51
|
Malviya M, Aretz Z, Molvi Z, Lee J, Pierre S, Wallisch P, Dao T, Scheinberg DA. Challenges and solutions for therapeutic TCR-based agents. Immunol Rev 2023; 320:58-82. [PMID: 37455333 PMCID: PMC11141734 DOI: 10.1111/imr.13233] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/18/2023] [Indexed: 07/18/2023]
Abstract
Recent development of methods to discover and engineer therapeutic T-cell receptors (TCRs) or antibody mimics of TCRs, and to understand their immunology and pharmacology, lag two decades behind therapeutic antibodies. Yet we have every expectation that TCR-based agents will be similarly important contributors to the treatment of a variety of medical conditions, especially cancers. TCR engineered cells, soluble TCRs and their derivatives, TCR-mimic antibodies, and TCR-based CAR T cells promise the possibility of highly specific drugs that can expand the scope of immunologic agents to recognize intracellular targets, including mutated proteins and undruggable transcription factors, not accessible by traditional antibodies. Hurdles exist regarding discovery, specificity, pharmacokinetics, and best modality of use that will need to be overcome before the full potential of TCR-based agents is achieved. HLA restriction may limit each agent to patient subpopulations and off-target reactivities remain important barriers to widespread development and use of these new agents. In this review we discuss the unique opportunities for these new classes of drugs, describe their unique antigenic targets, compare them to traditional antibody therapeutics and CAR T cells, and review the various obstacles that must be overcome before full application of these drugs can be realized.
Collapse
Affiliation(s)
- Manish Malviya
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Zita Aretz
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Physiology, Biophysics & Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Zaki Molvi
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Physiology, Biophysics & Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Jayop Lee
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Stephanie Pierre
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Tri-Institutional Medical Scientist Program, 1300 York Avenue, New York, NY 10021
| | - Patrick Wallisch
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| |
Collapse
|
52
|
Giordano Attianese GMP, Ash S, Irving M. Coengineering specificity, safety, and function into T cells for cancer immunotherapy. Immunol Rev 2023; 320:166-198. [PMID: 37548063 DOI: 10.1111/imr.13252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Adoptive T-cell transfer (ACT) therapies, including of tumor infiltrating lymphocytes (TILs) and T cells gene-modified to express either a T cell receptor (TCR) or a chimeric antigen receptor (CAR), have demonstrated clinical efficacy for a proportion of patients and cancer-types. The field of ACT has been driven forward by the clinical success of CD19-CAR therapy against various advanced B-cell malignancies, including curative responses for some leukemia patients. However, relapse remains problematic, in particular for lymphoma. Moreover, for a variety of reasons, relative limited efficacy has been demonstrated for ACT of non-hematological solid tumors. Indeed, in addition to pre-infusion challenges including lymphocyte collection and manufacturing, ACT failure can be attributed to several biological processes post-transfer including, (i) inefficient tumor trafficking, infiltration, expansion and retention, (ii) chronic antigen exposure coupled with insufficient costimulation resulting in T-cell exhaustion, (iii) a range of barriers in the tumor microenvironment (TME) mediated by both tumor cells and suppressive immune infiltrate, (iv) tumor antigen heterogeneity and loss, or down-regulation of antigen presentation machinery, (v) gain of tumor intrinsic mechanisms of resistance such as to apoptosis, and (vi) various forms of toxicity and other adverse events in patients. Affinity-optimized TCRs can improve T-cell function and innovative CAR designs as well as gene-modification strategies can be used to coengineer specificity, safety, and function into T cells. Coengineering strategies can be designed not only to directly support the transferred T cells, but also to block suppressive barriers in the TME and harness endogenous innate and adaptive immunity. Here, we review a selection of the remarkable T-cell coengineering strategies, including of tools, receptors, and gene-cargo, that have been developed in recent years to augment tumor control by ACT, more and more of which are advancing to the clinic.
Collapse
Affiliation(s)
- Greta Maria Paola Giordano Attianese
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sarah Ash
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
53
|
Patel M, Hudson O, Han J, Kondapalli L, Arora G, Hawi R, Andrikopoulou E, Estes C, Johnson AM, Lenneman C. Update on Immunotherapy Cardiotoxicity: Checkpoint Inhibitors, CAR T, and Beyond. Curr Treat Options Oncol 2023; 24:1489-1503. [PMID: 37624557 DOI: 10.1007/s11864-023-01130-y] [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] [Accepted: 07/18/2023] [Indexed: 08/26/2023]
Abstract
OPINION STATEMENT Immunotherapy is an innovative approach to cancer treatment that involves using the body's immune system to fight cancer. The landscape of immunotherapy is constantly evolving, as new therapies are developed and refined. Some of the most promising approaches in immunotherapy include immune checkpoint inhibitors (ICIs): these drugs target proteins on the surface of T-cells that inhibit their ability to attack cancer cells. By blocking these proteins, checkpoint inhibitors allow T-cells to recognize and destroy cancer cells more effectively. CAR T-cell therapy: this therapy involves genetically modifying a patient's own T-cells to recognize and attack cancer cells. CAR T-cell therapy exhibits favorable response in many patients with refractory hematological cancers with growing clinical trials in solid tumors. Immune system modulators: these drugs enhance the immune system's ability to fight cancer by stimulating the production of immune cells or inhibiting the activity of immune-suppressing cells. While immunotherapy has shown great promise in the treatment of cancer, it can also pose significant cardiac side effects. Some immunotherapy drugs like ICIs can cause myocarditis, which can lead to chest pain, shortness of breath, and heart failure. Other cardiac side effects of ICIs include arrhythmias, pericarditis, vasculitis, and accelerated atherosclerosis. It is important for patients receiving immunotherapy to be monitored closely for these side effects, as prompt treatment can help prevent serious complications. Patients should also report any symptoms to their healthcare providers right away, so that appropriate action can be taken. CAR T-cell therapy can also illicit an exaggerated immune response creating cytokine release syndrome (CRS) that may precipitate cardiovascular events: arrhythmias, myocardial infarction, and heart failure. Overall, while immune modulating therapy is a promising and expanding approach to cancer treatment, it is important to weigh the potential benefits against the risks and side effects, especially in patients with high risk for cardiovascular complications.
Collapse
Affiliation(s)
- Murti Patel
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Olivia Hudson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Jingnan Han
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Lavanya Kondapalli
- University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Garima Arora
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Riem Hawi
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | | | - Courtney Estes
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Abigail M Johnson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Carrie Lenneman
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
54
|
Apavaloaei A, Perreault C. Immunotargeting of a recurrent AML-specific neoantigen. NATURE CANCER 2023; 4:1403-1405. [PMID: 37783806 DOI: 10.1038/s43018-023-00634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
|
55
|
Schendel DJ. Evolution by innovation as a driving force to improve TCR-T therapies. Front Oncol 2023; 13:1216829. [PMID: 37810959 PMCID: PMC10552759 DOI: 10.3389/fonc.2023.1216829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Adoptive cell therapies continually evolve through science-based innovation. Specialized innovations for TCR-T therapies are described here that are embedded in an End-to-End Platform for TCR-T Therapy Development which aims to provide solutions for key unmet patient needs by addressing challenges of TCR-T therapy, including selection of target antigens and suitable T cell receptors, generation of TCR-T therapies that provide long term, durable efficacy and safety and development of efficient and scalable production of patient-specific (personalized) TCR-T therapy for solid tumors. Multiple, combinable, innovative technologies are used in a systematic and sequential manner in the development of TCR-T therapies. One group of technologies encompasses product enhancements that enable TCR-T therapies to be safer, more specific and more effective. The second group of technologies addresses development optimization that supports discovery and development processes for TCR-T therapies to be performed more quickly, with higher quality and greater efficiency. Each module incorporates innovations layered onto basic technologies common to the field of immunology. An active approach of "evolution by innovation" supports the overall goal to develop best-in-class TCR-T therapies for treatment of patients with solid cancer.
Collapse
Affiliation(s)
- Dolores J. Schendel
- Medigene Immunotherapies GmbH, Planegg, Germany
- Medigene AG, Planegg, Germany
| |
Collapse
|
56
|
Lee CH, Huh J, Buckley PR, Jang M, Pinho MP, Fernandes RA, Antanaviciute A, Simmons A, Koohy H. A robust deep learning workflow to predict CD8 + T-cell epitopes. Genome Med 2023; 15:70. [PMID: 37705109 PMCID: PMC10498576 DOI: 10.1186/s13073-023-01225-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: 01/30/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND T-cells play a crucial role in the adaptive immune system by triggering responses against cancer cells and pathogens, while maintaining tolerance against self-antigens, which has sparked interest in the development of various T-cell-focused immunotherapies. However, the identification of antigens recognised by T-cells is low-throughput and laborious. To overcome some of these limitations, computational methods for predicting CD8 + T-cell epitopes have emerged. Despite recent developments, most immunogenicity algorithms struggle to learn features of peptide immunogenicity from small datasets, suffer from HLA bias and are unable to reliably predict pathology-specific CD8 + T-cell epitopes. METHODS We developed TRAP (T-cell recognition potential of HLA-I presented peptides), a robust deep learning workflow for predicting CD8 + T-cell epitopes from MHC-I presented pathogenic and self-peptides. TRAP uses transfer learning, deep learning architecture and MHC binding information to make context-specific predictions of CD8 + T-cell epitopes. TRAP also detects low-confidence predictions for peptides that differ significantly from those in the training datasets to abstain from making incorrect predictions. To estimate the immunogenicity of pathogenic peptides with low-confidence predictions, we further developed a novel metric, RSAT (relative similarity to autoantigens and tumour-associated antigens), as a complementary to 'dissimilarity to self' from cancer studies. RESULTS TRAP was used to identify epitopes from glioblastoma patients as well as SARS-CoV-2 peptides, and it outperformed other algorithms in both cancer and pathogenic settings. TRAP was especially effective at extracting immunogenicity-associated properties from restricted data of emerging pathogens and translating them onto related species, as well as minimising the loss of likely epitopes in imbalanced datasets. We also demonstrated that the novel metric termed RSAT was able to estimate immunogenic of pathogenic peptides of various lengths and species. TRAP implementation is available at: https://github.com/ChloeHJ/TRAP . CONCLUSIONS This study presents a novel computational workflow for accurately predicting CD8 + T-cell epitopes to foster a better understanding of antigen-specific T-cell response and the development of effective clinical therapeutics.
Collapse
Affiliation(s)
- Chloe H Lee
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Jaesung Huh
- Visual Geometry Group, Department of Engineering Science, University of Oxford, Oxford, OX2 6NN, UK
| | - Paul R Buckley
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Myeongjun Jang
- Intelligent Systems Lab, Department of Computer Science, University of Oxford, Oxford, OX1 3QG, UK
| | - Mariana Pereira Pinho
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Ricardo A Fernandes
- Chinese Academy of Medical Sciences (CAMS) Oxford Institute (COI), University of Oxford, Oxford, OX3 7BN, UK
| | - Agne Antanaviciute
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
| | - Alison Simmons
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, OX3 9DS, UK
| | - Hashem Koohy
- MRC Human Immunology Unit, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DS, UK.
- Alan Turning Fellow in Health and Medicine, The Alan Turing Institute, London, UK.
| |
Collapse
|
57
|
Bouquet L, Bôle-Richard E, Warda W, Neto Da Rocha M, Trad R, Nicod C, Haderbache R, Genin D, Ferrand C, Deschamps M. RapaCaspase-9-based suicide gene applied to the safety of IL-1RAP CAR-T cells. Gene Ther 2023; 30:706-713. [PMID: 37173386 PMCID: PMC10506905 DOI: 10.1038/s41434-023-00404-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/30/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Even if adoptive cell transfer (ACT) has already shown great clinical efficiency in different types of disease, such as cancer, some adverse events consistently occur, and suicide genes are an interesting system to manage these events. Our team developed a new medical drug candidate, a chimeric antigen receptor (CAR) targeting interleukin-1 receptor accessory protein (IL-1RAP), which needs to be evaluated in clinical trials with a clinically applicable suicide gene system. To prevent side effects and ensure the safety of our candidate, we devised two constructs carrying an inducible suicide gene, RapaCasp9-G or RapaCasp9-A, containing a single-nucleotide polymorphism (rs1052576) affecting the efficiency of endogenous caspase 9. These suicide genes are activated by rapamycin and based on the fusion of human caspase 9 with a modified human FK-binding protein, allowing conditional dimerization. RapaCasp9-G- and RapaCasp9-A-expressing gene-modified T cells (GMTCs) were produced from healthy donors (HDs) and acute myeloid leukemia (AML) donors. The RapaCasp9-G suicide gene demonstrated better efficiency, and we showed its in vitro functionality in different clinically relevant culture conditions. Moreover, as rapamycin is not pharmacologically inert, we also demonstrated its safe use as part of our therapy.
Collapse
Affiliation(s)
- Lucie Bouquet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Elodie Bôle-Richard
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Walid Warda
- CanCell Therapeutics, 25 000, Besançon, France
| | | | - Rim Trad
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Clémentine Nicod
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Rafik Haderbache
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Delphine Genin
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Christophe Ferrand
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
- CanCell Therapeutics, 25 000, Besançon, France
| | - Marina Deschamps
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France.
- CanCell Therapeutics, 25 000, Besançon, France.
| |
Collapse
|
58
|
Yang K, Halima A, Chan TA. Antigen presentation in cancer - mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 2023; 20:604-623. [PMID: 37328642 DOI: 10.1038/s41571-023-00789-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour-immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA.
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA.
- National Center for Regenerative Medicine, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
59
|
Rihackova E, Rihacek M, Vyskocilova M, Valik D, Elbl L. Revisiting treatment-related cardiotoxicity in patients with malignant lymphoma-a review and prospects for the future. Front Cardiovasc Med 2023; 10:1243531. [PMID: 37711551 PMCID: PMC10499183 DOI: 10.3389/fcvm.2023.1243531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Treatment of malignant lymphoma has for years been represented by many cardiotoxic agents especially anthracyclines, cyclophosphamide, and thoracic irradiation. Although they are in clinical practice for decades, the precise mechanism of cardiotoxicity and effective prevention is still part of the research. At this article we discuss most routinely used anti-cancer drugs in chemotherapeutic regiments for malignant lymphoma with the focus on novel insight on molecular mechanisms of cardiotoxicity. Understanding toxicity at molecular levels may unveil possible targets of cardioprotective supportive therapy or optimization of current therapeutic protocols. Additionally, we review novel specific targeted therapy and its challenges in cardio-oncology.
Collapse
Affiliation(s)
- Eva Rihackova
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Michal Rihacek
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Biochemistry, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Maria Vyskocilova
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| | - Dalibor Valik
- Department of Laboratory Medicine, University Hospital Brno, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lubomir Elbl
- Department of Internal Medicine and Cardiology, University Hospital Brno and Faculty of Medicine of Masaryk University, Brno, Czech Republic
| |
Collapse
|
60
|
Foldvari Z, Knetter C, Yang W, Gjerdingen TJ, Bollineni RC, Tran TT, Lund-Johansen F, Kolstad A, Drousch K, Klopfleisch R, Leisegang M, Olweus J. A systematic safety pipeline for selection of T-cell receptors to enter clinical use. NPJ Vaccines 2023; 8:126. [PMID: 37607971 PMCID: PMC10444760 DOI: 10.1038/s41541-023-00713-y] [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: 10/19/2022] [Accepted: 07/31/2023] [Indexed: 08/24/2023] Open
Abstract
Cancer immunotherapy using T cell receptor-engineered T cells (TCR-Ts) represents a promising treatment option. However, technologies for pre-clinical safety assessment are incomplete or inaccessible to most laboratories. Here, TCR-T off-target reactivity was assessed in five steps: (1) Mapping target amino acids necessary for TCR-T recognition, followed by (2) a computational search for, and (3) reactivity screening against, candidate cross-reactive peptides in the human proteome. Natural processing and presentation of recognized peptides was evaluated using (4) short mRNAs, and (5) full-length proteins. TCR-Ts were screened for recognition of unintended HLA alleles, and as proxy for off-target reactivity in vivo, a syngeneic, HLA-A*02:01-transgenic mouse model was used. Validation demonstrated importance of studying recognition of full-length candidate off-targets, and that the clinically applied 1G4 TCR has a hitherto unknown reactivity to unintended HLA alleles, relevant for patient selection. This widely applicable strategy should facilitate evaluation of candidate therapeutic TCRs and inform clinical decision-making.
Collapse
Affiliation(s)
- Zsofia Foldvari
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Cathrine Knetter
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Weiwen Yang
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Thea Johanne Gjerdingen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Ravi Chand Bollineni
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Trung The Tran
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Kimberley Drousch
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, USA.
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
61
|
Cuevas MVR, Hardy MP, Larouche JD, Apavaloaei A, Kina E, Vincent K, Gendron P, Laverdure JP, Durette C, Thibault P, Lemieux S, Perreault C, Ehx G. BamQuery: a proteogenomic tool to explore the immunopeptidome and prioritize actionable tumor antigens. Genome Biol 2023; 24:188. [PMID: 37582761 PMCID: PMC10426134 DOI: 10.1186/s13059-023-03029-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
MHC-I-associated peptides deriving from non-coding genomic regions and mutations can generate tumor-specific antigens, including neoantigens. Quantifying tumor-specific antigens' RNA expression in malignant and benign tissues is critical for discriminating actionable targets. We present BamQuery, a tool attributing an exhaustive RNA expression to MHC-I-associated peptides of any origin from bulk and single-cell RNA-sequencing data. We show that many cryptic and mutated tumor-specific antigens can derive from multiple discrete genomic regions, abundantly expressed in normal tissues. BamQuery can also be used to predict MHC-I-associated peptides immunogenicity and identify actionable tumor-specific antigens de novo.
Collapse
Affiliation(s)
- Maria Virginia Ruiz Cuevas
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Marie-Pierre Hardy
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Jean-David Larouche
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Anca Apavaloaei
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Eralda Kina
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Krystel Vincent
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Patrick Gendron
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Jean-Philippe Laverdure
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Chantal Durette
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Pierre Thibault
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Chemistry, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Sébastien Lemieux
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Grégory Ehx
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Laboratory of Hematology, GIGA-I3, University of Liege, CHU of Liege, Liege, Belgium.
| |
Collapse
|
62
|
Pontoriero A, Critelli P, Chillari F, Ferrantelli G, Sciacca M, Brogna A, Parisi S, Pergolizzi S. Modulation of Radiation Doses and Chimeric Antigen Receptor T Cells: A Promising New Weapon in Solid Tumors-A Narrative Review. J Pers Med 2023; 13:1261. [PMID: 37623511 PMCID: PMC10455986 DOI: 10.3390/jpm13081261] [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: 06/29/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Tumor behavior is determined by its interaction with the tumor microenvironment (TME). Chimeric antigen receptor (CART) cell therapy represents a new form of cellular immunotherapy (IT). Immune cells present a different sensitivity to radiation therapy (RT). RT can affect tumor cells both modifying the TME and inducing DNA damage, with different effects depending on the low and high doses delivered, and can favor the expression of CART cells. CART cells are patients' T cells genetically engineered to recognize surface structure and to eradicate cancer cells. High-dose radiation therapy (HDRT, >10-20 Gy/fractions) converts immunologically "cold" tumors into "hot" ones by inducing necrosis and massive inflammation and death. LDRT (low-dose radiation therapy, >5-10 Gy/fractions) increases the expansion of CART cells and leads to non-immunogenetic death. An innovative approach, defined as the LATTICE technique, combines a high dose in higher FDG- uptake areas and a low dose to the tumor periphery. The association of RT and immune checkpoint inhibitors increases tumor immunogenicity and immune response both in irradiated and non-irradiated sites. The aim of this narrative review is to clarify the knowledge, to date, on CART cell therapy and its possible association with radiation therapy in solid tumors.
Collapse
Affiliation(s)
- Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Paola Critelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Federico Chillari
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Giacomo Ferrantelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Miriam Sciacca
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Anna Brogna
- Radiotherapy Unit, Medical Physics Unit, A.O.U. “G. Martino”, 98125 Messina, Italy;
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, 98125 Messina, Italy; (A.P.); (F.C.); (G.F.); (M.S.); (S.P.); (S.P.)
| |
Collapse
|
63
|
Du S, Yan J, Xue Y, Zhong Y, Dong Y. Adoptive cell therapy for cancer treatment. EXPLORATION (BEIJING, CHINA) 2023; 3:20210058. [PMID: 37933232 PMCID: PMC10624386 DOI: 10.1002/exp.20210058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/17/2023] [Indexed: 11/08/2023]
Abstract
Adoptive cell therapy (ACT) is a rapidly growing anti-cancer strategy that has shown promise in treating various cancer types. The concept of ACT involves activating patients' own immune cells ex vivo and then transferring them back to the patients to recognize and eliminate cancer cells. Currently, the commonly used ACT includes tumor-infiltrating lymphocytes (TILs), genetically engineered immune cells, and dendritic cells (DCs) vaccines. With the advancement of cell culture and genetic engineering techniques, ACT has been used in clinics to treat malignant hematological diseases and many new ACT-based regimens are in different stages of clinical trials. Here, representative ACT approaches are introduced and the opportunities and challenges for clinical translation of ACT are discussed.
Collapse
Affiliation(s)
- Shi Du
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Jingyue Yan
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yonger Xue
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yichen Zhong
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| | - Yizhou Dong
- Division of Pharmaceutics and PharmacologyCollege of PharmacyOhio State UniversityColumbusUSA
- Icahn Genomics InstitutePrecision Immunology InstituteDepartment of Oncological SciencesTisch Cancer InstituteFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkUSA
| |
Collapse
|
64
|
Rappazzo CG, Fernández-Quintero ML, Mayer A, Wu NC, Greiff V, Guthmiller JJ. Defining and Studying B Cell Receptor and TCR Interactions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:311-322. [PMID: 37459189 PMCID: PMC10495106 DOI: 10.4049/jimmunol.2300136] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/15/2023] [Indexed: 07/20/2023]
Abstract
BCRs (Abs) and TCRs (or adaptive immune receptors [AIRs]) are the means by which the adaptive immune system recognizes foreign and self-antigens, playing an integral part in host defense, as well as the emergence of autoimmunity. Importantly, the interaction between AIRs and their cognate Ags defies a simple key-in-lock paradigm and is instead a complex many-to-many mapping between an individual's massively diverse AIR repertoire, and a similarly diverse antigenic space. Understanding how adaptive immunity balances specificity with epitopic coverage is a key challenge for the field, and terms such as broad specificity, cross-reactivity, and polyreactivity remain ill-defined and are used inconsistently. In this Immunology Notes and Resources article, a group of experimental, structural, and computational immunologists define commonly used terms associated with AIR binding, describe methodologies to study these binding modes, as well as highlight the implications of these different binding modes for therapeutic design.
Collapse
Affiliation(s)
| | | | - Andreas Mayer
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Nicholas C. Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, 0372 Oslo, Norway
| | - Jenna J. Guthmiller
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
65
|
Knezevic L, Wachsmann TLA, Francis O, Dockree T, Bridgeman JS, Wouters A, de Wet B, Cole DK, Clement M, McLaren JE, Gostick E, Ladell K, Llewellyn-Lacey S, Price DA, van den Berg HA, Tabi Z, Sessions RB, Heemskerk MHM, Wooldridge L. High-affinity CD8 variants enhance the sensitivity of pMHCI antigen recognition via low-affinity TCRs. J Biol Chem 2023; 299:104981. [PMID: 37390984 PMCID: PMC10432799 DOI: 10.1016/j.jbc.2023.104981] [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: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 07/02/2023] Open
Abstract
CD8+ T cell-mediated recognition of peptide-major histocompatibility complex class I (pMHCI) molecules involves cooperative binding of the T cell receptor (TCR), which confers antigen specificity, and the CD8 coreceptor, which stabilizes the TCR/pMHCI complex. Earlier work has shown that the sensitivity of antigen recognition can be regulated in vitro by altering the strength of the pMHCI/CD8 interaction. Here, we characterized two CD8 variants with moderately enhanced affinities for pMHCI, aiming to boost antigen sensitivity without inducing non-specific activation. Expression of these CD8 variants in model systems preferentially enhanced pMHCI antigen recognition in the context of low-affinity TCRs. A similar effect was observed using primary CD4+ T cells transduced with cancer-targeting TCRs. The introduction of high-affinity CD8 variants also enhanced the functional sensitivity of primary CD8+ T cells expressing cancer-targeting TCRs, but comparable results were obtained using exogenous wild-type CD8. Specificity was retained in every case, with no evidence of reactivity in the absence of cognate antigen. Collectively, these findings highlight a generically applicable mechanism to enhance the sensitivity of low-affinity pMHCI antigen recognition, which could augment the therapeutic efficacy of clinically relevant TCRs.
Collapse
Affiliation(s)
- Lea Knezevic
- Faculty of Health Sciences, University of Bristol, Bristol, UK; Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Tassilo L A Wachsmann
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ore Francis
- Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Tamsin Dockree
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | | | - Anne Wouters
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - David K Cole
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK; Immunocore, Abingdon, UK
| | - Mathew Clement
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - James E McLaren
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Emma Gostick
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | | | - Zsuzsanna Tabi
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | | | - Mirjam H M Heemskerk
- Department of Haematology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
66
|
Ishii K, Davies JS, Sinkoe AL, Nguyen KA, Norberg SM, McIntosh CP, Kadakia T, Serna C, Rae Z, Kelly MC, Hinrichs CS. Multi-tiered approach to detect autoimmune cross-reactivity of therapeutic T cell receptors. SCIENCE ADVANCES 2023; 9:eadg9845. [PMID: 37494434 PMCID: PMC10371023 DOI: 10.1126/sciadv.adg9845] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
T cell receptor (TCR)-engineered T cell therapy using high-affinity TCRs is a promising treatment modality for cancer. Discovery of high-affinity TCRs especially against self-antigens can require approaches that circumvent central tolerance, which may increase the risk of cross-reactivity. Despite the potential for toxicity, no standardized approach to screen cross-reactivity has been established in the context of preclinical safety evaluation. Here, we describe a practical framework to prospectively detect clinically prohibitive cross-reactivity of therapeutic TCR candidates. Cross-reactivity screening consisted of multifaceted series of assays including assessment of p-MHC tetramer binding, cell line recognition, and reactivity against candidate peptide libraries. Peptide libraries were generated using conventional contact residue motif-guided search, amino acid substitution matrix-based search unguided by motif information, and combinatorial peptide library scan-guided search. We demonstrate the additive nature of a layered approach, which efficiently identifies unsafe cross-reactivity including one undetected by conventional motif-guided search. These findings have important implications for the safe development of TCR-based therapies.
Collapse
Affiliation(s)
- Kazusa Ishii
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - John S. Davies
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Andrew L. Sinkoe
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kilyna A. Nguyen
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Scott M. Norberg
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Crystal P. McIntosh
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tejas Kadakia
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Precigen, Germantown, MD, USA
| | - Carylinda Serna
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Oncology Department, Cell Therapy Unit, AstraZeneca, Gaithersburg, MD, USA
| | - Zachary Rae
- Single Cell Analysis Facility, CCR, NCI, NIH, Bethesda, MD, USA
- 10x Genomics, Pleasanton, CA, USA
| | | | - Christian S. Hinrichs
- Center for Immuno-Oncology, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Duncan and Nancy MacMillan Center of Excellence in Cancer Immunotherapy and Metabolism, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
67
|
Jiang N, Malone M, Chizari S. Antigen-specific and cross-reactive T cells in protection and disease. Immunol Rev 2023; 316:120-135. [PMID: 37209375 PMCID: PMC10524458 DOI: 10.1111/imr.13217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023]
Abstract
Human T cells have a diverse T-cell receptor (TCR) repertoire that endows them with the ability to identify and defend against a broad spectrum of antigens. The universe of possible antigens that T cells may encounter, however, is even larger. To effectively surveil such a vast universe, the T-cell repertoire must adopt a high degree of cross-reactivity. Likewise, antigen-specific and cross-reactive T-cell responses play pivotal roles in both protective and pathological immune responses in numerous diseases. In this review, we explore the implications of these antigen-driven T-cell responses, with a particular focus on CD8+ T cells, using infection, neurodegeneration, and cancer as examples. We also summarize recent technological advances that facilitate high-throughput profiling of antigen-specific and cross-reactive T-cell responses experimentally, as well as computational biology approaches that predict these interactions.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA, 19104
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael Malone
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| | - Shahab Chizari
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
68
|
Gouttefangeas C, Klein R, Maia A. The good and the bad of T cell cross-reactivity: challenges and opportunities for novel therapeutics in autoimmunity and cancer. Front Immunol 2023; 14:1212546. [PMID: 37409132 PMCID: PMC10319254 DOI: 10.3389/fimmu.2023.1212546] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana Maia
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
69
|
Fonseca AF, Antunes DA. CrossDome: an interactive R package to predict cross-reactivity risk using immunopeptidomics databases. Front Immunol 2023; 14:1142573. [PMID: 37377956 PMCID: PMC10291144 DOI: 10.3389/fimmu.2023.1142573] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
T-cell-based immunotherapies hold tremendous potential in the fight against cancer, thanks to their capacity to specifically targeting diseased cells. Nevertheless, this potential has been tempered with safety concerns regarding the possible recognition of unknown off-targets displayed by healthy cells. In a notorious example, engineered T-cells specific to MAGEA3 (EVDPIGHLY) also recognized a TITIN-derived peptide (ESDPIVAQY) expressed by cardiac cells, inducing lethal damage in melanoma patients. Such off-target toxicity has been related to T-cell cross-reactivity induced by molecular mimicry. In this context, there is growing interest in developing the means to avoid off-target toxicity, and to provide safer immunotherapy products. To this end, we present CrossDome, a multi-omics suite to predict the off-target toxicity risk of T-cell-based immunotherapies. Our suite provides two alternative protocols, i) a peptide-centered prediction, or ii) a TCR-centered prediction. As proof-of-principle, we evaluate our approach using 16 well-known cross-reactivity cases involving cancer-associated antigens. With CrossDome, the TITIN-derived peptide was predicted at the 99+ percentile rank among 36,000 scored candidates (p-value < 0.001). In addition, off-targets for all the 16 known cases were predicted within the top ranges of relatedness score on a Monte Carlo simulation with over 5 million putative peptide pairs, allowing us to determine a cut-off p-value for off-target toxicity risk. We also implemented a penalty system based on TCR hotspots, named contact map (CM). This TCR-centered approach improved upon the peptide-centered prediction on the MAGEA3-TITIN screening (e.g., from 27th to 6th, out of 36,000 ranked peptides). Next, we used an extended dataset of experimentally-determined cross-reactive peptides to evaluate alternative CrossDome protocols. The level of enrichment of validated cases among top 50 best-scored peptides was 63% for the peptide-centered protocol, and up to 82% for the TCR-centered protocol. Finally, we performed functional characterization of top ranking candidates, by integrating expression data, HLA binding, and immunogenicity predictions. CrossDome was designed as an R package for easy integration with antigen discovery pipelines, and an interactive web interface for users without coding experience. CrossDome is under active development, and it is available at https://github.com/AntunesLab/crossdome.
Collapse
Affiliation(s)
| | - Dinler A. Antunes
- Antunes Lab, Center for Nuclear Receptors and Cell Signaling (CNRCS), Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| |
Collapse
|
70
|
Ai Q, Li F, Zou S, Zhang Z, Jin Y, Jiang L, Chen H, Deng X, Peng C, Mou N, Wen C, Shen B, Zhan Q. Targeting KRAS G12V mutations with HLA class II-restricted TCR for the immunotherapy in solid tumors. Front Immunol 2023; 14:1161538. [PMID: 37287989 PMCID: PMC10243368 DOI: 10.3389/fimmu.2023.1161538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/08/2023] [Indexed: 06/09/2023] Open
Abstract
KRAS mutation is a significant driving factor of tumor, and KRASG12V mutation has the highest incidence in solid tumors such as pancreatic cancer and colorectal cancer. Thus, KRASG12V neoantigen-specific TCR-engineered T cells could be a promising cancer treatment approach for pancreatic cancer. Previous studies had reported that KRASG12V-reactive TCRs originated from patients' TILs could recognized KRASG12V neoantigen presented by specific HLA subtypes and remove tumor persistently in vitro and in vivo. However, TCR drugs are different from antibody drugs in that they are HLA-restricted. The different ethnic distribution of HLA greatly limits the applicability of TCR drugs in Chinese population. In this study, we have identified a KRASG12V-specific TCR which recognized classII MHC from a colorectal cancer patient. Interestingly, we observed that KRASG12V-specific TCR-engineered CD4+ T cells, not CD8+ T cells, demonstrated significant efficacy in vitro and in xenograft mouse model, exhibiting stable expression and targeting specificity of TCR when co-cultured with APCs presenting KRASG12V peptides. TCR-engineered CD4+ T cells were co-cultured with APCs loaded with neoantigen, and then HLA subtypes were identified by the secretion of IFN-γ. Collectively, our data suggest that TCR-engineered CD4+ T cells can be used to target KRASG12V mutation presented by HLA-DPB1*03:01 and DPB1*14:01, which provide a high population coverage and are more suitable for the clinical transformation for Chinese, and mediate tumor killing effect like CD8+ T cells. This TCR hold promise for precision therapy in immunotherapy of solid tumors as an attractive candidate.
Collapse
Affiliation(s)
- Qi Ai
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fanlu Li
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siyi Zou
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zehui Zhang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yangbing Jin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingxi Jiang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaxing Deng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenghong Peng
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Mou
- Department of Cell Therapy, Shanghai Genbase Biotechnology Co., Ltd, Shanghai, China
| | - Chenlei Wen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baiyong Shen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Zhan
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
71
|
Stedman HH. Confronting complexity in the design and conduct of high-risk rare disease clinical trials. Mol Ther 2023; 31:1191-1192. [PMID: 37141855 PMCID: PMC10329114 DOI: 10.1016/j.ymthe.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Affiliation(s)
- Hansell H Stedman
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
72
|
Krämer S, Moritz A, Stehl L, Hutt M, Hofmann M, Wagner C, Bunk S, Maurer D, Roth G, Wöhrle J. An ultra-high-throughput screen for the evaluation of peptide HLA-Binder interactions. Sci Rep 2023; 13:5290. [PMID: 37002335 PMCID: PMC10066226 DOI: 10.1038/s41598-023-32384-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/27/2023] [Indexed: 04/04/2023] Open
Abstract
Peptide human leukocyte antigen (pHLA) targeting therapeutics like T-cell receptor based adoptive cell therapy or bispecific T cell engaging receptor molecules hold great promise for the treatment of cancer. Comprehensive pre-clinical screening of therapeutic candidates is important to ensure patient safety but is challenging because of the size of the potential off-target space. By combining stabilized peptide-receptive HLA molecules with microarray printing and screening, we have developed an ultra-high-throughput screening platform named ValidaTe that enables large scale evaluation of pHLA-binder interactions. We demonstrate its potential by measuring and analyzing over 30.000 binding curves for a high-affinity T cell Engaging Receptor towards a large pHLA library. Compared to a dataset obtained by conventional bio-layer interferometry measurements, we illustrate that a massively increased throughput (over 650 fold) is obtained by our microarray screening, paving the way for use in pre-clinical safety screening of pHLA-targeting drugs.
Collapse
Affiliation(s)
| | | | - Luca Stehl
- BioCopy GmbH, 79312, Emmendingen, Germany
| | - Meike Hutt
- Immatics Biotechnologies GmbH, 72076, Tübingen, Germany
| | | | | | | | | | - Günter Roth
- BioCopy GmbH, 79312, Emmendingen, Germany
- BioCopy AG, 4123, Basel, Switzerland
| | | |
Collapse
|
73
|
de Rooij MA, Remst DF, van der Steen DM, Wouters AK, Hagedoorn RS, Kester MG, Meeuwsen MH, Wachsmann TL, de Ru AH, van Veelen PA, Verdegaal EM, Falkenburg JF, Heemskerk MH. A library of cancer testis specific T cell receptors for T cell receptor gene therapy. Mol Ther Oncolytics 2023; 28:1-14. [PMID: 36589698 PMCID: PMC9792401 DOI: 10.1016/j.omto.2022.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
To increase the number of cancer patients that can be treated with T cell receptor (TCR) gene therapy, we aimed to identify a set of high-affinity cancer-specific TCRs targeting different melanoma-associated antigens (MAGEs). In this study, peptides derived from MAGE genes with tumor-specific expression pattern were identified by human leukocyte antigen (HLA) peptidomics. Next, peptide-HLA tetramers were generated, and used to sort MAGE-specific CD8+ T cell clones from the allogeneic (allo) HLA repertoire of healthy donors. To evaluate the clinical potential, most potent TCRs were sequenced, transferred into peripheral blood-derived CD8+ T cells, and tested for antitumor efficacy. In total we identified, seven MAGE-specific TCRs that effectively target MAGE-A1, MAGE-A3, MAGE-A6, and MAGE-A9 in the context of HLA-A∗01:01, -A∗02:01, -A∗03:01, -B∗07:02, -B∗35:01, or -C∗07:02. TCR gene transfer into CD8⁺ T cells resulted in efficient reactivity against a variety of different tumor types, while no cross-reactivity was detected. In addition, major in vivo antitumor effects of MAGE-A1 specific TCR engineered CD8⁺ T cells were observed in the orthotopic xenograft model for established multiple myeloma. The identification of seven MAGE-specific TCRs expands the pool of cancer patients eligible for TCR gene therapy and increases possibilities for personalized TCR gene therapy.
Collapse
Affiliation(s)
- Marije A.J. de Rooij
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Dennis F.G. Remst
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Dirk M. van der Steen
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Anne K. Wouters
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Michel G.D. Kester
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Miranda H. Meeuwsen
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Tassilo L.A. Wachsmann
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics and Metabolics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Peter A. van Veelen
- Center for Proteomics and Metabolics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Els M.E. Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - J.H. Frederik Falkenburg
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Mirjam H.M. Heemskerk
- Department of Hematology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
74
|
The Melanoma-Associated Antigen Family A (MAGE-A): A Promising Target for Cancer Immunotherapy? Cancers (Basel) 2023; 15:cancers15061779. [PMID: 36980665 PMCID: PMC10046478 DOI: 10.3390/cancers15061779] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023] Open
Abstract
Early efforts to identify tumor-associated antigens over the last decade have provided unique cancer epitopes for targeted cancer therapy. MAGE-A proteins are a subclass of cancer/testis (CT) antigens that are presented on the cell surface by MHC class I molecules as an immune-privileged site. This is due to their restricted expression to germline cells and a wide range of cancers, where they are associated with resistance to chemotherapy, metastasis, and cancer cells with an increasing potential for survival. This makes them an appealing candidate target for designing an effective and specific immunotherapy, thereby suggesting that targeting oncogenic MAGE-As with cancer vaccination, adoptive T-cell transfer, or a combination of therapies would be promising. In this review, we summarize and discuss previous and ongoing (pre-)clinical studies that target these antigens, while bearing in mind the benefits and drawbacks of various therapeutic strategies, in order to speculate on future directions for MAGE-A-specific immunotherapies.
Collapse
|
75
|
Transgenic HA-1-Specific CD8 + T-Lymphocytes Selectively Target Leukemic Cells. Cancers (Basel) 2023; 15:cancers15051592. [PMID: 36900382 PMCID: PMC10000933 DOI: 10.3390/cancers15051592] [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: 01/31/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
A significant share of allogeneic hematopoietic stem cell transplantations (allo-HSCT) results in the relapse of malignant disease. The T cell immune response to minor histocompatibility antigens (MiHAs) promotes a favorable graft-versus-leukemia response. The immunogenic MiHA HA-1 is a promising target for leukemia immunotherapy, as it is predominantly expressed in hematopoietic tissues and presented by the common HLA A*02:01 allele. Adoptive transfer of HA-1-specific modified CD8+ T cells could complement allo-HSCT from HA-1- donors to HA-1+ recipients. Using bioinformatic analysis and a reporter T cell line, we discovered 13 T cell receptors (TCRs) specific for HA-1. Their affinities were measured by the response of the TCR-transduced reporter cell lines to HA-1+ cells. The studied TCRs showed no cross-reactivity to the panel of donor peripheral mononuclear blood cells with 28 common HLA alleles. CD8+ T cells after endogenous TCR knock out and introduction of transgenic HA-1-specific TCR were able to lyse hematopoietic cells from HA-1+ patients with acute myeloid, T-, and B-cell lymphocytic leukemia (n = 15). No cytotoxic effect was observed on cells from HA-1- or HLA-A*02-negative donors (n = 10). The results support the use of HA-1 as a target for post-transplant T cell therapy.
Collapse
|
76
|
Berman DM, Bell JI. Redirecting Polyclonal T Cells against Cancer with Soluble T-Cell Receptors. Clin Cancer Res 2023; 29:697-704. [PMID: 36255733 PMCID: PMC9932579 DOI: 10.1158/1078-0432.ccr-22-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/30/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
Cancer cells accumulate genetic mutations in coding proteins that may be presented by HLA as neoantigenic peptides (peptide HLA, pHLA). T cells scan for neoantigenic pHLA by the T-cell receptor (TCR):CD3 complex. This complex has the dual function of binding pHLA, by the TCR, and triggering T-cell activation by CD3. Checkpoint therapy activates exhausted T cells to kill cancer cells and generally work best against tumors with high neoantigen burden and in patients with neoantigenic-reactive T cells. TCR T-cell engagers (TCE) are a novel class of immunotherapy that bypasses these two requirements by redirecting polyclonal T cells, regardless of their native specificity, to kill a cancer cell independent of neoantigen burden. This is accomplished through deconstructing the membrane-bound TCR:CD3 complex into a soluble bispecific protein comprised of a targeting domain (TCR) and activating domain (usually anti-CD3 single-chain variable fragment). The pool of targets for TCR TCE is larger than for antibody therapeutics and includes >90% of human intra- or extracellular proteins. Most tumor-associated antigens for solid tumors are intracellular and accessible only by a TCR therapeutic. Tebentafusp, a TCR TCE directed to a peptide derived from the gp100 melanoma protein presented by HLA*A02:01, demonstrated a survival benefit in metastatic uveal melanoma (mUM). This survival benefit highlights the promise of TCR TCEs because mUM is a solid tumor with a very low neoantigen burden and has poor response to checkpoints and chemotherapy. Other TCR TCE programs are now in clinical studies for a broader range of tumors.
Collapse
Affiliation(s)
| | - John I. Bell
- Oxford University, Oxford, United Kingdom.,Corresponding Author: John I. Bell, Office of Regius Professor, Richard Doll Building, Old Road Campus, Oxford, OX3 7DG, United Kingdom. E-mail:
| |
Collapse
|
77
|
Baulu E, Gardet C, Chuvin N, Depil S. TCR-engineered T cell therapy in solid tumors: State of the art and perspectives. SCIENCE ADVANCES 2023; 9:eadf3700. [PMID: 36791198 PMCID: PMC9931212 DOI: 10.1126/sciadv.adf3700] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/06/2023] [Indexed: 05/25/2023]
Abstract
T cell engineering has changed the landscape of cancer immunotherapy. Chimeric antigen receptor T cells have demonstrated a remarkable efficacy in the treatment of B cell malignancies in hematology. However, their clinical impact on solid tumors has been modest so far. T cells expressing an engineered T cell receptor (TCR-T cells) represent a promising therapeutic alternative. The target repertoire is not limited to membrane proteins, and intrinsic features of TCRs such as high antigen sensitivity and near-to-physiological signaling may improve tumor cell detection and killing while improving T cell persistence. In this review, we present the clinical results obtained with TCR-T cells targeting different tumor antigen families. We detail the different methods that have been developed to identify and optimize a TCR candidate. We also discuss the challenges of TCR-T cell therapies, including toxicity assessment and resistance mechanisms. Last, we share some perspectives and highlight future directions in the field.
Collapse
Affiliation(s)
- Estelle Baulu
- Centre de Recherche en Cancérologie de Lyon, Lyon, France
- ErVaccine Technologies, Lyon, France
| | - Célia Gardet
- Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | - Stéphane Depil
- Centre de Recherche en Cancérologie de Lyon, Lyon, France
- ErVaccine Technologies, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
78
|
Mandlik DS, Mandlik SK, Choudhary HB. Immunotherapy for hepatocellular carcinoma: Current status and future perspectives. World J Gastroenterol 2023; 29:1054-1075. [PMID: 36844141 PMCID: PMC9950866 DOI: 10.3748/wjg.v29.i6.1054] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/23/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the world’s deadliest and fastest-growing tumors, with a poor prognosis. HCC develops in the context of chronic liver disease. Curative resection, surgery (liver transplantation), trans-arterial chemoembolization, radioembolization, radiofrequency ablation and chemotherapy are common treatment options for HCC, however, they will only assist a limited percentage of patients. Current treatments for advanced HCC are ineffective and aggravate the underlying liver condition. Despite promising preclinical and early-phase clinical trials for some drugs, existing systemic therapeutic methods for advanced tumor stages remain limited, underlining an unmet clinical need. In current years, cancer immunotherapy has made significant progress, opening up new treatment options for HCC. HCC, on the other hand, has a variety of causes and can affects the body’s immune system via a variety of mechanisms. With the speedy advancement of synthetic biology and genetic engineering, a range of innovative immunotherapies, such as immune checkpoint inhibitors [anti-programmed cell death-1 (PD-1), anti-cytotoxic T lymphocyte antigen-4, and anti-PD ligand 1 cell death antibodies], therapeutic cancer vaccines, engineered cytokines, and adoptive cell therapy have all been used for the treatment of advanced HCC. In this review, we summarize the present clinical and preclinical landscape of immunotherapies in HCC, critically discuss recent clinical trial outcomes, and address future perspectives in the field of liver cancer.
Collapse
Affiliation(s)
- Deepa S Mandlik
- Department of Pharmacology, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| | - Satish K Mandlik
- Department of Pharmaceutics, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| | - Heena B Choudhary
- Department of Pharmacology, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| |
Collapse
|
79
|
Ferrer-Curriu G, Soler-Botija C, Charvatova S, Motais B, Roura S, Galvez-Monton C, Monguió-Tortajada M, Iborra-Egea O, Emdin M, Lupón J, Aimo A, Bagó JR, Bayés-Genís A. Preclinical scenario of targeting myocardial fibrosis with chimeric antigen receptor (CAR) immunotherapy. Biomed Pharmacother 2023; 158:114061. [PMID: 36495661 DOI: 10.1016/j.biopha.2022.114061] [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: 10/10/2022] [Revised: 11/22/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is present in an important proportion of myocardial disorders. Injury activates cardiac fibroblasts, which deposit excess extracellular matrix, increasing tissue stiffness, impairing cardiac function, and leading to heart failure. Clinical therapies that directly target excessive fibrosis are limited, and more effective treatments are needed. Immunotherapy based on chimeric antigen receptor (CAR) T cells is a novel technique that redirects T lymphocytes toward specific antigens to eliminate the target cells. It is currently used in haematological cancers but has demonstrated efficacy in mouse models of hypertensive cardiac fibrosis, with activated fibroblasts as the target cells. CAR T cell therapy is associated with significant toxicities, but CAR natural killer cells can overcome efficacy and safety limitations. The use of CAR immunotherapy offers a potential alternative to current therapies for fibrosis reduction and restoration of cardiac function in patients with myocardial fibrosis.
Collapse
Affiliation(s)
- Gemma Ferrer-Curriu
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Carolina Soler-Botija
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Charvatova
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Benjamin Motais
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Santiago Roura
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), Vic, Barcelona 08500, Spain
| | - Carolina Galvez-Monton
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Monguió-Tortajada
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Oriol Iborra-Egea
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy; Interdisciplinary Center of Health Science, Scuola Superiore Sant'Anna, Pisa, Italy, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Josep Lupón
- Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy; Interdisciplinary Center of Health Science, Scuola Superiore Sant'Anna, Pisa, Italy, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Juli R Bagó
- Faculty of Medicine, University of Ostrava, 703 00 Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, 708 00 Ostrava, Czech Republic; Faculty of Science, University of Ostrava, 701 00 Ostrava, Czech Republic
| | - Antoni Bayés-Genís
- ICREC Research Program, Health Science Research Institute Germans Trias i Pujol (IGTP), Can Ruti Campus, Badalona, Spain; CIBERCV, Instituto de Salud Carlos III, Madrid, Spain; Cardiology Service, Germans Trias i Pujol University Hospital, Badalona, Spain; Department of Medicine, UAB, Barcelona, Spain; Bellvitge Biomedical Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain.
| |
Collapse
|
80
|
Itoh M, Kawagoe S, Nakagawa H, Asahina A, Okano HJ. Generation of induced pluripotent stem cell (iPSC) from NY-ESO-I-specific cytotoxic T cells isolated from the melanoma patient with minor HLAs: The practical pilot study for the adoptive immunotherapy for melanoma using iPSC technology. Exp Dermatol 2023; 32:126-134. [PMID: 36222007 DOI: 10.1111/exd.14686] [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: 08/05/2022] [Revised: 09/23/2022] [Accepted: 10/10/2022] [Indexed: 11/29/2022]
Abstract
Melanoma is one of the most severe skin cancers, derived from melanocytes. Among various therapies for melanoma, adoptive immunotherapy using tumor-infiltrating lymphocytes/chimeric antigen receptor-T cells (TCs) is advanced in recent years; however, the efficacy is still limited, and major challenges remain in terms of safety and cell supply. To solve the issues of adoptive immunotherapy, we utilized induced pluripotent stem cells (iPSCs), which have an unlimited proliferative ability and various differentiation capability. First, we monoclonally isolated CD8+ TCs specifically reactive with NY-ESO-1, one of tumor antigens, from the melanoma patient's monocytes after stimulated with NY-ESO-1 peptide by manual procedure, and cultured NY-ESO-1-specific TCs until proliferated and formed colonies. iPSCs were consequently generated from colony-forming TCs by exogenous expression of reprogramming factors using Sendai virus vector. After the RAG2 gene in TC-derived iPSCs (T-iPSCs) was knocked out for preventing T-cell receptor (TCR) rearrangement, T-iPSCs were re-differentiated into rejuvenated cytotoxic TCs. We confirmed that TCR of T-iPSC-derived TC was maintained as the same of original TCs. In conclusion, T-iPSCs have a potential to be an unlimited cell source for providing cytotoxic TCs. Our study could be a "touchstone" to develop iPSC-based adoptive immunotherapy for the treatment of melanoma for the future clinical use.
Collapse
Affiliation(s)
- Munenari Itoh
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Shiho Kawagoe
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Hidemi Nakagawa
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Akihiko Asahina
- The Jikei University School of Medicine, Department of Dermatology, Tokyo, Japan
| | - Hirotaka James Okano
- The Jikei University School of Medicine, Division of Regenerative Medicine, Tokyo, Japan
| |
Collapse
|
81
|
Martinov T, Greenberg PD. Targeting Driver Oncogenes and Other Public Neoantigens Using T Cell Receptor-Based Cellular Therapy. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:331-351. [PMID: 37655310 PMCID: PMC10470615 DOI: 10.1146/annurev-cancerbio-061521-082114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
T cell reactivity to tumor-specific neoantigens can drive endogenous and therapeutically induced antitumor immunity. However, most tumor-specific neoantigens are unique to each patient (private) and targeting them requires personalized therapy. A smaller subset of neoantigens includes epitopes that span recurrent mutation hotspots, translocations, or gene fusions in oncogenic drivers and tumor suppressors, as well as epitopes that arise from viral oncogenic proteins. Such antigens are likely to be shared across patients (public), uniformly expressed within a tumor, and required for cancer cell survival and fitness. Although a limited number of these public neoantigens are naturally immunogenic, recent studies affirm their clinical utility. In this review, we highlight efforts to target mutant KRAS, mutant p53, and epitopes derived from oncogenic viruses using T cells engineered with off-the-shelf T cell receptors. We also discuss the challenges and strategies to achieving more effective T cell therapies, particularly in the context of solid tumors.
Collapse
Affiliation(s)
- Tijana Martinov
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Philip D Greenberg
- Program in Immunology and Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Immunology Department, University of Washington, Seattle, Washington, USA
| |
Collapse
|
82
|
van Amerongen RA, Morton LT, Chaudhari UG, Remst DF, Hagedoorn RS, van den Berg CW, Freund C, Falkenburg JF, Heemskerk MH. Human iPSC-derived preclinical models to identify toxicity of tumor-specific T cells with clinical potential. Mol Ther Methods Clin Dev 2023; 28:249-261. [PMID: 36816758 PMCID: PMC9931760 DOI: 10.1016/j.omtm.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
The balance between safety and efficacy of T cell therapies remains challenging and T cell mediated toxicities have occurred. The stringent selection of tumor-specific targets and careful selection of tumor-specific T cells using T cell toxicity screenings are essential. In vitro screening options against vital organs or specialized cell subsets would be preferably included in preclinical pipelines, but options remain limited. Here, we set up preclinical models with human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes, epicardial cells, and kidney organoids to investigate toxicity risks of tumor-specific T cells more thoroughly. CD8+T cells reactive against PRAME, HA-1H, CD20, or WT1, currently used or planned to be used in phase I/II clinical studies, were included. Using these hiPSC-derived preclinical models, we demonstrated that WT1-specific T cells caused on-target toxicity that correlated with target gene expression. Multiple measures of T cell reactivity demonstrated this toxicity on the level of T cells and hiPSC-derived target cells. In addition, phenotypic analysis illustrated interaction and crosstalk between infiltrated T cells and kidney organoids. In summary, we demonstrated the benefit of hiPSC-derived models in determining toxicity risks of tumor-specific T cells. Furthermore, our data emphasizes the additional value of other measures of T cell reactivity on top of the commonly used cytokine levels.
Collapse
Affiliation(s)
- Rosa A. van Amerongen
- Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Laura T. Morton
- Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Umesh G. Chaudhari
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Dennis F.G. Remst
- Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology and Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | - Christian Freund
- LUMC hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands
| | | | - Mirjam H.M. Heemskerk
- Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands,Corresponding author: Mirjam H.M. Heemskerk, Department of Hematology, Leiden University Medical Center, 2333ZA Leiden, the Netherlands.
| |
Collapse
|
83
|
Brochier W, Bricard O, Coulie PG. Facts and Hopes in Cancer Antigens Recognized by T Cells. Clin Cancer Res 2023; 29:309-315. [PMID: 36044396 DOI: 10.1158/1078-0432.ccr-21-3798] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/13/2022] [Accepted: 08/15/2022] [Indexed: 01/19/2023]
Abstract
T cells are key effectors of our immune response against tumors and exert their antitumor effects upon recognizing a variety of tumor-specific peptides presented by HLA molecules on the surface of tumor cells. The identification of the tumor-specific antigens of a given tumor is not required for immune checkpoint therapy (ICT), which mainly reactivates existing tumor-specific T cells together with T cells of unknown specificities. To decrease the activation of non-tumor-specific T cells, active or passive immunizations against tumor-specific antigens are considered. These immunizations require the identification of at least some of the tumor-specific antigens displayed on the tumor cells of a patient. While this has become an easy task for tumors with a large number of mutations generating neoantigens, it remains difficult for the remainder. Here, we review some facts about human tumor-specific or tumor-associated antigens, as well as some hopes for their future use in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Orian Bricard
- de Duve Institute, UCLouvain, Brussels, Belgium.,Immunology Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Pierre G Coulie
- de Duve Institute, UCLouvain, Brussels, Belgium.,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| |
Collapse
|
84
|
Qin R, An C, Chen W. Physical-Chemical Regulation of Membrane Receptors Dynamics in Viral Invasion and Immune Defense. J Mol Biol 2023; 435:167800. [PMID: 36007627 PMCID: PMC9394170 DOI: 10.1016/j.jmb.2022.167800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 02/04/2023]
Abstract
Mechanical cues dynamically regulate membrane receptors functions to trigger various physiological and pathological processes from viral invasion to immune defense. These cues mainly include various types of dynamic mechanical forces and the spatial confinement of plasma membrane. However, the molecular mechanisms of how they couple with biochemical cues in regulating membrane receptors functions still remain mysterious. Here, we review recent advances in methodologies of single-molecule biomechanical techniques and in novel biomechanical regulatory mechanisms of critical ligand recognition of viral and immune receptors including SARS-CoV-2 spike protein, T cell receptor (TCR) and other co-stimulatory immune receptors. Furthermore, we provide our perspectives of the general principle of how force-dependent kinetics determine the dynamic functions of membrane receptors and of biomechanical-mechanism-driven SARS-CoV-2 neutralizing antibody design and TCR engineering for T-cell-based therapies.
Collapse
Affiliation(s)
- Rui Qin
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Chenyi An
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Wei Chen
- Department of Cell Biology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China; Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory for Modern Optical Instrumentation Key Laboratory for Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
85
|
Norberg SM, Hinrichs CS. Engineered T cell therapy for viral and non-viral epithelial cancers. Cancer Cell 2023; 41:58-69. [PMID: 36400016 PMCID: PMC9839504 DOI: 10.1016/j.ccell.2022.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/13/2022] [Accepted: 10/17/2022] [Indexed: 11/18/2022]
Abstract
Engineered T cell therapy has shown remarkable efficacy in hematologic malignancies and has the potential for application to common epithelial cancers. Diverse T cell therapy strategies including adoptive transfer of tumor-infiltrating lymphocytes, chimeric antigen receptor (CAR)-T cells, and T cell receptor (TCR)-T cells have been studied in clinical trials. Recent research has established treatment of human papillomavirus (HPV)-associated cancers with TCR-T cells as a model for proof-of-principle studies in epithelial cancers. These studies and others have provided critical insight into mechanisms of tumor regression, therapeutic targets, treatment safety, treatment design, and barriers to curative cell therapies for common types of cancer. This perspective will review and consolidate understanding gained from clinical trials to treat viral and non-viral epithelial cancers with cell and gene therapy and will examine how past experience may guide future strategy in treatment and biomarker discovery.
Collapse
Affiliation(s)
- Scott M Norberg
- National Cancer Institute, Center for Immuno-Oncology, Bethesda, MD 20892, USA
| | - Christian S Hinrichs
- Rutgers Cancer Institute of New Jersey, Duncan and Nancy MacMillan Cancer Immunology and Metabolism Center of Excellence, New Brunswick, NJ 08901, USA.
| |
Collapse
|
86
|
Hiltensperger M, Krackhardt AM. Current and future concepts for the generation and application of genetically engineered CAR-T and TCR-T cells. Front Immunol 2023; 14:1121030. [PMID: 36949949 PMCID: PMC10025359 DOI: 10.3389/fimmu.2023.1121030] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Adoptive cell therapy (ACT) has seen a steep rise of new therapeutic approaches in its immune-oncology pipeline over the last years. This is in great part due to the recent approvals of chimeric antigen receptor (CAR)-T cell therapies and their remarkable efficacy in certain soluble tumors. A big focus of ACT lies on T cells and how to genetically modify them to target and kill tumor cells. Genetically modified T cells that are currently utilized are either equipped with an engineered CAR or a T cell receptor (TCR) for this purpose. Both strategies have their advantages and limitations. While CAR-T cell therapies are already used in the clinic, these therapies face challenges when it comes to the treatment of solid tumors. New designs of next-generation CAR-T cells might be able to overcome these hurdles. Moreover, CARs are restricted to surface antigens. Genetically engineered TCR-T cells targeting intracellular antigens might provide necessary qualities for the treatment of solid tumors. In this review, we will summarize the major advancements of the CAR-T and TCR-T cell technology. Moreover, we will cover ongoing clinical trials, discuss current challenges, and provide an assessment of future directions within the field.
Collapse
Affiliation(s)
- Michael Hiltensperger
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- IIIrd Medical Department, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- *Correspondence: Michael Hiltensperger, ; Angela M. Krackhardt,
| | - Angela M. Krackhardt
- German Cancer Consortium (DKTK), partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- IIIrd Medical Department, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- *Correspondence: Michael Hiltensperger, ; Angela M. Krackhardt,
| |
Collapse
|
87
|
Shin J, Parker MFL, Zhu I, Alanizi A, Rodriguez CI, Liu R, Watchmaker PB, Kalita M, Blecha J, Luu J, Wright B, Lapi SE, Flavell RR, Okada H, Tlsty TD, Roybal KT, Wilson DM. Antigen-Dependent Inducible T-Cell Reporter System for PET Imaging of Breast Cancer and Glioblastoma. J Nucl Med 2023; 64:137-144. [PMID: 35981900 PMCID: PMC9841254 DOI: 10.2967/jnumed.122.264284] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/28/2023] Open
Abstract
For the past several decades, chimeric antigen receptor T-cell therapies have shown promise in the treatment of cancers. These treatments would greatly benefit from companion imaging biomarkers to follow the trafficking of T cells in vivo. Methods: Using synthetic biology, we engineered T cells with a chimeric receptor synthetic intramembrane proteolysis receptor (SNIPR) that induces overexpression of an exogenous reporter gene cassette on recognition of specific tumor markers. We then applied a SNIPR-based PET reporter system to 2 cancer-relevant antigens, human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor variant III (EGFRvIII), commonly expressed in breast and glial tumors, respectively. Results: Antigen-specific reporter induction of the SNIPR PET T cells was confirmed in vitro using green fluorescent protein fluorescence, luciferase luminescence, and the HSV-TK PET reporter with 9-(4-18F-fluoro-3-[hydroxymethyl]butyl)guanine ([18F]FHBG). T cells associated with their target antigens were successfully imaged using PET in dual-xenograft HER2+/HER2- and EGFRvIII+/EGFRvIII- animal models, with more than 10-fold higher [18F]FHBG signals seen in antigen-expressing tumors versus the corresponding controls. Conclusion: The main innovation found in this work was PET detection of T cells via specific antigen-induced signals, in contrast to reporter systems relying on constitutive gene expression.
Collapse
Affiliation(s)
- Jaehoon Shin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Matthew F L Parker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Iowis Zhu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Aryn Alanizi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Carlos I Rodriguez
- Department of Pathology, University of California, San Francisco, San Francisco, California
| | - Raymond Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Payal B Watchmaker
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Mausam Kalita
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Joseph Blecha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Justin Luu
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Brian Wright
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
- Helen Diller Cancer Center, University of California, San Francisco, San Francisco, California
| | - Hideho Okada
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
- Helen Diller Cancer Center, University of California, San Francisco, San Francisco, California
| | - Thea D Tlsty
- Department of Pathology, University of California, San Francisco, San Francisco, California;
| | - Kole T Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California;
- Parker Institute for Cancer Immunotherapy, San Francisco, California
- Helen Diller Cancer Center, University of California, San Francisco, San Francisco, California
- Chan Zuckerberg Biohub, San Francisco, California
- Gladstone UCSF Institute for Genetic Immunology, San Francisco, California; and
- UCSF Cell Design Institute, San Francisco, California
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California;
| |
Collapse
|
88
|
Immisch L, Papafotiou G, Gallarín Delgado N, Scheuplein V, Paschen A, Blankenstein T, Willimsky G. Targeting the recurrent Rac1P29S neoepitope in melanoma with heterologous high-affinity T cell receptors. Front Immunol 2023; 14:1119498. [PMID: 36875127 PMCID: PMC9978334 DOI: 10.3389/fimmu.2023.1119498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Recurrent neoepitopes are cancer-specific antigens common among groups of patients and therefore ideal targets for adoptive T cell therapy. The neoepitope FSGEYIPTV carries the Rac1P29S amino acid change caused by a c.85C>T missense mutation, which is the third most common hotspot mutation in melanoma. Here, we isolated and characterized TCRs to target this HLA-A*02:01-binding neoepitope by adoptive T cell therapy. Peptide immunization elicited immune responses in transgenic mice expressing a diverse human TCR repertoire restricted to HLA-A*02:01, which enabled isolation of high-affinity TCRs. TCR-transduced T cells induced cytotoxicity against Rac1P29S expressing melanoma cells and we observed regression of Rac1P29S expressing tumors in vivo after adoptive T cell therapy (ATT). Here we found that a TCR raised against a heterologous mutation with higher peptide-MHC affinity (Rac2P29L) more efficiently targeted the common melanoma mutation Rac1P29S. Overall, our study provides evidence for the therapeutic potential of Rac1P29S-specific TCR-transduced T cells and reveal a novel strategy by generating more efficient TCRs by heterologous peptides.
Collapse
Affiliation(s)
- Lena Immisch
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, partner site Berlin, Berlin, Germany
| | - George Papafotiou
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, partner site Berlin, Berlin, Germany
| | - Nerea Gallarín Delgado
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vivian Scheuplein
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium, partner site Essen, Essen, Germany
| | - Thomas Blankenstein
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gerald Willimsky
- Institute of Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany.,German Cancer Consortium, partner site Berlin, Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
89
|
Welty NE, Gill SI. Cancer Immunotherapy Beyond Checkpoint Blockade: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:563-578. [PMID: 36636439 PMCID: PMC9830230 DOI: 10.1016/j.jaccao.2022.11.006] [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: 07/19/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 12/24/2022] Open
Abstract
Avoidance of immune destruction is recognized as one of the hallmarks of cancer development. Although first predicted as a potential antitumor treatment modality more than 50 years ago, the widespread clinical use of cancer immunotherapies has only recently become a reality. Cancer immunotherapy works by reactivation of a stalled pre-existing immune response or by eliciting a de novo immune response, and its toolkit comprises antibodies, vaccines, cytokines, and cell-based therapies. The treatment paradigm in some malignancies has completely changed over the past 10 to 15 years. Massive efforts in preclinical development have led to a surge of clinical trials testing innovative therapeutic approaches as monotherapy and, increasingly, in combination. Here we provide an overview of approved and emerging antitumor immune therapies, focusing on the rich landscape of therapeutic approaches beyond those that block the canonical PD-1/PD-L1 and CTLA-4 axes and placing them in the context of the latest understanding of tumor immunology.
Collapse
Key Words
- BiTE, bispecific T cell engager
- CAR, chimeric antigen receptor
- CRS, cytokine-release syndrome
- FDA, U.S. Food and Drug Administration
- HLA, human leukocyte antigen
- ICI, immune checkpoint inhibitor
- IL, interleukin
- NK, natural killer
- NSCLC, non–small cell lung cancer
- TIL, tumor-infiltrating lymphocyte
- alloHCT, allogeneic hematopoietic stem cell transplantation
- cancer
- immune therapy
- immunotherapy
- innovation
- mAb, monoclonal antibody
- treatment
Collapse
Affiliation(s)
- Nathan E. Welty
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saar I. Gill
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA,Address for correspondence: Dr Saar I. Gill, Smilow Center for Translational Research, Room 8-101, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
90
|
Ganatra S, Dani SS, Yang EH, Zaha VG, Nohria A. Cardiotoxicity of T-Cell Antineoplastic Therapies: JACC: CardioOncology Primer. JACC CardioOncol 2022; 4:616-623. [PMID: 36636447 PMCID: PMC9830211 DOI: 10.1016/j.jaccao.2022.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/27/2022] [Indexed: 01/11/2023] Open
Abstract
T-cell therapies, such as chimeric antigen receptor (CAR) T-cell, bispecific T-cell engager (BiTE) and tumor-infiltrating lymphocyte (TIL) therapies, fight cancer cells harboring specific tumor antigens. However, activation of the immune response by these therapies can lead to a systemic inflammatory response, termed cytokine release syndrome (CRS), that can result in adverse events, including cardiotoxicity. Retrospective studies have shown that cardiovascular complications occur in 10% to 20% of patients who develop high-grade CRS after CAR T-cell therapy and can include cardiomyopathy, heart failure, arrhythmias, and myocardial infarction. While cardiotoxicities have been less commonly reported with BiTE and TIL therapies, systematic surveillance for cardiotoxicity has not been performed. Patients undergoing T-cell therapies should be screened for cardiovascular conditions that may not be able to withstand the hemodynamic perturbations imposed by CRS. Generalized management of CRS, including the use of the interleukin-6 antagonist, tocilizumab, for high-grade CRS, is used to mitigate the risk of cardiotoxicity.
Collapse
Key Words
- BCMA, B-cell maturation antigen
- BiTE therapy
- BiTE, bispecific T-cell engager
- CAR T-cell therapy
- CAR, chimeric antigen receptor
- CRS, cytokine release syndrome
- HF, heart failure
- ICSR, individual case safety report
- IL, interleukin
- LVEF, left ventricular ejection fraction
- MACE, major adverse cardiovascular event(s)
- TIL, tumor-infiltrating lymphocyte
- arrhythmia
- cardiomyopathy
- cardiotoxicity
- heart failure
- tumor-infiltrating lymphocytes
Collapse
Affiliation(s)
- Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital & Medical Center, Burlington, Massachusetts, USA
- Address for correspondence: Dr Sarju Ganatra, Department of Cardiovascular Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, Massachusetts 01805, USA. @SarjuGanatraMD
| | - Sourbha S. Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital & Medical Center, Burlington, Massachusetts, USA
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, UCLA-Cardio-Oncology Program, University of California-Los Angeles, Los Angeles, California, USA
| | - Vlad G. Zaha
- Cardio-Oncology Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health & Hospital System, Dallas, Texas, USA
| | - Anju Nohria
- Cardio-Oncology Program, Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
91
|
Shah S, Al-Omari A, Cook KW, Paston SJ, Durrant LG, Brentville VA. What do cancer-specific T cells 'see'? DISCOVERY IMMUNOLOGY 2022; 2:kyac011. [PMID: 38567060 PMCID: PMC10917189 DOI: 10.1093/discim/kyac011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 12/02/2022] [Indexed: 04/04/2024]
Abstract
Complex cellular interactions between the immune system and cancer can impact tumour development, growth, and progression. T cells play a key role in these interactions; however, the challenge for T cells is to recognize tumour antigens whilst minimizing cross-reactivity with antigens associated with healthy tissue. Some tumour cells, including those associated with viral infections, have clear, tumour-specific antigens that can be targeted by T cells. A high mutational burden can lead to increased numbers of mutational neoantigens that allow very specific immune responses to be generated but also allow escape variants to develop. Other cancer indications and those with low mutational burden are less easily distinguished from normal tissue. Recent studies have suggested that cancer-associated alterations in tumour cell biology including changes in post-translational modification (PTM) patterns may also lead to novel antigens that can be directly recognized by T cells. The PTM-derived antigens provide tumour-specific T-cell responses that both escape central tolerance and avoid the necessity for individualized therapies. PTM-specific CD4 T-cell responses have shown tumour therapy in murine models and highlight the importance of CD4 T cells as well as CD8 T cells in reversing the immunosuppressive tumour microenvironment. Understanding which cancer-specific antigens can be recognized by T cells and the way that immune tolerance and the tumour microenvironment shape immune responses to cancer is vital for the future development of cancer therapies.
Collapse
Affiliation(s)
- Sabaria Shah
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Abdullah Al-Omari
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Katherine W Cook
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Samantha J Paston
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| | - Victoria A Brentville
- Scancell Limited, University of Nottingham Biodiscovery Institute, University Park, Nottingham, UK
| |
Collapse
|
92
|
Nanobody-based CAR T cells targeting intracellular tumor antigens. Biomed Pharmacother 2022; 156:113919. [DOI: 10.1016/j.biopha.2022.113919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
|
93
|
Aggarwal P, Luo W, Pehlivan KC, Hoang H, Rajappa P, Cripe TP, Cassady KA, Lee DA, Cairo MS. Pediatric versus adult high grade glioma: Immunotherapeutic and genomic considerations. Front Immunol 2022; 13:1038096. [PMID: 36483545 PMCID: PMC9722734 DOI: 10.3389/fimmu.2022.1038096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022] Open
Abstract
High grade gliomas are identified as malignant central nervous tumors that spread rapidly and have a universally poor prognosis. Historically high grade gliomas in the pediatric population have been treated similarly to adult high grade gliomas. For the first time, the most recent classification of central nervous system tumors by World Health Organization has divided adult from pediatric type diffuse high grade gliomas, underscoring the biologic differences between these tumors in different age groups. The objective of our review is to compare high grade gliomas in the adult versus pediatric patient populations, highlighting similarities and differences in epidemiology, etiology, pathogenesis and therapeutic approaches. High grade gliomas in adults versus children have varying clinical presentations, molecular biology background, and response to chemotherapy, as well as unique molecular targets. However, increasing evidence show that they both respond to recently developed immunotherapies. This review summarizes the distinctions and commonalities between the two in disease pathogenesis and response to therapeutic interventions with a focus on immunotherapy.
Collapse
Affiliation(s)
- Payal Aggarwal
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Wen Luo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | | | - Hai Hoang
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States
| | - Prajwal Rajappa
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Timothy P. Cripe
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kevin A. Cassady
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Dean A. Lee
- Center for Childhood Cancer Research, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, United States,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States,Department of Medicine, New York Medical College, Valhalla, NY, United States,Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, United States,*Correspondence: Mitchell S. Cairo,
| |
Collapse
|
94
|
[Preclinical study of T cell receptor specifically reactive with KRAS G12V mutation in the treatment of malignant tumors]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2022; 54. [PMID: 36241231 PMCID: PMC9568380 DOI: 10.19723/j.issn.1671-167x.2022.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE KRAS gene is one of the most common mutations of proto-oncogenes in human tumors, G12V is one of the most common mutation types for KRAS. It's challenging to chemically acquire the targeted drug for this mutation. Recent studies reported that this mutation peptides can form a neoepitope for T cell recognition. Our study aims to clone the T cell receptor (TCR) which specifically recognizes the neoepitope for KRAS G12V mutation and constructs TCR engineered T cells (TCR-T), and to investigate if TCR-Ts have strong antitumor response ability. METHODS In this study, tumor infiltrating lymphocytes were obtained from one colorectal cancer patient carrying KRAS G12V mutation. Tumor-reactive TCR was obtained by single-cell RT-5' rapid-amplification of cDNA ends PCR analysis and introduced into peripheral blood lymphocytes to generate TCR-Ts. RESULTS We obtained a high-affinity TCR sequence that specifically recognized the HLA-A*11:01-restricted KRAS G12V8-16 epitope: KVA11-01. KVA11-01 TCR-T could significantly kill various tumor cells such as PANC-1, SW480 and HeLa (overexpressing HLA-A*11:01 and KRAS G12V), and secreting high levels of interferon-γ (IFN-γ). Non-specific killing experiments suggested KVA11-01 specifically recognized tumor cells expressing both mutant KRAS G12V and HLA-A*11:01. In vivo assay, tumor inhibition experiments demonstrated that infusion of approximately 1E7 KVA11-01 TCR-T could significantly inhibit the growth of subcuta-neously transplanted tumors of PANC-1 and HeLa (overexpressing HLA-A*11:01 and KRAS G12V) cells in nude mice. No destruction of the morphologies of the liver, spleen and brain were observed. We also found that KVA11-01 TCR-T could significantly infiltrate into tumor tissue and had a better homing ability. CONCLUSION KVA11-01 TCR-T cells can effectively target a variety of malignant tumor cells carrying KRAS G12V mutation through in vitro and in vivo assay. KVA11-01 TCR-T cells have excellent biological activity, high specificity of target antigen and homing ability into solid tumor tissue. KVA11-01 TCR-T is expected to be an effective treatment for patients with KRAS G12V mutant solid malignancies.
Collapse
|
95
|
Vazquez-Lombardi R, Jung JS, Schlatter FS, Mei A, Mantuano NR, Bieberich F, Hong KL, Kucharczyk J, Kapetanovic E, Aznauryan E, Weber CR, Zippelius A, Läubli H, Reddy ST. High-throughput T cell receptor engineering by functional screening identifies candidates with enhanced potency and specificity. Immunity 2022; 55:1953-1966.e10. [PMID: 36174557 DOI: 10.1016/j.immuni.2022.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/19/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022]
Abstract
A major challenge in adoptive T cell immunotherapy is the discovery of natural T cell receptors (TCRs) with high activity and specificity to tumor antigens. Engineering synthetic TCRs for increased tumor antigen recognition is complicated by the risk of introducing cross-reactivity and by the poor correlation that can exist between binding affinity and activity of TCRs in response to antigen (peptide-MHC). Here, we developed TCR-Engine, a method combining genome editing, computational design, and deep sequencing to engineer the functional activity and specificity of TCRs on the surface of a human T cell line at high throughput. We applied TCR-Engine to successfully engineer synthetic TCRs for increased potency and specificity to a clinically relevant tumor-associated antigen (MAGE-A3) and validated their translational potential through multiple in vitro and in vivo assessments of safety and efficacy. Thus, TCR-Engine represents a valuable technology for engineering of safe and potent synthetic TCRs for immunotherapy applications.
Collapse
Affiliation(s)
- Rodrigo Vazquez-Lombardi
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland; Engimmune Therapeutics AG, Hegenheimermattweg 167A, 4123 Allschwil, Switzerland.
| | - Johanna S Jung
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Fabrice S Schlatter
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Anna Mei
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | - Florian Bieberich
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Kai-Lin Hong
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jakub Kucharczyk
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Edo Kapetanovic
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Erik Aznauryan
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Cédric R Weber
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Alfred Zippelius
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Heinz Läubli
- Department of Biomedicine, Universitätsspital Basel, Basel, Switzerland
| | - Sai T Reddy
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland.
| |
Collapse
|
96
|
Britain DM, Town JP, Weiner OD. Progressive enhancement of kinetic proofreading in T cell antigen discrimination from receptor activation to DAG generation. eLife 2022; 11:e75263. [PMID: 36125261 PMCID: PMC9536835 DOI: 10.7554/elife.75263] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
T cells use kinetic proofreading to discriminate antigens by converting small changes in antigen-binding lifetime into large differences in cell activation, but where in the signaling cascade this computation is performed is unknown. Previously, we developed a light-gated immune receptor to probe the role of ligand kinetics in T cell antigen signaling. We found significant kinetic proofreading at the level of the signaling lipid diacylglycerol (DAG) but lacked the ability to determine where the multiple signaling steps required for kinetic discrimination originate in the upstream signaling cascade (Tiseher and Weiner, 2019). Here, we uncover where kinetic proofreading is executed by adapting our optogenetic system for robust activation of early signaling events. We find the strength of kinetic proofreading progressively increases from Zap70 recruitment to LAT clustering to downstream DAG generation. Leveraging the ability of our system to rapidly disengage ligand binding, we also measure slower reset rates for downstream signaling events. These data suggest a distributed kinetic proofreading mechanism, with proofreading steps both at the receptor and at slower resetting downstream signaling complexes that could help balance antigen sensitivity and discrimination.
Collapse
Affiliation(s)
- Derek M Britain
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Jason P Town
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Orion David Weiner
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
97
|
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: 41] [Impact Index Per Article: 13.7] [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
|
98
|
Kim GB, Fritsche J, Bunk S, Mahr A, Unverdorben F, Tosh K, Kong H, Maldini CR, Lau C, Srivatsa S, Jiang S, Glover J, Dopkin D, Zhang CX, Schuster H, Kowalewski DJ, Goldfinger V, Ott M, Fuhrmann D, Baues M, Boesmueller H, Schraeder C, Schimmack G, Song C, Hoffgaard F, Roemer M, Tsou CC, Hofmann M, Treiber T, Hutt M, Alten L, Jaworski M, Alpert A, Missel S, Reinhardt C, Singh H, Schoor O, Walter S, Wagner C, Maurer D, Weinschenk T, Riley JL. Quantitative immunopeptidomics reveals a tumor stroma-specific target for T cell therapy. Sci Transl Med 2022; 14:eabo6135. [PMID: 36044599 PMCID: PMC10130759 DOI: 10.1126/scitranslmed.abo6135] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
T cell receptor (TCR)-based immunotherapy has emerged as a promising therapeutic approach for the treatment of patients with solid cancers. Identifying peptide-human leukocyte antigen (pHLA) complexes highly presented on tumors and rarely expressed on healthy tissue in combination with high-affinity TCRs that when introduced into T cells can redirect T cells to eliminate tumor but not healthy tissue is a key requirement for safe and efficacious TCR-based therapies. To discover promising shared tumor antigens that could be targeted via TCR-based adoptive T cell therapy, we employed population-scale immunopeptidomics using quantitative mass spectrometry across ~1500 tumor and normal tissue samples. We identified an HLA-A*02:01-restricted pan-cancer epitope within the collagen type VI α-3 (COL6A3) gene that is highly presented on tumor stroma across multiple solid cancers due to a tumor-specific alternative splicing event that rarely occurs outside the tumor microenvironment. T cells expressing natural COL6A3-specific TCRs demonstrated only modest activity against cells presenting high copy numbers of COL6A3 pHLAs. One of these TCRs was affinity-enhanced, enabling transduced T cells to specifically eliminate tumors in vivo that expressed similar copy numbers of pHLAs as primary tumor specimens. The enhanced TCR variants exhibited a favorable safety profile with no detectable off-target reactivity, paving the way to initiate clinical trials using COL6A3-specific TCRs to target an array of solid tumors.
Collapse
Affiliation(s)
- Gloria B Kim
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jens Fritsche
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Sebastian Bunk
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Andrea Mahr
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Felix Unverdorben
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Kevin Tosh
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hong Kong
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Colby R Maldini
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chui Lau
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sriram Srivatsa
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuguang Jiang
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua Glover
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Derek Dopkin
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carolyn X Zhang
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Heiko Schuster
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Daniel J Kowalewski
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | | | - Martina Ott
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - David Fuhrmann
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Maike Baues
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Hans Boesmueller
- Institute of Pathology and Neuropathology, Eberhard Karls University, 72076 Tuebingen, Germany
| | - Christoph Schraeder
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Gisela Schimmack
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Colette Song
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Franziska Hoffgaard
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Michael Roemer
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Chih-Chiang Tsou
- Immatics US, 2201 W. Holcombe Blvd., Suite 205, Houston, TX 77030, USA
| | - Martin Hofmann
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Thomas Treiber
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Meike Hutt
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Leonie Alten
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Maike Jaworski
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Amir Alpert
- Immatics US, 2201 W. Holcombe Blvd., Suite 205, Houston, TX 77030, USA
| | - Sarah Missel
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Carsten Reinhardt
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Harpreet Singh
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany.,Immatics US, 2201 W. Holcombe Blvd., Suite 205, Houston, TX 77030, USA
| | - Oliver Schoor
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Steffen Walter
- Immatics US, 2201 W. Holcombe Blvd., Suite 205, Houston, TX 77030, USA
| | - Claudia Wagner
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Dominik Maurer
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany
| | - Toni Weinschenk
- Immatics Biotechnologies GmbH, Paul-Ehrlich-Str. 15, 72076 Tuebingen, Germany.,Immatics US, 2201 W. Holcombe Blvd., Suite 205, Houston, TX 77030, USA
| | - James L Riley
- Department of Microbiology, Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
99
|
Eakins RA, Chobrutskiy A, Teer JK, Patel DN, Hsiang M, Huda TI, Zaman S, Sexton WJ, Coppola D, Falasiri S, Blanck G, Chobrutskiy BI. Chemical complementarity between tumor resident, T-cell receptor CDR3s and MAGEA3/6 correlates with increased melanoma survival: Potential relevance to MAGE vaccine auto-reactivity. Mol Immunol 2022; 150:58-66. [PMID: 35987136 DOI: 10.1016/j.molimm.2022.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/18/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022]
Abstract
Cancer testis antigens have been of interest as possible targets for cancer immunotherapies. To better understand the opportunities for the use of such immunotherapy targets, we used a chemical complementarity scoring algorithm and an original web tool to establish aspects of electrostatic complementarity of the CTAs, MAGEA3 and MAGEA6, with melanoma specimen resident, T-cell receptor (TCR) complementarity determining region 3 (CDR3) amino acid sequences. Greater electrostatic complementarity between T-cell receptor CDR3 and tumor CTAs MAGEA3/6 was associated with a greater probability of overall survival, for both the cancer genome atlas and Moffitt Cancer Center samples; and was associated with high levels of T-cell cytotoxicity-related gene expression. Most importantly, this approach allowed for the highly efficient screening of specific segments of the MAGEA3/6 antigens which indicated that certain MAGE segments would have either more or less risk of auto-reactivity. In sum, the chemical complementarity algorithm, and its efficient application via the web tool, adaptivematch.com, offers a convenient opportunity to identify likely parameters important for immunotherapy considerations and melanoma patient risk stratifications.
Collapse
Affiliation(s)
- Rachel A Eakins
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - Andrea Chobrutskiy
- Department of Pediatrics, Oregon Health and Science University Hospital, Portland, OR 97239, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Dhruv N Patel
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - Monica Hsiang
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - Taha I Huda
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - Saif Zaman
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - Wade J Sexton
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Domenico Coppola
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Shayan Falasiri
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida 33612, USA; Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA.
| | - Boris I Chobrutskiy
- Department of Internal Medicine, Oregon Health and Science University Hospital, Portland, OR 97239, USA
| |
Collapse
|
100
|
Duncan BB, Dunbar CE, Ishii K. Applying a Clinical Lens to Animal Models of CAR-T Cell Therapies. Mol Ther Methods Clin Dev 2022; 27:17-31. [PMID: 36156878 PMCID: PMC9478925 DOI: 10.1016/j.omtm.2022.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Chimeric antigen receptor (CAR)-T cells have emerged as a promising treatment modality for various hematologic and solid malignancies over the past decade. Animal models remain the cornerstone of pre-clinical evaluation of human CAR-T cell products and are generally required by regulatory agencies prior to clinical translation. However, pharmacokinetics and pharmacodynamics of adoptively transferred T cells are dependent on various recipient factors, posing challenges for accurately predicting human engineered T cell behavior in non-human animal models. For example, murine xenograft models did not forecast now well-established cytokine-driven systemic toxicities of CAR-T cells seen in humans, highlighting the limitations of animal models that do not perfectly recapitulate complex human immune systems. Understanding the concordance as well as discrepancies between existing pre-clinical animal data and human clinical experiences, along with established advantages and limitations of each model, will facilitate investigators’ ability to appropriately select and design animal models for optimal evaluation of future CAR-T cell products. We summarize the current state of animal models in this field, and the advantages and disadvantages of each approach depending on the pre-clinical questions being asked.
Collapse
|