1
|
Goloudina A, Le Chevalier F, Authié P, Charneau P, Majlessi L. Shared neoantigens for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200978. [PMID: 40256120 PMCID: PMC12008704 DOI: 10.1016/j.omton.2025.200978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Exploration of neoantigens holds the potential to be productive in immuno-oncotherapy. Among tumor-specific antigens, neoantigens result from genetic instability that gives rise to non-synonymous somatic mutations, highly specific to tumor cells. In addition to point mutations, gene rearrangements, indels leading to frameshifts, chromosomal translocations or inversions that may lead to fusion proteins, alternative mRNA splicing, and integration of genetic material of oncogenic viruses into the host genome provide consistent sources of neoantigens that are absent in healthy tissues. Out of these alterations, 2%-3% may generate T cell neoepitopes, possibly detectable by TCRs. Neoantigens are absent in healthy tissues and are thus at low risk of triggering autoimmunity. In addition, the host lymphocytes have not been rendered tolerant toward them and it is possible to induce immune responses against them. Here, we overview the two categories of neoantigens, i.e., private and shared, and their use in immuno-oncotherapy in selected pre-clinical and clinical studies. The vast majority of commonly occurring tumor-specific mutations are cancer causing and are permanently expressed by all malignant tumor cells, preventing the latter from escaping vaccine-induced anti-neoantigen immunity. The use of public neoantigens combined with efficient vaccine platforms can provide non-personalized "off-the-shelf" therapeutic vaccine candidates for broad-spectrum immunotherapy purposes.
Collapse
Affiliation(s)
- Anastasia Goloudina
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Fabien Le Chevalier
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Authié
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Pierre Charneau
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| | - Laleh Majlessi
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, 28 rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
2
|
Cesano A, Augustin R, Barrea L, Bedognetti D, Bruno TC, Carturan A, Hammer C, Ho WS, Kather JN, Kirchhoff T, Lu RO, McQuade J, Najjar YG, Pietrobon V, Ruella M, Shen R, Soldati L, Spencer C, Betof Warner A, Warren S, Ziv E, Marincola FM. Advances in the understanding and therapeutic manipulation of cancer immune responsiveness: a Society for Immunotherapy of Cancer (SITC) review. J Immunother Cancer 2025; 13:e008876. [PMID: 39824527 PMCID: PMC11749597 DOI: 10.1136/jitc-2024-008876] [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: 01/19/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025] Open
Abstract
Cancer immunotherapy-including immune checkpoint inhibition (ICI) and adoptive cell therapy (ACT)-has become a standard, potentially curative treatment for a subset of advanced solid and liquid tumors. However, most patients with cancer do not benefit from the rapidly evolving improvements in the understanding of principal mechanisms determining cancer immune responsiveness (CIR); including patient-specific genetically determined and acquired factors, as well as intrinsic cancer cell biology. Though CIR is multifactorial, fundamental concepts are emerging that should be considered for the design of novel therapeutic strategies and related clinical studies. Recent advancements as well as novel approaches to address the limitations of current treatments are discussed here, with a specific focus on ICI and ACT.
Collapse
Affiliation(s)
| | - Ryan Augustin
- University of Pittsburgh Department of Medicine, Pittsburgh, Pennsylvania, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Tullia C Bruno
- University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Winson S Ho
- University of California San Francisco, San Francisco, California, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Tomas Kirchhoff
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Rongze O Lu
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Jennifer McQuade
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Marco Ruella
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rhine Shen
- Kite Pharma Inc, Santa Monica, California, USA
| | | | - Christine Spencer
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | | | - Elad Ziv
- University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
3
|
Liu Q, Wu P, Lei J, Bai P, Zhong P, Yang M, Wei P. Old concepts, new tricks: How peptide vaccines are reshaping cancer immunotherapy? Int J Biol Macromol 2024; 279:135541. [PMID: 39270889 DOI: 10.1016/j.ijbiomac.2024.135541] [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: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Over the past few decades, research on cancer immunotherapy has firmly established immune cells as key players in effective cancer treatment. Peptide vaccines directly targeting immune cells have demonstrated immense potential due to their specificity and applicability. However, developing peptide vaccines to generate tumor-reactive T cells remains challenging, primarily due to suboptimal immunogenicity and overcoming the immunosuppressive tumor microenvironment (TME). In this review, we discuss various elements of effective peptide vaccines, including antigen selection, peptide epitope optimization, vaccine adjuvants, and the combination of multiple immunotherapies, in addition to recent advances in tumor neoantigens as well as epitopes bound by non-classical human leukocyte antigen (HLA) molecules, to increase the understanding of cancer peptide vaccines and provide multiple references for the design of subsequent T cell-based peptide vaccines.
Collapse
Affiliation(s)
- Qingyang Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Peihua Wu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Jun Lei
- Hubei Key Laboratory of Cell Homeostasis, State Key Laboratory of Virology, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Laboratory Medicine, Xixi Hospital of Hangzhou, Hangzhou, China
| | - Peng Bai
- In Vivo Pharmacology Unit, WuXi AppTec, Nantong, Jiangsu, China
| | - Peiluan Zhong
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Min Yang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China.
| |
Collapse
|
4
|
Song LN, Wang B, Cai JL, Zhang PL, Chen SP, Zhou ZJ, Dai Z. Stratifying ICIs-responsive tumor microenvironment in HCC: from parsing out immune-hypoxic crosstalk to clinically applicable MRI-radiomics models. Br J Cancer 2024; 130:1356-1364. [PMID: 38355839 PMCID: PMC11014931 DOI: 10.1038/s41416-023-02463-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 10/04/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND We aimed to redefine Immune checkpoint inhibitors (ICIs)-responsive "hot" TME and develop a corresponding stratification model to maximize ICIs-efficacy in Hepatocellular Carcinoma (HCC). METHODS Hypoxic scores were designed, and the relevance to immunotherapy responses were validated in pan-cancers through single cell analysis. Multi-omics analysis using the hypoxic scores and immune infiltrate abundance was performed to redefine the ICIs-responsive TME subtype in HCC patients from TCGA (n = 363) and HCCDB database (n = 228). The immune hypoxic stress index (IHSI) was constructed to stratify the ICIs-responsive TME subtype, with exploring biological mechanism in vitro and in vivo. MRI-radiomics models were built for clinical applicability. RESULTS The hypoxic scores were lower in the dominant cell-subclusters of responders in pan-cancers. The higher immune infiltrate-normoxic (HIN) subtype was redefined as the ICIs-responsive TME. Stratification of the HIN subtype using IHSI effectively identified ICIs-responders in Melanoma (n = 122) and urological cancer (n = 22). TRAF3IP3, the constituent gene of IHSI, was implicated in ICIs-relevant "immune-hypoxic" crosstalk by stimulating MAVS/IFN-I pathway under normoxic condition. MRI-radiomics models assessing TRAF3IP3 with HIF1A expression (AUC > 0.80) screened ICIs-Responders in HCC cohort (n = 75). CONCLUSION The hypoxic-immune stratification redefined ICIs-responsive TME and provided MRI-Radiomics models for initial ICIs-responders screening, with IHSI facilitating further identification.
Collapse
Affiliation(s)
- Li-Na Song
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Biao Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jia-Liang Cai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Pei-Ling Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Shi-Ping Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zheng-Jun Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China
| | - Zhi Dai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
McShan AC, Flores-Solis D, Sun Y, Garfinkle SE, Toor JS, Young MC, Sgourakis NG. Conformational plasticity of RAS Q61 family of neoepitopes results in distinct features for targeted recognition. Nat Commun 2023; 14:8204. [PMID: 38081856 PMCID: PMC10713829 DOI: 10.1038/s41467-023-43654-9] [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: 08/28/2022] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
The conformational landscapes of peptide/human leucocyte antigen (pHLA) protein complexes encompassing tumor neoantigens provide a rationale for target selection towards autologous T cell, vaccine, and antibody-based therapeutic modalities. Here, using complementary biophysical and computational methods, we characterize recurrent RAS55-64 Q61 neoepitopes presented by the common HLA-A*01:01 allotype. We integrate sparse NMR restraints with Rosetta docking to determine the solution structure of NRASQ61K/HLA-A*01:01, which enables modeling of other common RAS55-64 neoepitopes. Hydrogen/deuterium exchange mass spectrometry experiments alongside molecular dynamics simulations reveal differences in solvent accessibility and conformational plasticity across a panel of common Q61 neoepitopes that are relevant for recognition by immunoreceptors. Finally, we predict binding and provide structural models of NRASQ61K antigens spanning the entire HLA allelic landscape, together with in vitro validation for HLA-A*01:191, HLA-B*15:01, and HLA-C*08:02. Our work provides a basis to delineate the solution surface features and immunogenicity of clinically relevant neoepitope/HLA targets for cancer therapy.
Collapse
Affiliation(s)
- Andrew C McShan
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- School of Chemistry & Biochemistry, Georgia Institute of Technology, 901 Atlantic Dr NW, Atlanta, GA, 30318, USA
| | - David Flores-Solis
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA, 95064, USA
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold Straße 3A, 37075, Göttingen, Germany
| | - Yi Sun
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samuel E Garfinkle
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jugmohit S Toor
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, CA, 95064, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, 48202, USA
| | - Michael C Young
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Nikolaos G Sgourakis
- Center for Computational and Genomic Medicine, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Anand S, Littler DR, Mobbs JI, Braun A, Baker DG, Tennant L, Purcell AW, Vivian JP, Rossjohn J. Complimentary electrostatics dominate T Cell Receptor binding to a psoriasis-associated-peptide-antigen presented by Human Leukocyte Antigen (HLA) C*06:02. J Biol Chem 2023:104930. [PMID: 37330172 PMCID: PMC10371836 DOI: 10.1016/j.jbc.2023.104930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/05/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
Psoriasis is a chronic skin disease characterised by hyperproliferative epidermal lesions infiltrated by autoreactive T cells. Individuals expressing the Human Leukocyte antigen (HLA) C*06:02 allele are at highest risk for developing psoriasis. An autoreactive T cell clone (termed Vα3S1/Vβ13S1) isolated from psoriatic plaques is selective for HLA-C*06:02-presenting a peptide derived from the melanocyte-specific auto-antigen ADAMTSL5 (VRSRRCLRL). Here we determine the crystal structure of this psoriatic TCR-HLA-C*06:02- ADAMTSL5 complex with a stabilised peptide. Docking of the TCR involves an extensive complementary charge network formed between negatively charged TCR residues interleaving with exposed arginine residues from the self-peptide and the HLA-C*06:02 α1 helix. We probed these interactions through mutagenesis and activation assays. The charged interface spans the polymorphic region of the C1/C2 HLA group. Notably the peptide binding groove of HLA C*06:02 appears exquisitely suited for presenting highly charged Arg-rich epitopes recognised by this acidic psoriatic TCR. Overall, we provide a structural basis for understanding engagement of melanocyte antigen-presenting cells by a TCR implicated in psoriasis, while simultaneously expanding our knowledge of how TCRs engage HLA-C.
Collapse
Affiliation(s)
- Sushma Anand
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Dene R Littler
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Jesse I Mobbs
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Asolina Braun
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Daniel G Baker
- Janssen Research & Development, LLC, Horsham, Philadelphia, Pennsylvania, USA
| | - Luke Tennant
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Anthony W Purcell
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Julian P Vivian
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton Victoria, Australia; Institute of Infection and Immunity, Cardiff University, School of Medicine, Cardiff, UK.
| |
Collapse
|
7
|
Lei J, Zhou Z, Fang J, Sun Z, He M, He B, Chen Q, Paek C, Chen P, Zhou J, Wang H, Tang M, Yin L, Chen Y. Aspirin induces immunogenic cell death and enhances cancer immunotherapy in colorectal cancer. Int Immunopharmacol 2023; 121:110350. [PMID: 37290325 DOI: 10.1016/j.intimp.2023.110350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/07/2023] [Accepted: 05/13/2023] [Indexed: 06/10/2023]
Abstract
The use of aspirin is associated with reduced incidence of colorectal cancer (CRC). However, the detailed mechanism remains unclear. In this study, we reported that colon cancer cells treated with aspirin showed the hallmarks of immunogenic cell death (ICD), including surface expression of calreticulin (CRT) and heat shock protein 70 (HSP70). Mechanistically, aspirin induced endoplasmic reticulum (ER) stress in colon cancer cells. In addition, aspirin decreased the expression of the glucose transporters, GLUT3, and reduced the key enzyme of glycolysis, including HK2, PFKM, PKM2 and LDHA. The changes of tumor glycolysis after aspirin treatment were associated with c-MYC downregulation. Moreover, aspirin potentiated the antitumor efficacy of anti-PD-1 antibody and anti-CTLA-4 antibody in CT26 tumors. However, this antitumor activity of aspirin in combination with anti-PD-1 antibody was abolished by the depletion of CD8+ T cells. Vaccination with tumor antigens is one of the strategies for activating T-cell response against tumors. Here, we demonstrated that aspirin-treated tumor cells in combination with tumor antigens (AH1 peptide) or protective substituted peptide (A5 peptide) could be served as a potent vaccine to eradicate tumors. Overall, our data indicated that aspirin can be used as an inducer of ICD for CRC therapy.
Collapse
Affiliation(s)
- Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Zihao Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jialing Fang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Zaiqiao Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Mengting He
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Boxiao He
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Qian Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Chonil Paek
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Peng Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Jin Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Hongjian Wang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Mingliang Tang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China.
| | - Yongshun Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province 430072, China.
| |
Collapse
|
8
|
Mattila J, Sormunen S, Heikkilä N, Mattila IP, Saramäki J, Arstila TP. Analysis of thymic generation of shared T-cell receptor α repertoire associated with recognition of tumor antigens shows no preference for neoantigens over wild-type antigens. Cancer Med 2023; 12:13486-13496. [PMID: 37114587 PMCID: PMC10315763 DOI: 10.1002/cam4.6002] [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: 11/28/2022] [Revised: 04/06/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The number of mutations in cancer cells is an important predictor of a positive response to cancer immunotherapy. It has been suggested that the neoantigens produced by these mutations are more immunogenic than nonmutated tumor antigens, which are likely to be protected by immunological tolerance. However, the mechanisms of tolerance as regards tumor antigens are incompletely understood. METHODS Here, we have analyzed the impact of thymic negative selection on shared T-cell receptor (TCR) repertoire associated with the recognition of either mutated or nonmutated tumor antigens by comparing previously known TCR-antigen-pairs to TCR repertoires of 21 immunologically healthy individuals. RESULTS Our results show that TCRα chains associated with either type of tumor antigens are readily generated in the thymus, at a frequency similar to TCRα chains associated with nonself. In the peripheral repertoire, the relative clone size of nonself-associated chains is higher than that of the tumor antigens, but importantly, there is no difference between TCRα chains associated with mutated or nonmutated tumor antigens. CONCLUSION This suggests that the tolerance mechanisms protecting nonmutated tumor antigens are non-deletional and therefore potentially reversible. As unmutated antigens are, unlike mutations, shared by a large number of patients, they may offer advantages in designing immunological approaches to cancer treatment.
Collapse
Affiliation(s)
- Joonatan Mattila
- Research Programs Unit, Translational Immunology, Haartmaninkatu 3 (PL 21) 00014, and MedicumUniversity of HelsinkiHelsinkiFinland
| | - Silja Sormunen
- Department of Computer ScienceAalto UniversityEspooFinland
| | - Nelli Heikkilä
- Research Programs Unit, Translational Immunology, Haartmaninkatu 3 (PL 21) 00014, and MedicumUniversity of HelsinkiHelsinkiFinland
- Faculty of Medicine, Center for Vaccinology, Department of Pathology and ImmunologyUniversity of GenevaGenevaSwitzerland
| | - Ilkka P. Mattila
- Department of Pediatric Cardiac and Transplantation SurgeryHospital for Children and Adolescents, Helsinki University Central HospitalHelsinkiFinland
| | - Jari Saramäki
- Department of Computer ScienceAalto UniversityEspooFinland
| | - T. Petteri Arstila
- Research Programs Unit, Translational Immunology, Haartmaninkatu 3 (PL 21) 00014, and MedicumUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
9
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 174:xv-xxv. [PMID: 37039770 DOI: 10.1016/s0091-679x(23)00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
10
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 180:xv-xxv. [PMID: 37890936 DOI: 10.1016/s0091-679x(23)00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
11
|
Tumor antigens and vaccines in colorectal cancer. MEDICINE IN DRUG DISCOVERY 2022. [DOI: 10.1016/j.medidd.2022.100144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
12
|
Bieri U, Enderlin D, Buser L, Wettstein MS, Eberli D, Moch H, Hermanns T, Poyet C. Modified immunoscore improves the prediction of progression-free survival in patients with non-muscle-invasive bladder cancer: A digital pathology study. Front Oncol 2022; 12:964672. [PMID: 36212478 PMCID: PMC9539272 DOI: 10.3389/fonc.2022.964672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour-infiltrating lymphocytes (TIL), known to be of prognostic value in various solid tumours, have been in the focus of research in the last years. TIL are often quantified via IMMUNOSCORE ® (IS), a scoring system based on TIL cell densities. Recent studies were able to replicate these findings for muscle-invasive bladder cancer (MIBC), however data regarding non-muscle-invasive bladder cancer (NMIBC) are scarce. This study aimed to evaluate the value of a modified Immunoscore (mIS) as a predictive marker for NMIBC prognosis using tissue-micro-arrays (TMAs). We analysed two TMAs containing 316 samples from 158 patients with NMIBC, stained for CD3, CD8, CD45RO and FOXP3. Stained TIL were captured by digital pathology, cumulated, averaged, and reported as density (stained cells per mm²). The mIS was then constructed based on density of all four immune-cell types. Clinical, pathological and follow-up data were collected retrospectively. Univariable and multivariable cox regression analysis was performed to assess the potential value of mIS as a predictor for progression free survival (PFS) and recurrence-free-survival (RFS). Patients within "European Organisation for Research and Treatment of Cancer" (EORTC) risk groups were further substratified in high mIS and low mIS subgroups. Finally log-rank test was used to compare the different survival curves. The median age in our cohort was 68 years (Interquartile Range (IQR): 60 - 76), and 117 (74%) patients were male. A total of 26 patients (16.5%) were classified as EORTC low risk, 45 (28.5%) as intermediate risk and 87 (55.1%) as high risk. Patients in the EORTC high risk group with low mIS showed a shorter PFS in comparison to high mIS (HR 2.9, CI 0.79 - 11.0, p=0.082). In contrast, no predictive potential regarding PFS was observed in intermediate or low risk groups. Furthermore, mIS was not able to predict RFS in any EORTC risk group. mIS could be utilized to predict prognosis more accurately in high-risk patients with NMIBC by identifying those with higher or lower risk of progression. Therefore, mIS could be used to allocate these highrisk patients to more streamlined follow-up or more aggressive treatment strategies.
Collapse
Affiliation(s)
- Uwe Bieri
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Dominik Enderlin
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Lorenz Buser
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Marian S. Wettstein
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Cédric Poyet
- Department of Urology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Poole A, Karuppiah V, Hartt A, Haidar JN, Moureau S, Dobrzycki T, Hayes C, Rowley C, Dias J, Harper S, Barnbrook K, Hock M, Coles C, Yang W, Aleksic M, Lin AB, Robinson R, Dukes JD, Liddy N, Van der Kamp M, Plowman GD, Vuidepot A, Cole DK, Whale AD, Chillakuri C. Therapeutic high affinity T cell receptor targeting a KRAS G12D cancer neoantigen. Nat Commun 2022; 13:5333. [PMID: 36088370 PMCID: PMC9464187 DOI: 10.1038/s41467-022-32811-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 08/16/2022] [Indexed: 11/09/2022] Open
Abstract
Neoantigens derived from somatic mutations are specific to cancer cells and are ideal targets for cancer immunotherapy. KRAS is the most frequently mutated oncogene and drives the pathogenesis of several cancers. Here we show the identification and development of an affinity-enhanced T cell receptor (TCR) that recognizes a peptide derived from the most common KRAS mutant, KRASG12D, presented in the context of HLA-A*11:01. The affinity of the engineered TCR is increased by over one million-fold yet fully able to distinguish KRASG12D over KRASWT. While crystal structures reveal few discernible differences in TCR interactions with KRASWT versus KRASG12D, thermodynamic analysis and molecular dynamics simulations reveal that TCR specificity is driven by differences in indirect electrostatic interactions. The affinity enhanced TCR, fused to a humanized anti-CD3 scFv, enables selective killing of cancer cells expressing KRASG12D. Our work thus reveals a molecular mechanism that drives TCR selectivity and describes a soluble bispecific molecule with therapeutic potential against cancers harboring a common shared neoantigen.
Collapse
Affiliation(s)
- Andrew Poole
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | | | - Annabelle Hartt
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, USA
| | - Jaafar N Haidar
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Sylvie Moureau
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Tomasz Dobrzycki
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Conor Hayes
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | | | - Jorge Dias
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Stephen Harper
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Keir Barnbrook
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Miriam Hock
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Charlotte Coles
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Wei Yang
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Milos Aleksic
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Aimee Bence Lin
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Ross Robinson
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Joe D Dukes
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Nathaniel Liddy
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Marc Van der Kamp
- School of Biochemistry, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, USA
| | - Gregory D Plowman
- Eli Lilly & Co, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Annelise Vuidepot
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - David K Cole
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA
| | - Andrew D Whale
- Immunocore Ltd., 92 Park Drive, Milton Park, Abingdon, OX14 4RY, USA.
| | | |
Collapse
|
14
|
Yu G, He X, Li X, Wu Y. Driving neoantigen-based cancer vaccines for personalized immunotherapy into clinic: A burdensome journey to promising land. Biomed Pharmacother 2022; 153:113464. [DOI: 10.1016/j.biopha.2022.113464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022] Open
|
15
|
Neoantigen: A Promising Target for the Immunotherapy of Colorectal Cancer. DISEASE MARKERS 2022; 2022:8270305. [PMID: 35211210 PMCID: PMC8863477 DOI: 10.1155/2022/8270305] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/28/2022] [Indexed: 02/05/2023]
Abstract
At present, there are various treatment strategies for colorectal cancer, including surgery, chemotherapy, radiotherapy, and targeted therapy. In recent years, with the continuous development of immunotherapy, immune checkpoint inhibitors (ICIs) can significantly improve the treatment of advanced colorectal cancer patients with high levels of microsatellite instability. In addition to ICIs, neoantigens, as a class of tumor-specific antigens (TSA), are regarded as new immunotherapy targets for many cancer species and are being explored for antitumor therapy. Immunotherapy strategies based on neoantigens include tumor vaccines and adoptive cell therapy (ACT). These methods aim to eliminate tumor cells by enhancing the immune response of host T-cells to neoantigens. In addition, for MSS colorectal cancer, such “cold tumors” with low mutation rates and stable microsatellites are not sensitive to ICIs, whereas neoantigens could provide a promising immunotherapeutic avenue. In this review, we summarized the current status of colorectal cancer neoantigen prediction and current clinical trials of neoantigens and discussed the difficulties and limitations of neoantigens-based therapies for the treatment of CRC.
Collapse
|
16
|
Neoantigen Cancer Vaccines: Generation, Optimization, and Therapeutic Targeting Strategies. Vaccines (Basel) 2022; 10:vaccines10020196. [PMID: 35214655 PMCID: PMC8877108 DOI: 10.3390/vaccines10020196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/30/2022] Open
Abstract
Alternatives to conventional cancer treatments are highly sought after for high-risk malignancies that have a poor response to established treatment modalities. With research advancing rapidly in the past decade, neoantigen-based immunotherapeutic approaches represent an effective and highly tolerable therapeutic option. Neoantigens are tumor-specific antigens that are not expressed in normal cells and possess significant immunogenic potential. Several recent studies have described the conceptual framework and methodologies to generate neoantigen-based vaccines as well as the formulation of appropriate clinical trials to advance this approach for patient care. This review aims to describe some of the key studies in the recent literature in this rapidly evolving field and summarize the current advances in neoantigen identification and selection, vaccine generation and delivery, and the optimization of neoantigen-based therapeutic strategies, including the early data from pivotal clinical studies.
Collapse
|
17
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2022; 172:xiii-xxiii. [PMID: 36064230 PMCID: PMC10087864 DOI: 10.1016/s0091-679x(22)00139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Wang P, Chen Y, Wang C. Beyond Tumor Mutation Burden: Tumor Neoantigen Burden as a Biomarker for Immunotherapy and Other Types of Therapy. Front Oncol 2021; 11:672677. [PMID: 33996601 PMCID: PMC8117238 DOI: 10.3389/fonc.2021.672677] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy has significantly improved the clinical outcome of patients with cancer. However, the immune response rate varies greatly, possibly due to lack of effective biomarkers that can be used to distinguish responders from non-responders. Recently, clinical studies have associated high tumor neoantigen burden (TNB) with improved outcomes in patients treated with immunotherapy. Therefore, TNB has emerged as a biomarker for immunotherapy and other types of therapy. In the present review, the potential application of TNB as a biomarker was evaluated. The methods of neoantigen prediction were summarized and the mechanisms involved in TNB were investigated. The impact of high TNB and increased number of infiltrating immune cells on the efficacy of immunotherapy was also addressed. Finally, the future challenges of TNB were discussed.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Biotherapy, Cancer Center, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Yueyun Chen
- Department of Biotherapy, Cancer Center, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Chun Wang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Xia J, Bai P, Fan W, Li Q, Li Y, Wang D, Yin L, Zhou Y. NEPdb: A Database of T-Cell Experimentally-Validated Neoantigens and Pan-Cancer Predicted Neoepitopes for Cancer Immunotherapy. Front Immunol 2021; 12:644637. [PMID: 33927717 PMCID: PMC8078594 DOI: 10.3389/fimmu.2021.644637] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/12/2021] [Indexed: 12/30/2022] Open
Abstract
T-cell recognition of somatic mutation-derived cancer neoepitopes can lead to tumor regression. Due to the difficulty to identify effective neoepitopes, constructing a database for sharing experimentally validated cancer neoantigens will be beneficial to precise cancer immunotherapy. Meanwhile, the routine neoepitope prediction in silico is important but laborious for clinical use. Here we present NEPdb, a database that contains more than 17,000 validated human immunogenic neoantigens and ineffective neoepitopes within human leukocyte antigens (HLAs) via curating published literature with our semi-automatic pipeline. Furthermore, NEPdb also provides pan-cancer level predicted HLA-I neoepitopes derived from 16,745 shared cancer somatic mutations, using state-of-the-art predictors. With a well-designed search engine and visualization modes, this database would enhance the efficiency of neoantigen-based cancer studies and treatments. NEPdb is freely available at http://nep.whu.edu.cn/.
Collapse
Affiliation(s)
- Jiaqi Xia
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Peng Bai
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Weiliang Fan
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qiming Li
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yongzheng Li
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Dehe Wang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China.,Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|