1
|
Carri I, Schwab E, Trivino JC, von Euw EM, Nielsen M, Mordoh J, Barrio MM. VACCIMEL, an allogeneic melanoma vaccine, efficiently triggers T cell immune responses against neoantigens and alloantigens, as well as against tumor-associated antigens. Front Immunol 2025; 15:1496204. [PMID: 39840067 PMCID: PMC11747570 DOI: 10.3389/fimmu.2024.1496204] [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: 09/13/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
VACCIMEL is a therapeutic cancer vaccine composed of four irradiated allogeneic human melanoma cell lines rationally selected to cover a wide range of melanoma tumor-associated antigens (TAA). We previously demonstrated that vaccination in the adjuvant setting prolonged the distant-metastasis-free survival of cutaneous melanoma patients and that T cells reactive to TAA and the patient's private neoantigens increased during treatment. However, immune responses directed to vaccine antigens that may arise from VACCIMEL's somatic mutations and human polymorphisms remain unexplored. To study these immunogens, we performed whole-exome sequencing of paired tumor and germinal samples from four vaccinated patients and the vaccine cells. VACCIMEL variants were called by comparing the vaccine and the patient's exomes, and non-synonymous coding variants were used to predict T cell epitopes. Candidates were ranked based on their mRNA expression in VACCIMEL, predicted peptide-HLA (pHLA) presentation, and pHLA stability. Then, the immune responses to prioritized epitope candidates were tested using IFNγ ELISpot assays on vaccinated patients' PBMC samples. The comparison of the vaccine with the patients' germinal exomes revealed on average 9481 coding non-synonymous variants, suggesting that VACCIMEL offers a high number of potential antigens. Between 0,05 and 0,2% of these variants were also found in the tumors of three vaccinated patients; however, one patient with a high tumor mutational burden (TMB) shared 19,5% somatic variants. The assessment of T cell responses showed that vaccinated patients mounted highly diverse responses against VACCIMEL peptides. Notably, effector T cells targeting the patient's tumor antigens, comprising neoantigens and TAA, were found in higher frequencies than T cells targeting VACCIMEL-exclusive antigens. On the other hand, we observed that the immunogenic epitopes are not conserved across patients, despite sharing HLA and that immune responses fluctuate over time. Finally, a positive correlation between VACCIMEL antigen expression and the intensity of the T cell responses was found. Our results demonstrate that the immune system simultaneously responds to a high number of antigens, either vaccinal or private, proving that immune responses against epitopes not expressed in the patient's tumors were not detrimental to the immune recognition of neoantigens and TAA.
Collapse
Affiliation(s)
- Ibel Carri
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Erika Schwab
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Erika M. von Euw
- Translational Oncology Research Labs, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Morten Nielsen
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) – Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - José Mordoh
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas (FUCA), Fundación Cáncer, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
2
|
Thrift WJ, Lounsbury NW, Broadwell Q, Heidersbach A, Freund E, Abdolazimi Y, Phung QT, Chen J, Capietto AH, Tong AJ, Rose CM, Blanchette C, Lill JR, Haley B, Delamarre L, Bourgon R, Liu K, Jhunjhunwala S. Towards designing improved cancer immunotherapy targets with a peptide-MHC-I presentation model, HLApollo. Nat Commun 2024; 15:10752. [PMID: 39737928 DOI: 10.1038/s41467-024-54887-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Based on the success of cancer immunotherapy, personalized cancer vaccines have emerged as a leading oncology treatment. Antigen presentation on MHC class I (MHC-I) is crucial for the adaptive immune response to cancer cells, necessitating highly predictive computational methods to model this phenomenon. Here, we introduce HLApollo, a transformer-based model for peptide-MHC-I (pMHC-I) presentation prediction, leveraging the language of peptides, MHC, and source proteins. HLApollo provides end-to-end treatment of MHC-I sequences and deconvolution of multi-allelic data, using a negative-set switching strategy to mitigate misassigned negatives in unlabelled ligandome data. HLApollo shows a 12.65% increase in average precision (AP) on ligandome data and a 4.1% AP increase on immunogenicity test data compared to next-best models. Incorporating protein features from protein language models yields further gains and reduces the need for gene expression measurements. Guided by clinical use, we demonstrate pan-allelic generalization which effectively captures rare alleles in underrepresented ancestries.
Collapse
Affiliation(s)
- William John Thrift
- Early Clinical Development Artificial Intelligence, Genentech, South San Francisco, CA, USA
| | | | - Quade Broadwell
- Early Clinical Development Artificial Intelligence, Genentech, South San Francisco, CA, USA
| | - Amy Heidersbach
- Molecular Biology Department, Genentech, South San Francisco, CA, USA
| | - Emily Freund
- Molecular Biology Department, Genentech, South San Francisco, CA, USA
| | - Yassan Abdolazimi
- Molecular Biology Department, Genentech, South San Francisco, CA, USA
| | - Qui T Phung
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Jieming Chen
- Oncology Bioinformatics, Genentech, South San Francisco, CA, USA
| | | | - Ann-Jay Tong
- Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Christopher M Rose
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | | | - Jennie R Lill
- Microchemistry, Proteomics and Lipidomics, Genentech, South San Francisco, CA, USA
| | - Benjamin Haley
- Molecular Biology Department, Genentech, South San Francisco, CA, USA
| | | | - Richard Bourgon
- Oncology Bioinformatics, Genentech, South San Francisco, CA, USA
- Computational Science, Freenome, South San Francisco, CA, USA
| | - Kai Liu
- Early Clinical Development Artificial Intelligence, Genentech, South San Francisco, CA, USA.
- Artificial Intelligence, SES AI, Woburn, MA, USA.
| | | |
Collapse
|
3
|
Baron M, Labreche K, Veyri M, Désiré N, Bouzidi A, Seck-Thiam F, Charlotte F, Rousseau A, Morin V, Nakid-Cordero C, Abbar B, Picca A, Le Cann M, Balegroune N, Gauthier N, Theodorou I, Touat M, Morel V, Bielle F, Samri A, Alentorn A, Sanson M, Roos-Weil D, Haioun C, Poullot E, De Septenville AL, Davi F, Guihot A, Boelle PY, Leblond V, Coulet F, Spano JP, Choquet S, Autran B. Epstein-Barr virus and immune status imprint the immunogenomics of non-Hodgkin lymphomas occurring in immune-suppressed environments. Haematologica 2024; 109:3615-3630. [PMID: 38841782 PMCID: PMC11532699 DOI: 10.3324/haematol.2023.284332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Non-Hodgkin lymphomas (NHL) commonly occur in immunodeficient patients, both those infected by human immunodeficiency virus (HIV) and those who have been transplanted, and are often driven by Epstein-Barr virus (EBV) with cerebral localization, raising the question of tumor immunogenicity, a critical issue for treatment responses. We investigated the immunogenomics of 68 lymphoproliferative disorders from 51 immunodeficient (34 post-transplant, 17 HIV+) and 17 immunocompetent patients. Overall, 72% were large B-cell lymphoma and 25% were primary central nervous system lymphoma, while 40% were EBV+. Tumor whole-exome and RNA sequencing, along with a bioinformatics pipeline allowed analysis of tumor mutational burden, tumor landscape and tumor microenvironment and prediction of tumor neoepitopes. Both tumor mutational burden (2.2 vs. 3.4/Mb, P=0.001) and numbers of neoepitopes (40 vs. 200, P=0.00019) were lower in EBV+ than in EBV- NHL, regardless of the immune status. In contrast both EBV and the immune status influenced the tumor mutational profile, with HNRNPF and STAT3 mutations observed exclusively in EBV+ and immunodeficient NHL, respectively. Peripheral blood T-cell responses against tumor neoepitopes were detected in all EBV- cases but in only half of the EBV+ ones, including responses against IgH-derived MHC-class-II restricted neoepitopes. The tumor microenvironment analysis showed higher CD8 T-cell infiltrates in EBV+ versus EBV- NHL, together with a more tolerogenic profile composed of regulatory T cells, type-M2 macrophages and an increased expression of negative immune-regulators. Our results highlight that the immunogenomics of NHL in patients with immunodeficiency primarily relies on the tumor EBV status, while T-cell recognition of tumor- and IgH-specific neoepitopes is conserved in EBV- patients, offering potential opportunities for future T-cell-based immune therapies.
Collapse
Affiliation(s)
- Marine Baron
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris, France; Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris.
| | - Karim Labreche
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Marianne Veyri
- Sorbonne Université, INSERM, Pierre et Louis Institute of Epidemiology and Public Health, F-75013 Paris France, Theravir Team, Department of Medical Oncology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Nathalie Désiré
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Amira Bouzidi
- Sorbonne Université, INSERM, Research Unit on Cardiovascular and Metabolic Disease UMR ICAN, Department of Endocrine Biochemistry and Oncology, AP-HP, Hôpital-Pitié-Salpêtrière, F-75013 Paris
| | - Fatou Seck-Thiam
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Frédéric Charlotte
- Sorbonne Université, Department of Anatomy and Pathologic Cytology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Alice Rousseau
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Véronique Morin
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Cécilia Nakid-Cordero
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Baptiste Abbar
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Alberto Picca
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Marie Le Cann
- Department of Clinical Haematology, AP-HP, Hôpital Kremlin Bicêtre, F-94270 Le Kremlin
| | - Noureddine Balegroune
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Nicolas Gauthier
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | | | - Mehdi Touat
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Véronique Morel
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Franck Bielle
- Sorbonne Université, Department of Neuropathology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013, Paris
| | - Assia Samri
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Agusti Alentorn
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Marc Sanson
- Sorbonne Université, INSERM, CNRS, Brain and Spine Institute, ICM, Department of Neurology 2-Mazarin, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Damien Roos-Weil
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Corinne Haioun
- Lymphoid malignancies Unit, AP-HP, Mondor Hospital, F-94000 Créteil
| | - Elsa Poullot
- Department of Anatomy and Pathologic Cytology, AP-HP, Mondor Hospital, F-94000 Créteil
| | - Anne Langlois De Septenville
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Department of Biological Hematology, AP-HP, Hôpital Pitié-Salpêtrière, Paris
| | - Frédéric Davi
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Department of Biological Hematology, AP-HP, Hôpital Pitié-Salpêtrière, Paris
| | - Amélie Guihot
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Pierre-Yves Boelle
- Sorbonne Université, CinBioS, UMS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, 75013 Paris
| | - Véronique Leblond
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Florence Coulet
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Microsatellites Instability and Cancer, CRSA, Department of Medical Genetics, AP-HP, Pitié-Salpêtrière Hospital, F-75013 Paris
| | - Jean-Philippe Spano
- Sorbonne Université, INSERM, Pierre et Louis Institute of Epidemiology and Public Health, F-75013 Paris France, Theravir Team, Department of Medical Oncology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Sylvain Choquet
- Sorbonne Université, Department of Clinical Haematology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| | - Brigitte Autran
- Sorbonne Université, INSERM U1135, Center for Immunology and Infectious Diseases (CIMI), Department of Immunology, AP-HP, Hôpital Pitié-Salpêtrière, F-75013 Paris
| |
Collapse
|
4
|
Bresser K, Nicolet BP, Jeko A, Wu W, Loayza-Puch F, Agami R, Heck AJR, Wolkers MC, Schumacher TN. Gene and protein sequence features augment HLA class I ligand predictions. Cell Rep 2024; 43:114325. [PMID: 38870014 DOI: 10.1016/j.celrep.2024.114325] [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: 10/20/2023] [Revised: 04/22/2024] [Accepted: 05/22/2024] [Indexed: 06/15/2024] Open
Abstract
The sensitivity of malignant tissues to T cell-based immunotherapies depends on the presence of targetable human leukocyte antigen (HLA) class I ligands. Peptide-intrinsic factors, such as HLA class I affinity and proteasomal processing, have been established as determinants of HLA ligand presentation. However, the role of gene and protein sequence features as determinants of epitope presentation has not been systematically evaluated. We perform HLA ligandome mass spectrometry to evaluate the contribution of 7,135 gene and protein sequence features to HLA sampling. This analysis reveals that a number of predicted modifiers of mRNA and protein abundance and turnover, including predicted mRNA methylation and protein ubiquitination sites, inform on the presence of HLA ligands. Importantly, integration of such "hard-coded" sequence features into a machine learning approach augments HLA ligand predictions to a comparable degree as experimental measures of gene expression. Our study highlights the value of gene and protein features for HLA ligand predictions.
Collapse
Affiliation(s)
- Kaspar Bresser
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, the Netherlands; Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Benoit P Nicolet
- Sanquin Blood Supply Foundation, Department of Research, T cell differentiation lab, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Landsteiner Laboratory, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Anita Jeko
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, the Netherlands
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, the Netherlands
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, the Netherlands
| | - Monika C Wolkers
- Sanquin Blood Supply Foundation, Department of Research, T cell differentiation lab, Amsterdam, The Netherlands; Amsterdam UMC, University of Amsterdam, Landsteiner Laboratory, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Ton N Schumacher
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, the Netherlands; Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
5
|
Weisbrod L, Capriotti L, Hofmann M, Spieler V, Dersch H, Voedisch B, Schmidt P, Knake S. FASTMAP-a flexible and scalable immunopeptidomics pipeline for HLA- and antigen-specific T-cell epitope mapping based on artificial antigen-presenting cells. Front Immunol 2024; 15:1386160. [PMID: 38779658 PMCID: PMC11109385 DOI: 10.3389/fimmu.2024.1386160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
The study of peptide repertoires presented by major histocompatibility complex (MHC) molecules and the identification of potential T-cell epitopes contribute to a multitude of immunopeptidome-based treatment approaches. Epitope mapping is essential for the development of promising epitope-based approaches in vaccination as well as for innovative therapeutics for autoimmune diseases, infectious diseases, and cancer. It also plays a critical role in the immunogenicity assessment of protein therapeutics with regard to safety and efficacy concerns. The main challenge emerges from the highly polymorphic nature of the human leukocyte antigen (HLA) molecules leading to the requirement of a peptide mapping strategy for a single HLA allele. As many autoimmune diseases are linked to at least one specific antigen, we established FASTMAP, an innovative strategy to transiently co-transfect a single HLA allele combined with a disease-specific antigen into a human cell line. This approach allows the specific identification of HLA-bound peptides using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Using FASTMAP, we found a comparable spectrum of endogenous peptides presented by the most frequently expressed HLA alleles in the world's population compared to what has been described in literature. To ensure a reliable peptide mapping workflow, we combined the HLA alleles with well-known human model antigens like coagulation factor VIII, acetylcholine receptor subunit alpha, protein structures of the SARS-CoV-2 virus, and myelin basic protein. Using these model antigens, we have been able to identify a broad range of peptides that are in line with already published and in silico predicted T-cell epitopes of the specific HLA/model antigen combination. The transient co-expression of a single affinity-tagged MHC molecule combined with a disease-specific antigen in a human cell line in our FASTMAP pipeline provides the opportunity to identify potential T-cell epitopes/endogenously processed MHC-bound peptides in a very cost-effective, fast, and customizable system with high-throughput potential.
Collapse
Affiliation(s)
- Luisa Weisbrod
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Luigi Capriotti
- Analytical Biochemistry, Research and Development, CSL Behring AG, Bern, Switzerland
| | - Marco Hofmann
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Valerie Spieler
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Herbert Dersch
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Bernd Voedisch
- Recombinant Protein Discovery, CSL Innovation GmbH, Marburg, Germany
| | - Peter Schmidt
- Protein Biochemistry, Bio21 Institute, CSL Limited, Parkville, VIC, Australia
| | - Susanne Knake
- Department of Neurology, Epilepsy Center Hessen, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
6
|
Borch A, Carri I, Reynisson B, Alvarez HMG, Munk KK, Montemurro A, Kristensen NP, Tvingsholm SA, Holm JS, Heeke C, Moss KH, Hansen UK, Schaap-Johansen AL, Bagger FO, de Lima VAB, Rohrberg KS, Funt SA, Donia M, Svane IM, Lassen U, Barra C, Nielsen M, Hadrup SR. IMPROVE: a feature model to predict neoepitope immunogenicity through broad-scale validation of T-cell recognition. Front Immunol 2024; 15:1360281. [PMID: 38633261 PMCID: PMC11021644 DOI: 10.3389/fimmu.2024.1360281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/07/2024] [Indexed: 04/19/2024] Open
Abstract
Background Mutation-derived neoantigens are critical targets for tumor rejection in cancer immunotherapy, and better tools for neoepitope identification and prediction are needed to improve neoepitope targeting strategies. Computational tools have enabled the identification of patient-specific neoantigen candidates from sequencing data, but limited data availability has hindered their capacity to predict which of the many neoepitopes will most likely give rise to T cell recognition. Method To address this, we make use of experimentally validated T cell recognition towards 17,500 neoepitope candidates, with 467 being T cell recognized, across 70 cancer patients undergoing immunotherapy. Results We evaluated 27 neoepitope characteristics, and created a random forest model, IMPROVE, to predict neoepitope immunogenicity. The presence of hydrophobic and aromatic residues in the peptide binding core were the most important features for predicting neoepitope immunogenicity. Conclusion Overall, IMPROVE was found to significantly advance the identification of neoepitopes compared to other current methods.
Collapse
Affiliation(s)
- Annie Borch
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Ibel Carri
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Birkir Reynisson
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Heli M. Garcia Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Kamilla K. Munk
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | | | - Siri A. Tvingsholm
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Jeppe Sejerø Holm
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Christina Heeke
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Keith Henry Moss
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Ulla Kring Hansen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | | | | | | | | | - Samuel A. Funt
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Marco Donia
- National Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Ulrik Lassen
- Department of Oncology, Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Carolina Barra
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
7
|
Wan YTR, Koşaloğlu‐Yalçın Z, Peters B, Nielsen M. A large-scale study of peptide features defining immunogenicity of cancer neo-epitopes. NAR Cancer 2024; 6:zcae002. [PMID: 38288446 PMCID: PMC10823584 DOI: 10.1093/narcan/zcae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
Accurate prediction of immunogenicity for neo-epitopes arising from a cancer associated mutation is a crucial step in many bioinformatics pipelines that predict outcome of checkpoint blockade treatments or that aim to design personalised cancer immunotherapies and vaccines. In this study, we performed a comprehensive analysis of peptide features relevant for prediction of immunogenicity using the Cancer Epitope Database and Analysis Resource (CEDAR), a curated database of cancer epitopes with experimentally validated immunogenicity annotations from peer-reviewed publications. The developed model, ICERFIRE (ICore-based Ensemble Random Forest for neo-epitope Immunogenicity pREdiction), extracts the predicted ICORE from the full neo-epitope as input, i.e. the nested peptide with the highest predicted major histocompatibility complex (MHC) binding potential combined with its predicted likelihood of antigen presentation (%Rank). Key additional features integrated into the model include assessment of the BLOSUM mutation score of the neo-epitope, and antigen expression levels of the wild-type counterpart which is often reflecting a neo-epitope's abundance. We demonstrate improved and robust performance of ICERFIRE over existing immunogenicity and epitope prediction models, both in cross-validation and on external validation datasets.
Collapse
Affiliation(s)
- Yat-tsai Richie Wan
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, DK 28002, Denmark
| | - Zeynep Koşaloğlu‐Yalçın
- Center for Infectious Disease and Vaccine Research, La Jolla Institute of Immunology, La Jolla, CA 92037, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute of Immunology, La Jolla, CA 92037, USA
| | - Morten Nielsen
- Department of Health Technology, Technical University of Denmark, Kgs. Lyngby, DK 28002, Denmark
| |
Collapse
|
8
|
Barra C, Nilsson JB, Saksager A, Carri I, Deleuran S, Garcia Alvarez HM, Høie MH, Li Y, Clifford JN, Wan YTR, Moreta LS, Nielsen M. In Silico Tools for Predicting Novel Epitopes. Methods Mol Biol 2024; 2813:245-280. [PMID: 38888783 DOI: 10.1007/978-1-0716-3890-3_17] [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] [Indexed: 06/20/2024]
Abstract
Identifying antigens within a pathogen is a critical task to develop effective vaccines and diagnostic methods, as well as understanding the evolution and adaptation to host immune responses. Historically, antigenicity was studied with experiments that evaluate the immune response against selected fragments of pathogens. Using this approach, the scientific community has gathered abundant information regarding which pathogenic fragments are immunogenic. The systematic collection of this data has enabled unraveling many of the fundamental rules underlying the properties defining epitopes and immunogenicity, and has resulted in the creation of a large panel of immunologically relevant predictive (in silico) tools. The development and application of such tools have proven to accelerate the identification of novel epitopes within biomedical applications reducing experimental costs. This chapter introduces some basic concepts about MHC presentation, T cell and B cell epitopes, the experimental efforts to determine those, and focuses on state-of-the-art methods for epitope prediction, highlighting their strengths and limitations, and catering instructions for their rational use.
Collapse
Affiliation(s)
- Carolina Barra
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark.
| | | | - Astrid Saksager
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | - Ibel Carri
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| | - Sebastian Deleuran
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | - Heli M Garcia Alvarez
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| | - Magnus Haraldson Høie
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | - Yuchen Li
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | | | - Yat-Tsai Richie Wan
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | - Lys Sanz Moreta
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
| | - Morten Nielsen
- Section for Bioinformatics, Health Tech, Technical University of Denmark, Lyngby, Denmark
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Argentina
| |
Collapse
|
9
|
Contemplating immunopeptidomes to better predict them. Semin Immunol 2023; 66:101708. [PMID: 36621290 DOI: 10.1016/j.smim.2022.101708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
The identification of T-cell epitopes is key for a complete molecular understanding of immune recognition mechanisms in infectious diseases, autoimmunity and cancer. T-cell epitopes further provide targets for personalized vaccines and T-cell therapy, with several therapeutic applications in cancer immunotherapy and elsewhere. T-cell epitopes consist of short peptides displayed on Major Histocompatibility Complex (MHC) molecules. The recent advances in mass spectrometry (MS) based technologies to profile the ensemble of peptides displayed on MHC molecules - the so-called immunopeptidome - had a major impact on our understanding of antigen presentation and MHC ligands. On the one hand, these techniques enabled researchers to directly identify hundreds of thousands of peptides presented on MHC molecules, including some that elicited T-cell recognition. On the other hand, the data collected in these experiments revealed fundamental properties of antigen presentation pathways and significantly improved our ability to predict naturally presented MHC ligands and T-cell epitopes across the wide spectrum of MHC alleles found in human and other organisms. Here we review recent computational developments to analyze experimentally determined immunopeptidomes and harness these data to improve our understanding of antigen presentation and MHC binding specificities, as well as our ability to predict MHC ligands. We further discuss the strengths and limitations of the latest approaches to move beyond predictions of antigen presentation and tackle the challenges of predicting TCR recognition and immunogenicity.
Collapse
|
10
|
Frentzen A, Greenbaum JA, Kim H, Peters B, Koşaloğlu-Yalçın Z. Estimating tissue-specific peptide abundance from public RNA-Seq data. Front Genet 2023; 14:1082168. [PMID: 36713080 PMCID: PMC9878344 DOI: 10.3389/fgene.2023.1082168] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Several novel MHC class I epitope prediction tools additionally incorporate the abundance levels of the peptides' source antigens and have shown improved performance for predicting immunogenicity. Such tools require the user to input the MHC alleles and peptide sequences of interest, as well as the abundance levels of the peptides' source proteins. However, such expression data is often not directly available to users, and retrieving the expression level of a peptide's source antigen from public databases is not trivial. We have developed the Peptide eXpression annotator (pepX), which takes a peptide as input, identifies from which proteins the peptide can be derived, and returns an estimate of the expression level of those source proteins from selected public databases. We have also investigated how the abundance level of a peptide can be best estimated in cases when it can originate from multiple transcripts and proteins and found that summing up transcript-level expression values performs best in distinguishing ligands from decoy peptides.
Collapse
Affiliation(s)
- Angela Frentzen
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Jason A. Greenbaum
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Haeuk Kim
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, United States
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, United States,Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Zeynep Koşaloğlu-Yalçın
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, San Diego, CA, United States,*Correspondence: Zeynep Koşaloğlu-Yalçın,
| |
Collapse
|