY, Shinde R, Murphy ME, Claiborne DT. The African-centric P47S Variant of TP53 Confers Immune Dysregulation and Impaired Response to Immune Checkpoint Inhibition. CANCER RESEARCH COMMUNICATIONS 2023; 3:1200-1211. [PMID: 37441266 PMCID: PMC10335007 DOI: 10.1158/2767-9764.crc-23-0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
The tumor suppressor TP53 is the most frequently mutated gene in cancer and is mutationally inactivated in 50% of sporadic tumors. Inactivating mutations in TP53 also occur in Li Fraumeni syndrome (LFS). In addition to germline mutations in TP53 in LFS that completely inactivate this protein, there are many more germline mutant forms of TP53 in human populations that partially inactivate this protein: we call these partially inactivating mutations "hypomorphs." One of these hypomorphs is a SNP that exists in 6%-10% of Africans and 1%-2% of African Americans, which changes proline at amino acid 47 to serine (Pro47Ser; P47S). We previously showed that the P47S variant of p53 is intrinsically impaired for tumor suppressor function, and that this SNP is associated with increased cancer risk in mice and humans. Here we show that this SNP also influences the tumor microenvironment, and the immune microenvironment profile in P47S mice is more protumorigenic. At basal levels, P47S mice show impaired memory T-cell formation and function, along with increased anti-inflammatory (so-called "M2") macrophages. We show that in tumor-bearing P47S mice, there is an increase in immunosuppressive myeloid-derived suppressor cells and decreased numbers of activated dendritic cells, macrophages, and B cells, along with evidence for increased T-cell exhaustion in the tumor microenvironment. Finally, we show that P47S mice demonstrate an incomplete response to anti-PD-L1 therapy. Our combined data suggest that the African-centric P47S variant leads to both intrinsic and extrinsic defects in tumor suppression. Significance Findings presented here show that the P47S variant of TP53 influences the immune microenvironment, and the immune response to cancer. This is the first time that a naturally occurring genetic variant of TP53 has been shown to negatively impact the immune microenvironment and the response to immunotherapy.
Collapse
Affiliation(s)
- David C. Stieg
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Joshua L. D. Parris
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Tyler Hong Loong Yang
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Gauri Mirji
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Sarah Kim Reiser
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Nivitha Murali
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Madison Werts
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Thibaut Barnoud
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - David Y. Lu
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Rahul Shinde
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Maureen E. Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania
| | - Daniel T. Claiborne
- Program in Immunology, Microenvironment, and Metastasis, The Wistar Institute, Philadelphia, Pennsylvania
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Benavente S. Remodeling the tumor microenvironment to overcome treatment resistance in HPV-negative head and neck cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:291-313. [PMID: 37457128 PMCID: PMC10344731 DOI: 10.20517/cdr.2022.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/02/2023] [Accepted: 05/22/2023] [Indexed: 07/18/2023]
Abstract
Despite intensive efforts and refined techniques, overall survival in HPV-negative head and neck cancer remains poor. Robust immune priming is required to elicit a strong and durable antitumor immune response in immunologically cold and excluded tumors like HPV-negative head and neck cancer. This review highlights how the tumor microenvironment could be affected by different immune and stromal cell types, weighs the need to integrate metabolic regulation of the tumor microenvironment into cancer treatment strategies and summarizes the emerging clinical applicability of personalized immunotherapeutic strategies in HPV-negative head and neck cancer.
Collapse
Affiliation(s)
- Sergi Benavente
- Correspondence to: Dr. Sergi Benavente, Department of Radiation Oncology, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119, Barcelona 08035, Spain. E-mail:
| |
Collapse
|
11
|
Baude J, Limagne E, Ladjohounlou R, Mirjolet C. Combining radiotherapy and NK cell-based therapies: The time has come. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:31-60. [PMID: 37438020 DOI: 10.1016/bs.ircmb.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Natural killer (NK) cells are innate lymphoid cells that play an essential role in the anti-tumor response through immunosurveillance, multiple mechanisms of cytotoxicity and the synthesis of cytokines modulating the immune tumor microenvironment (TME). After the dramatic advances in immunotherapy targeting T cells including the success of checkpoint inhibitors or autologous chimeric antigen receptor (CAR) expressing T cells in clinical practice, NK cells have gained growing interest for the development of new therapies. Although NK cells have shown promising responses in leukemia patients, the effects of NK-targeted therapies are currently limited in the treatment of solid tumors. Thus, radiotherapy could provide a valuable solution to improve treatments targeting NK cells. Indeed, ionizing radiations represent a powerful immuno-modulator that can either induce a pro-inflammatory and anti-tumor TME, or conversely lead to immunosuppression of effector immune cells in favor of tumor growth and therapeutic escape, depending on how it is delivered and tumor models. However, the effects of ionizing radiation on NK cells are only partially understood. Therefore, we review the effects of radiotherapy on the NK cell-mediated anti-tumor response, and propose potential strategies to reinvigorate NK cells by combining radiotherapy with NK cell-targeted therapies.
Collapse
Affiliation(s)
- Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France
| | - Emeric Limagne
- TIReCS Team, UMR INSERM 1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, Dijon, France; University of Bourgogne Franche-Comté, Dijon, France
| | - Riad Ladjohounlou
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France
| | - Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, Centre Georges-François Leclerc, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| |
Collapse
|
12
|
Zhang XZ, Tao SP, Liang SX, Chen SB, Liu FS, Jiang W, Chen MJ. Nomogram based on circulating lymphocyte subsets for predicting radiation pneumonia in esophageal squamous cell carcinoma. Front Immunol 2022; 13:938795. [PMID: 36105795 PMCID: PMC9465326 DOI: 10.3389/fimmu.2022.938795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Currently, the relationship between radiation pneumonia (RP) and circulating immune cell in patients with esophageal squamous cell carcinoma (ESCC) remains unclear. This study aimed to explore the relationship between RP and circulating lymphocyte subsets in patients with ESCC receiving chemoradiotherapy (CRT), and develop a nomogram model to predict RP. Since we should implement clinical intervention to ≥ grade 2 RP, a nomogram model for ≥ grade 2 RP was also established to provide an early warning. Patients and methods This study retrospectively included 121 patients with ESCC receiving CRT from Guangxi Medical University Cancer Hospital from 2013 to 2021. Independent factors associated with occurrence of RP and ≥ grade 2 RP were identified by univariate and multivariate logistic regression analysis in the training cohort, and incorporated into nomograms. The predictive accuracy and discrimination of the model was assessed using Concordance Index (C-index), calibration curve and decision curve analysis (DCA). And each model was internally validated. Additionally, to verify the optimized predictive performance of the nomograms, the area under the ROC curve (AUC) of each nomogram was compared to that of single independent risk factors, lung V10 and lung V20, respectively. Moreover, each model was further evaluated for risk stratification to identify populations at high risk of RP and ≥ grade 2 RP. Results Multivariate analysis suggested that TNM stage, post-RT percentage of CD8+ T cell, and lung V15 were independent predictive factors of RP. Besides, pre- and post-RT percentage of CD8+ T cell, and V15 were independent factors of ≥ grade 2 RP. The C-indexes of RP and ≥ grade 2 RP nomograms were 0.809 (95% CI: 0.715-0.903) and 0.787 (95% CI: 0.685-0.889) in the training cohort, respectively. And the C-indexes of RP and ≥ grade 2 RP nomograms were 0.718 (95% CI: 0.544-0.892) and 0.621 (95% CI: 0.404-0.837) in the validation cohort, respectively. The calibration curves showed that the predicted values of model agreed well with actual observations. Moreover, DCA results indicated the applicability and accuracy of the models to predict RP and ≥ grade 2 RP. After stratification, the incidence of the high-risk group was significantly higher than that of the low-risk group with respect to either RP or ≥ grade 2 RP. Conclusion TNM stage, post-RT percentage of CD8+ T cell, and lung V15 were the independent predictors of RP toxicity. Pre- and post-RT percentage of CD8+ T cell, and lung V15 were the independent factors of ≥ grade 2 RP toxicity. The nomograms based on circulating lymphocyte subsets can robustly predict RP and ≥ grade 2 RP, guiding clinicians in risk stratification and early intervention.
Collapse
Affiliation(s)
- Xiao-zhen Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Su-ping Tao
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shi-xiong Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shu-bin Chen
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fu-shuang Liu
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wei Jiang
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Mao-jian Chen, ; Wei Jiang,
| | - Mao-jian Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Mao-jian Chen, ; Wei Jiang,
| |
Collapse
|