1
|
Li W, Mandel U, van Faassen H, Parker MJ, Legg MSG, Hussack G, Clausen H, Evans SV. Structure of the Fab fragment of a humanized 5E5 antibody to a cancer-specific Tn-MUC1 epitope. Acta Crystallogr D Struct Biol 2025; 81:223-233. [PMID: 40221891 PMCID: PMC12054364 DOI: 10.1107/s2059798325002554] [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: 12/06/2024] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
The structure of the humanized Fab from murine monoclonal antibody 5E5 specific for tumor antigen Tn-MUC1 has been determined to 1.57 Å resolution. Despite undertaking thousands of crystallization trials of the humanized 5E5 (h-5E5) Fab in the presence of either the singly or doubly glycosylated peptide antigens corresponding to Tn-MUC1, the Fab is only observed unliganded in the crystal. The conformations of the complementarity-determining regions (CDRs) of the combining site on the h-5E5 Fab do not differ significantly from those reported for liganded murine scFv at 3.0 Å resolution. While the affinity of the murine 5E5 has previously been reported as KD = 1.7 nM for the 24-mer Tn-MUC1 peptide PPAHGVT*SAPDTRPAPGS*T*APPAH prepared by in vitro glycosylation of a synthetic 24-mer MUC1 peptide, the KD of the h-5E5 Fab for the shorter doubly glycosylated glycopeptide antigens PAPGS*T*AP and APGS*T*AP was measured here as only 41 and 61 µM, respectively. Interestingly, the single Fab molecule in the asymmetric unit of space group C2 is observed packed head-to-head with a symmetry-related Fab across a crystallographic twofold axis such that a polypeptide loop from the light chain of each Fab is observed to insert into the antigen-binding pocket of the symmetry-related Fab. While this might suggest that binding of the Tn-MUC1 peptides may have been inhibited by a homophilic association, none was detected. The humanization process has imposed changes in the framework regions of the Fv which may have affected the Vh-Vl interface.
Collapse
MESH Headings
- Immunoglobulin Fab Fragments/chemistry
- Immunoglobulin Fab Fragments/immunology
- Mucin-1/chemistry
- Mucin-1/immunology
- Humans
- Crystallography, X-Ray
- Epitopes/chemistry
- Epitopes/immunology
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/immunology
- Antigens, Tumor-Associated, Carbohydrate/chemistry
- Antigens, Tumor-Associated, Carbohydrate/immunology
- Models, Molecular
- Mice
- Animals
- Glycosylation
Collapse
Affiliation(s)
- Wei Li
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 3055 STN CSCVictoriaBCV8P 3P6Canada
| | - Ulla Mandel
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health SciencesUniversity of CopenhagenBlegdamsvej 32200Copenhagen NDenmark
| | - Henk van Faassen
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1A 0R6Canada
| | - Matthew J. Parker
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 3055 STN CSCVictoriaBCV8P 3P6Canada
| | - Max S. G. Legg
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 3055 STN CSCVictoriaBCV8P 3P6Canada
| | - Greg Hussack
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1A 0R6Canada
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health SciencesUniversity of CopenhagenBlegdamsvej 32200Copenhagen NDenmark
| | - Stephen V. Evans
- Department of Biochemistry and MicrobiologyUniversity of VictoriaPO Box 3055 STN CSCVictoriaBCV8P 3P6Canada
| |
Collapse
|
2
|
Voena C, Ambrogio C, Iannelli F, Chiarle R. ALK in cancer: from function to therapeutic targeting. Nat Rev Cancer 2025; 25:359-378. [PMID: 40055571 DOI: 10.1038/s41568-025-00797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 05/01/2025]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase (RTK) that acts as an oncogenic driver in solid and haematological malignancies in both children and adults. Although ALK-expressing (ALK+) tumours show strong initial responses to the series of ALK inhibitors currently available, many patients will develop resistance. In this Review, we discuss recent advances in ALK oncogenic signalling, together with existing and promising new modalities to treat ALK-driven tumours, including currently approved ALK-directed therapies, namely tyrosine kinase inhibitors, and novel approaches such as ALK-specific immune therapies. Although ALK inhibitors have changed the management and clinical history of ALK+ tumours, they are still insufficient to cure most of the patients. Therefore, more effort is needed to further improve outcomes and prevent the tumour resistance, recurrence and metastatic spread that many patients with ALK+ tumours experience. Here, we outline how a multipronged approach directed against ALK and other essential pathways that sustain the persistence of ALK+ tumours, together with potent or specific immunotherapies, could achieve this goal. We envision that the lessons learned from treating ALK+ tumours in the clinic could ultimately accelerate the implementation of innovative combination therapies to treat tumours driven by other tyrosine kinases or oncogenes with similar properties.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fabio Iannelli
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy.
- Division of Hematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Pathology, Children's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
4
|
Yu R, Ji X, Zhang P, Zhang H, Qu H, Dong W. The potential of chimeric antigen receptor -T cell therapy for endocrine cancer. World J Surg Oncol 2025; 23:153. [PMID: 40264184 PMCID: PMC12012980 DOI: 10.1186/s12957-025-03745-x] [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: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
Endocrine cancer, a relatively rare and heterogeneous tumor with diverse clinical features. The facile synthesis of hormones further complicates endocrine cancer treatment. Thus, the development of safe and effective systemic treatment approaches, such as chimeric antigen receptor (CAR) T cell therapy, is imperative to enhance the prognosis of patients with endocrine cancer. Although this therapy has achieved good results in the treatment of hematological malignancies, it encounters diverse complications and challenges in the context of endocrine cancer. This review delineates the generation of CAR-T cells, examines the potential of CAR-T cell therapy for endocrine cancer, enumerates pivotal antigens linked to endocrine cancer, encapsulates the challenges confronted with CAR-T cell therapy for endocrine cancer, and expounds upon strategies to overcome these limitations. The primary objective is to provide insightful perspectives that can contribute to the advancement of CAR-T cell therapy in the field of endocrine cancer.
Collapse
Affiliation(s)
- Ruonan Yu
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, Liaoning, 110840, China.
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
5
|
Pesce E, Sodini A, Palmieri E, Valensin S, Tinti C, Rossi M, De Rosa A, Fragai M, Papi F, Cordiglieri C, Berti F, Grifantini R, Micoli F, Nativi C. GMMA decorated with mucin 1 Tn/STn mimetics elicit specific antibodies response and inhibit tumor growth. NPJ Vaccines 2025; 10:71. [PMID: 40234452 PMCID: PMC12000591 DOI: 10.1038/s41541-025-01127-8] [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: 09/30/2024] [Accepted: 03/25/2025] [Indexed: 04/17/2025] Open
Abstract
Carbohydrate-based therapeutic vaccines are actively pursued as targeted immunotherapy to treat cancer. Aberrant glycosylation is indeed of paramount importance in tumors, leading to the formation of "neo-epitopes", known as tumor-associated carbohydrate antigens (TACAs), crucial in cancer onset, development and spread. Accordingly, the over-simplified mucin-type O-glycans Tn and STn have been confirmed among the most promising candidates for the development of cancer vaccines. In this work, we first propose genetically manipulated bacteria outer membrane vesicles (OMVs), namely GMMA, as a vaccine formulation platform to display glycan antigens. GMMA were glycosylated with multiple copies of structurally locked Tn mimetic or STn mimetic as cancer vaccine prototypes. These constructs, in non-adjuvanted formulations, showed sounding immunogenic properties in vivo and impressive efficacy in a mouse model of aggressive triple-negative breast cancer. This example of tailor-made therapeutic vaccine might revolutionize the approach to cancer therapy.
Collapse
Affiliation(s)
- Elisa Pesce
- INGM, Istituto Nazionale Genetica Molecolare 'Romeo ed Enrica Invernizzi' and Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | - Andrea Sodini
- Department of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI), Florence, 50019, Italy
| | - Elena Palmieri
- GSK Vaccines Institute for Global Health S.r.l. (GVGH), 53100, Siena, Italy
| | - Silvia Valensin
- Laboratory Animal Research Unit (LARU), Fondazione Life Sciences, via Fiorentina, 1, 53100, Siena, Italy
| | - Cristina Tinti
- Laboratory Animal Research Unit (LARU), Fondazione Life Sciences, via Fiorentina, 1, 53100, Siena, Italy
- Hyper Antibody Research & Development Lab (HARD Lab), Fondazione Toscana Life Sciences, via Fiorentina, 1, 53100, Siena, Italy
| | - Marco Rossi
- Hyper Antibody Research & Development Lab (HARD Lab), Fondazione Toscana Life Sciences, via Fiorentina, 1, 53100, Siena, Italy
| | - Antonella De Rosa
- Laboratory Animal Research Unit (LARU), Fondazione Life Sciences, via Fiorentina, 1, 53100, Siena, Italy
| | - Marco Fragai
- Department of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI), Florence, 50019, Italy
- CeRM/CIRMMP, University of Florence, Via L. Sacconi 6, Sesto Fiorentino (FI), Florence, 50019, Italy
| | - Francesco Papi
- Department of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI), Florence, 50019, Italy
- GSK, 53100, Siena, Italy
| | - Chiara Cordiglieri
- INGM, Istituto Nazionale Genetica Molecolare 'Romeo ed Enrica Invernizzi' and Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy
| | | | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare 'Romeo ed Enrica Invernizzi' and Department of Clinical Sciences and Community Health, University of Milan, 20122, Milan, Italy.
- CheckmAb Srl, 20122, Milan, Italy.
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health S.r.l. (GVGH), 53100, Siena, Italy.
| | - Cristina Nativi
- Department of Chemistry, DICUS, University of Florence, Via della Lastruccia 3,13, Sesto Fiorentino (FI), Florence, 50019, Italy.
| |
Collapse
|
6
|
Ogasawara M, Miyashita M, Yamagishi Y, Ota S. Combined immunotherapy employing Wilms' tumor 1 peptide-pulsed dendritic cells and hormone or chemotherapeutic agents in patients with metastatic castration resistant prostate cancer. Ther Apher Dial 2025. [PMID: 40223232 DOI: 10.1111/1744-9987.70016] [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: 12/26/2024] [Accepted: 03/27/2025] [Indexed: 04/15/2025]
Abstract
INTRODUCTION Metastatic castration-resistant prostate cancer (mCRPC) has a poor prognosis. This study evaluated the safety, immune responses, and clinical outcomes of Wilms' tumor 1 (WT1) peptide-loaded dendritic cell (DC) vaccination combined with hormone or chemotherapeutic agents in mCRPC patients. METHODS WT1 peptide-loaded mature DCs were administered intradermally and the adjuvant OK-432 every 2-4 weeks. WT1-specific immune responses were assayed using ELISpot, HLA-tetramer, and CD107a assays. RESULTS Vaccination was well tolerated with no severe adverse events. WT1-specific immune responses were significantly enhanced in patients with stable disease (SD), along with reduced regulatory T cells. A PSA reduction of >50% was achieved in 35.7% of patients. Median overall survival (mOS) was 28.5 months, exceeding the Halabi nomogram's estimate (19.0 months). Patients with WT1-specific immune responses exhibited significantly longer mOS, suggesting a link between WT1-specific immunity and favorable outcomes. CONCLUSION This immunotherapy approach shows promise for improving survival in mCRPC patients.
Collapse
Affiliation(s)
- Masahiro Ogasawara
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan
- Institute for Artificial Organ, Transplantation and Cell Therapy, Sapporo, Japan
| | - Mamiko Miyashita
- Institute for Artificial Organ, Transplantation and Cell Therapy, Sapporo, Japan
| | - Yuka Yamagishi
- Cell Processing Center, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Shuichi Ota
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan
| |
Collapse
|
7
|
Stadler S, Blasco RB, Singh VK, Damm-Welk C, Ben-Hamza A, Welters C, Hansmann L, Chiarle R, Woessmann W. Endogenous CD4+ T Cells That Recognize ALK and the NPM1::ALK Fusion Protein Can Be Expanded from Human Peripheral Blood. Cancer Immunol Res 2025; 13:487-495. [PMID: 39774774 PMCID: PMC11964841 DOI: 10.1158/2326-6066.cir-24-0445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/08/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
Anaplastic lymphoma kinase (ALK) fusion proteins resulting from chromosomal rearrangements are promising targets for cancer immunotherapy. Although ALK-specific CD8+ T cells and epitopes presented on MHC class I have been identified in patients with ALK-positive malignancies, little is known about ALK-specific CD4+ T cells. We screened peripheral blood of 10 patients with ALK-positive anaplastic large-cell lymphoma in remission and six healthy donors for CD4+ T-cell responses to the whole ALK fusion protein, nucleophosmin 1 (NPM1)::ALK. ALK-specific CD4+ T cells were detected in 15 individuals after stimulation with autologous dendritic cells pulsed with long-overlapping ALK peptide pools. CD4+ T-cell epitopes were predominantly located within three specific regions (p102-188, p257-356, and p593-680) in the ALK portion of the fusion protein. We detected CD4+ T cells in one patient that recognized the NPM1::ALK fusion neoepitope and identified a corresponding T-cell receptor (TCR) by TCRαβ single-cell sequencing. The NPM1::ALK fusion-specific TCR was HLA-DR13-restricted and conferred antigen specificity when expressed in a TCR- reporter cell line (58α-β-). Together, our data provide evidence of ALK-specific CD4+ T cells in human peripheral blood, describe target epitopes in patients, and support the consideration of CD4+ T cells in the development of ALK-specific immunotherapies.
Collapse
Affiliation(s)
- Serena Stadler
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen, Germany
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology, and Tumor Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Rafael B. Blasco
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
| | - Vijay Kumar Singh
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen, Germany
| | - Christine Damm-Welk
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Amin Ben-Hamza
- Department of Hematology, Oncology, and Tumor Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carlotta Welters
- Department of Hematology, Oncology, and Tumor Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Leo Hansmann
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology, and Tumor Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Roberto Chiarle
- Department of Pathology, Boston Children’s Hospital, Boston, MA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Hematopathology division, IRCCS Istituto Europeo di Oncologia, Milan, Italy
| | - Wilhelm Woessmann
- Department of Pediatric Hematology and Oncology, Justus-Liebig University, Giessen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Bai Y, Wang N, Xie S, Zhang J. Paradigm and efficiency of industrial waste resource utilization: Evidence from China. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 380:124922. [PMID: 40090091 DOI: 10.1016/j.jenvman.2025.124922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
Achieving industrial symbiosis and zero emissions requires integrated frameworks for optimizing industrial waste valorization. However, existing studies often overlook the synergy between operational paradigms and multidimensional efficiency. This study bridges this gap through a mixed-methods investigation of 48 Chinese industrial waste resource (IWR) utilization centers, combining qualitative analysis of 1.90 GB of textual case data with a Multi-Criteria Decision Making-Grey Relational Projection Method (MCDM-GRPM) quantitative analysis framework. Two key contributions emerge: (1) redefining industrial waste as IWRs and formulating a "technology-enterprise-policy" paradigm that integrates policy incentives, cross-sector collaboration, and technological innovation to facilitate closed-loop recycling; and (2) developing a multidimensional efficiency evaluation system incorporating technical, economic, and ecological criteria to assess IWR utilization performance. Findings demonstrate that the proposed paradigm operationalizes the 3R principles, transforming waste into high-value resources while fostering industrial symbiosis. Efficiency analysis reveals notable disparities among centers, with top performers (e.g., DMU25, DMU21) leveraging geographic advantages, waste-type characteristics, and industrial diversification. High efficiency in individual dimensions does not ensure overall performance, underscoring the need for balanced, multicriteria-driven strategies. By integrating qualitative and quantitative insights, this study provides a replicable framework for advancing circular economy transitions and promoting industrial symbiosis, aligning economic and environmental objectives.
Collapse
Affiliation(s)
- Yubing Bai
- Beijing Information Science and Technology University, School of Management Science and Engineering, Beijing, 102206, China
| | - Ning Wang
- Beijing Information Science and Technology University, School of Management Science and Engineering, Beijing, 102206, China; Beijing Key Lab of Green Development Decision Making Based on Big Data, Beijing, 102206, China
| | - Shuxin Xie
- State Key Laboratory of Regional Environment and Sustainability, Beijing Normal University, Beijing, 100875, China
| | - Jian Zhang
- Beijing Information Science and Technology University, School of Management Science and Engineering, Beijing, 102206, China; Beijing Knowledge Management Research Center, Beijing, 102206, China.
| |
Collapse
|
9
|
Eskandari A, Leow TC, Rahman MBA, Oslan SN. Advances in Therapeutic Cancer Vaccines, Their Obstacles, and Prospects Toward Tumor Immunotherapy. Mol Biotechnol 2025; 67:1336-1366. [PMID: 38625508 DOI: 10.1007/s12033-024-01144-3] [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: 01/26/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Over the past few decades, cancer immunotherapy has experienced a significant revolution due to the advancements in immune checkpoint inhibitors (ICIs) and adoptive cell therapies (ACTs), along with their regulatory approvals. In recent times, there has been hope in the effectiveness of cancer vaccines for therapy as they have been able to stimulate de novo T-cell reactions against tumor antigens. These tumor antigens include both tumor-associated antigen (TAA) and tumor-specific antigen (TSA). Nevertheless, the constant quest to fully achieve these abilities persists. Therefore, this review offers a broad perspective on the existing status of cancer immunizations. Cancer vaccine design has been revolutionized due to the advancements made in antigen selection, the development of antigen delivery systems, and a deeper understanding of the strategic intricacies involved in effective antigen presentation. In addition, this review addresses the present condition of clinical tests and deliberates on their approaches, with a particular emphasis on the immunogenicity specific to tumors and the evaluation of effectiveness against tumors. Nevertheless, the ongoing clinical endeavors to create cancer vaccines have failed to produce remarkable clinical results as a result of substantial obstacles, such as the suppression of the tumor immune microenvironment, the identification of suitable candidates, the assessment of immune responses, and the acceleration of vaccine production. Hence, there are possibilities for the industry to overcome challenges and enhance patient results in the coming years. This can be achieved by recognizing the intricate nature of clinical issues and continuously working toward surpassing existing limitations.
Collapse
Affiliation(s)
- Azadeh Eskandari
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | | | - Siti Nurbaya Oslan
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
10
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 PMCID: PMC11946770 DOI: 10.3390/vaccines13030285] [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: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut’s Institute of Technology Ladkrabang, Bangkok 10520, Thailand;
| |
Collapse
|
11
|
Yang M, Zhong P, Wei P. Living Bacteria: A New Vehicle for Vaccine Delivery in Cancer Immunotherapy. Int J Mol Sci 2025; 26:2056. [PMID: 40076679 PMCID: PMC11900161 DOI: 10.3390/ijms26052056] [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: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer vaccines, aimed at evolving the human immune system to eliminate tumor cells, have long been explored as a method of cancer treatment with significant clinical potential. Traditional delivery systems face significant challenges in directly targeting tumor cells and delivering adequate amounts of antigen due to the hostile tumor microenvironment. Emerging evidence suggests that certain bacteria naturally home in on tumors and modulate antitumor immunity, making bacterial vectors a promising vehicle for precision cancer vaccines. Live bacterial vehicles offer several advantages, including tumor colonization, precise drug delivery, and immune stimulation, making them a compelling option for cancer immunotherapy. In this review, we explore the mechanisms of action behind living bacteria-based vaccines, recent progress in popular bacterial chassis, and strategies for specific payload delivery and biocontainment to ensure safety. These approaches will lay the foundation for developing an affordable, widely applicable cancer vaccine delivery system. This review also discusses the challenges and future opportunities in harnessing bacterial-based vaccines for enhanced therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
| | | | - Pengcheng Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China; (M.Y.); (P.Z.)
| |
Collapse
|
12
|
Wang W, Bunyatov M, Lopez-Barbosa N, DeLisa MP. Engineering affinity-matured variants of an anti-polysialic acid monoclonal antibody with superior cytotoxicity-mediating potency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637914. [PMID: 40027839 PMCID: PMC11870402 DOI: 10.1101/2025.02.12.637914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Monoclonal antibodies (mAbs) that specifically recognize cell surface glycans associated with cancer and infectious disease hold tremendous value for both basic research and clinical applications. However, high-quality anti-glycan mAbs, especially those with sufficiently high affinity and specificity, remain scarce, highlighting the need for protein engineering approaches based on rational design or directed evolution that enable optimization of antigen-binding properties. To this end, we sought to enhance the affinity of a polysialic acid (polySia)-specific antibody called mAb735, which was raised by animal immunization and possesses only modest affinity, using a combination of rational design and directed evolution. The application of these approaches led to the discovery of affinity-matured IgG variants with up to ∼7-fold stronger affinity for polySia relative to the parental antibody. The higher affinity IgG variants were observed to opsonize polySia- positive cancer cells more avidly, which in turn resulted in significantly greater cytotoxicity as determined by both antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) assays. Collectively, these results demonstrate the effective application of both rational and random molecular evolution techniques to an important anti-glycan antibody, providing insights into its carbohydrate recognition while at the same time uncovering variants with greater therapeutic promise due to their enhanced affinity and potency.
Collapse
Affiliation(s)
- Weiyao Wang
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, NY 14853 USA
| | - Mehman Bunyatov
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, NY 14853 USA
| | - Natalia Lopez-Barbosa
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, NY 14853 USA
| | - Matthew P. DeLisa
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Olin Hall, Ithaca, NY 14853 USA
- Cornell Institute of Biotechnology, Cornell University, 130 Biotechnology Building, Ithaca, NY 14853 USA
| |
Collapse
|
13
|
Zhou Y, Wei Y, Tian X, Wei X. Cancer vaccines: current status and future directions. J Hematol Oncol 2025; 18:18. [PMID: 39962549 PMCID: PMC11834487 DOI: 10.1186/s13045-025-01670-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Cancer continues to be a major global health burden, with high morbidity and mortality. Building on the success of immune checkpoint inhibitors and adoptive cellular therapy, cancer vaccines have garnered significant interest, but their clinical success remains modest. Benefiting from advancements in technology, many meticulously designed cancer vaccines have shown promise, warranting further investigations to reach their full potential. Cancer vaccines hold unique benefits, particularly for patients resistant to other therapies, and they offer the ability to initiate broad and durable T cell responses. In this review, we highlight the antigen selection for cancer vaccines, introduce the immune responses induced by vaccines, and propose strategies to enhance vaccine immunogenicity. Furthermore, we summarize key features and notable clinical advances of various vaccine platforms. Lastly, we delve into the mechanisms of tumor resistance and explore the potential benefits of combining cancer vaccines with standard treatments and other immunomodulatory approaches to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yingqiong Zhou
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
14
|
Velz J, Freudenmann LK, Medici G, Dubbelaar M, Mohme M, Ghasemi DR, Scheid J, Kowalewski DJ, Patterson AB, Zeitlberger AM, Lamszus K, Westphal M, Eyrich M, Messing-Jünger M, Röhrig A, Reinhard H, Beccaria K, Craveiro RB, Frey BM, Sill M, Nahnsen S, Gauder M, Kapolou K, Silginer M, Weiss T, Wirsching HG, Roth P, Grotzer M, Krayenbühl N, Bozinov O, Regli L, Rammensee HG, Rushing EJ, Sahm F, Walz JS, Weller M, Neidert MC. Mapping naturally presented T cell antigens in medulloblastoma based on integrative multi-omics. Nat Commun 2025; 16:1364. [PMID: 39904979 PMCID: PMC11794601 DOI: 10.1038/s41467-025-56268-0] [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: 05/15/2023] [Accepted: 01/14/2025] [Indexed: 02/06/2025] Open
Abstract
Medulloblastoma is the most frequent malignant primary brain tumor in children. Despite recent advances in integrated genomics, the prognosis in children with high-risk medulloblastoma remains devastating, and new tumor-specific therapeutic approaches are needed. Here, we present an atlas of naturally presented T cell antigens in medulloblastoma. We map the human leukocyte antigen (HLA)-presented peptidomes of 28 tumors and perform comparative immunopeptidome profiling against an in-house benign database. Medulloblastoma is shown to be a rich source of tumor-associated antigens, naturally presented on HLA class I and II molecules. Remarkably, most tumor-associated peptides and proteins are subgroup-specific, whereas shared presentation among all subgroups of medulloblastoma (WNT, SHH, Group 3 and Group 4) is rare. Functional testing of top-ranking novel candidate antigens demonstrates the induction of peptide-specific T cell responses, supporting their potential for T cell immunotherapy. This study is an in-depth mapping of naturally presented T cell antigens in medulloblastoma. Integration of immunopeptidomics, transcriptomics, and epigenetic data leads to the identification of a large set of actionable targets that can be further used for the translation into the clinical setting by facilitating the informed design of immunotherapeutic approaches to children with medulloblastoma.
Collapse
Affiliation(s)
- Julia Velz
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Divison of Pediatric Neurosurgery, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lena K Freudenmann
- Institute of Immunology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
| | - Gioele Medici
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marissa Dubbelaar
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David R Ghasemi
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Scheid
- Institute of Immunology, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | | | - Angelica B Patterson
- Institute of Immunobiology, Cantonal Hospital St.Gallen, St. Gallen, Switzerland
- Department of Neurosurgery, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Anna M Zeitlberger
- Department of Neurosurgery, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Eyrich
- Department of Pediatric Haematology, Oncology and Stem Cell Transplantation, University Children's Hospital, University Medical Center, University of Würzburg, Würzburg, Germany
| | | | - Andreas Röhrig
- Department of Neurosurgery, Asklepios Children's Hospital, Sankt Augustin, Germany
| | - Harald Reinhard
- Department of Pediatrics, Asklepios Children's Hospital, Sankt Augustin, Germany
| | - Kévin Beccaria
- Department of Pediatric Neurosurgery, Necker Enfants Malades Hospital, APHP, Université Paris Cite, Paris, France
| | - Rogeiro B Craveiro
- Department of Orthodontic, Dental Clinic, University Hospital of RWTH Aachen, Aachen, Germany
| | - Beat M Frey
- Blood Transfusion Service, Swiss Red Cross, Schlieren, Switzerland
| | - Martin Sill
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
- Department for Computer Science, Biomedical Data Science, University of Tübingen, Tübingen, Germany
- M3 Research Center, University Hospital, Tübingen, Baden- Württemberg, Germany
- Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard-Karls University of Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Marie Gauder
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Konstantina Kapolou
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Manuela Silginer
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Hans-Georg Wirsching
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Patrick Roth
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Michael Grotzer
- Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Niklaus Krayenbühl
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
- Divison of Pediatric Neurosurgery, University Children's Hospital Zurich, Zurich, Switzerland
| | - Oliver Bozinov
- Department of Neurosurgery, Cantonal Hospital St.Gallen, St.Gallen, Switzerland
| | - Luca Regli
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Hans-Georg Rammensee
- Institute of Immunology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Elisabeth J Rushing
- Department of Neuropathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Felix Sahm
- Department of Neuropathology, Heidelberg University Hospital, and CCU Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juliane S Walz
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marian C Neidert
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.
- Department of Neurosurgery, Cantonal Hospital St.Gallen, St.Gallen, Switzerland.
| |
Collapse
|
15
|
Graciotti M, Kandalaft LE. Vaccines for cancer prevention: exploring opportunities and navigating challenges. Nat Rev Drug Discov 2025; 24:134-150. [PMID: 39622986 DOI: 10.1038/s41573-024-01081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 02/06/2025]
Abstract
Improved understanding of cancer immunology has gradually brought increasing attention towards cancer-preventive vaccines as an important tool in the fight against cancer. The aim of this approach is to reduce cancer occurrence by inducing a specific immune response targeting tumours at an early stage before they can fully develop. The great advantage of preventive cancer vaccines lies in the potential to harness a less-compromised immune system in vaccine recipients before their immune responses become affected by the advanced status of the disease itself or by aggressive treatments such as chemotherapy. Successful implementation of immunoprevention against oncogenic viruses such as hepatitis B and papillomavirus has led to a dramatic decrease in virally induced cancers. Extending this approach to other cancers holds great promise but remains a major challenge. Here, we provide a comprehensive review of preclinical evidence supporting this approach, encouraging results from pioneering clinical studies as well as a discussion on the key aspects and open questions to address in order to design potent prophylactic cancer vaccines in the near future.
Collapse
Affiliation(s)
- Michele Graciotti
- Center of Experimental Therapeutics, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Lana E Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
- Department of Oncology, University of Lausanne (UNIL), Lausanne, Switzerland.
- AGORA Cancer Research Center, Lausanne, Lausanne, Switzerland.
- Swiss Medical Network, Genolier Innovation Network, Genolier Clinic, Genolier, Switzerland.
| |
Collapse
|
16
|
Šťastná-Marková M, Hainz P, Kryštofová J, Macková J, Roubalová K, Vydra J, Němečková Š. Absence of pre-transplant T cell response against LAA is associated with Flt3-ITD mutation and increased relapse-risk in AML patients with HSCT. Immunotherapy 2025; 17:185-190. [PMID: 40099461 PMCID: PMC11951724 DOI: 10.1080/1750743x.2025.2478804] [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: 09/10/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
AIMS This study aimed to examine changes in the repertoire of functional T-cells specific for six leukemia-associated antigens (LAA), including WT1, PRAME, MUC1, CCNA1, NPM1, and NPM1c, during immune reconstitution following allogeneic transplantation of hematopoietic stem cells (HSCT) in patients with acute myeloid leukemia. PATIENTS & METHODS LAA-specific T cell response was measured by ELISPOT- IFNγ and intracellular cytokine staining in 47 patients before starting conditioning therapy (baseline) and 7 months after HSCT. RESULTS The positive cumulative LAA-specific T cell response before HSCT was associated with a decreased risk of relapse after HSCT. The prevalent genetic aberration - an internal tandem duplication of Fms 3 - related receptor tyrosine kinase, which has been previously implicated in immune escape mechanisms, is presented here for the first time as a factor associated with the absence of an adaptive T cell response against multiple LAAs. T-cell specific responses against wild-type and mutated NPM1 antigens were less frequent in the study cohort and did not correlate with mutations in the NPM1 gene. CONCLUSIONS Our results showed that the T-cell response to LAA can be reconstituted after HSCT. Measurement of functional pre-transplant T-cell responses against multiple LAAs could help to find patients with an increased risk of relapse.
Collapse
Affiliation(s)
- Markéta Šťastná-Marková
- Transplantation and Intensive Care Unit, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology of the 1st Faculty of Medicine of Charles University, Prague, Czech Republic
| | - Petr Hainz
- Department of Immunology, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jitka Kryštofová
- Department of Immunology, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jana Macková
- Department of Immunology, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Kateřina Roubalová
- Department of Immunology, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jan Vydra
- Transplantation and Intensive Care Unit, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology of the 1st Faculty of Medicine of Charles University, Prague, Czech Republic
| | - Šárka Němečková
- Department of Immunology, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
17
|
Li N, Qin H, Zhu F, Ding H, Chen Y, Lin Y, Deng R, Ma T, Lv Y, Xiong C, Li R, Wei Y, Shi J, Chen H, Zhao Y, Zhou G, Guo H, Lv M, Lin Y, Han B, Nie G, Zhao R. Potent prophylactic cancer vaccines harnessing surface antigens shared by tumour cells and induced pluripotent stem cells. Nat Biomed Eng 2025; 9:215-233. [PMID: 39730914 DOI: 10.1038/s41551-024-01309-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/01/2024] [Indexed: 12/29/2024]
Abstract
The development of prophylactic cancer vaccines typically involves the selection of combinations of tumour-associated antigens, tumour-specific antigens and neoantigens. Here we show that membranes from induced pluripotent stem cells can serve as a tumour-antigen pool, and that a nanoparticle vaccine consisting of self-assembled commercial adjuvants wrapped by such membranes robustly stimulated innate immunity, evaded antigen-specific tolerance and activated B-cell and T-cell responses, which were mediated by epitopes from the abundant number of antigens shared between the membranes of tumour cells and pluripotent stem cells. In mice, the vaccine elicited systemic antitumour memory T-cell and B-cell responses as well as tumour-specific immune responses after a tumour challenge, and inhibited the progression of melanoma, colon cancer, breast cancer and post-operative lung metastases. Harnessing antigens shared by pluripotent stem cell membranes and tumour membranes may facilitate the development of universal cancer vaccines.
Collapse
Affiliation(s)
- Nan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Hao Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yang Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Yixuan Lin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Ronghui Deng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Tianyu Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yuanyuan Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Changhao Xiong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Rong Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Jian Shi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Hanqing Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, P. R. China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, P. R. China
| | - Guangbiao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Hua Guo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Mengyao Lv
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yongfang Lin
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Bing Han
- Department of Orthodontics, Cranial-Facial Growth and Development Center, Peking University School and Hospital of Stomatology, Beijing, P. R. China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, P. R. China.
| |
Collapse
|
18
|
Gielis S, Flumens D, van der Heijden S, Versteven M, De Reu H, Bartholomeus E, Schippers J, Campillo-Davo D, Berneman ZN, Anguille S, Smits E, Ogunjimi B, Lion E, Laukens K, Meysman P. Analysis of Wilms' tumor protein 1 specific TCR repertoire in AML patients uncovers higher diversity in patients in remission than in relapsed. Ann Hematol 2025; 104:317-333. [PMID: 39259326 PMCID: PMC11868354 DOI: 10.1007/s00277-024-05919-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/26/2024] [Indexed: 09/13/2024]
Abstract
The Wilms' tumor protein 1 (WT1) is a well-known and prioritized tumor-associated antigen expressed in numerous solid and blood tumors. Its abundance and immunogenicity have led to the development of different WT1-specific immune therapies. The driving player in these therapies, the WT1-specific T-cell receptor (TCR) repertoire, has received much less attention. Importantly, T cells with high affinity against the WT1 self-antigen are normally eliminated after negative selection in the thymus and are thus rare in peripheral blood. Here, we developed computational models for the robust and fast identification of WT1-specific TCRs from TCR repertoire data. To this end, WT137-45 (WT1-37) and WT1126-134 (WT1-126)-specific T cells were isolated from WT1 peptide-stimulated blood of healthy individuals. The TCR repertoire from these WT1-specific T cells was sequenced and used to train a pattern recognition model for the identification of WT1-specific TCR patterns for the WT1-37 or WT1-126 epitopes. The resulting computational models were applied on an independent published dataset from acute myeloid leukemia (AML) patients, treated with hematopoietic stem cell transplantation, to track WT1-specific TCRs in silico. Several WT1-specific TCRs were found in AML patients. Subsequent clustering analysis of all repertoires indicated the presence of more diverse TCR patterns within the WT1-specific TCR repertoires of AML patients in complete remission in contrast to relapsing patients. We demonstrate the possibility of tracking WT1-37 and WT1-126-specific TCRs directly from TCR repertoire data using computational methods, eliminating the need for additional blood samples and experiments for the two studied WT1 epitopes.
Collapse
MESH Headings
- Humans
- WT1 Proteins/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/blood
- Female
- Remission Induction
- Male
- Middle Aged
- Adult
- Recurrence
- Epitopes, T-Lymphocyte/immunology
- Hematopoietic Stem Cell Transplantation
Collapse
Affiliation(s)
- Sofie Gielis
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| | - Donovan Flumens
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Sanne van der Heijden
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
| | - Maarten Versteven
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Esther Bartholomeus
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Jolien Schippers
- Genetics, Pharmacology and Physiopathology of Heart, Blood Vessels and Skeleton (GENCOR) department, University of Antwerp, Edegem, Belgium
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
- Center for Cell Therapy & Regenerative Medicine (CCRG), Antwerp University Hospital, Edegem, Belgium
| | - Sébastien Anguille
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
- Center for Cell Therapy & Regenerative Medicine (CCRG), Antwerp University Hospital, Edegem, Belgium
- Division of Hematology, Antwerp University Hospital, Edegem, Belgium
| | - Evelien Smits
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Center for Oncological Research (CORE), Integrated Personalized & Precision Oncology Network (IPPON), University of Antwerp, Wilrijk, Belgium
- Center for Cell Therapy & Regenerative Medicine (CCRG), Antwerp University Hospital, Edegem, Belgium
| | - Benson Ogunjimi
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, Edegem, Belgium
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute, University of Antwerp, Wilrijk, Belgium
| | - Eva Lion
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
- Center for Cell Therapy & Regenerative Medicine (CCRG), Antwerp University Hospital, Edegem, Belgium
| | - Kris Laukens
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium
| | - Pieter Meysman
- Adrem Data Lab, Department of Computer Science, University of Antwerp, Antwerp, Belgium.
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium.
- Biomedical Informatics Research Network Antwerp (Biomina), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
19
|
Jamy O, Cicic D. REGAL: galinpepimut-S vs. best available therapy as maintenance therapy for acute myeloid leukemia in second remission. Future Oncol 2025; 21:73-81. [PMID: 39606837 PMCID: PMC11760237 DOI: 10.1080/14796694.2024.2433935] [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: 10/26/2023] [Accepted: 11/21/2024] [Indexed: 11/29/2024] Open
Abstract
Patients with relapsed or refractory (r/r) acute myeloid leukemia (AML) have very poor long-term outcomes. Allogeneic stem cell transplantation (allo-SCT) can potentially cure some of these patients who are able to achieve a second or greater remission with salvage chemotherapy. Unfortunately, several barriers exist to transplantation and not all patients with r/r AML are able to proceed to allo-SCT. Therefore, novel therapies to decrease the risk of relapse in these patients are urgently needed. Wilms tumor 1 (WT1) protein has emerged as an encouraging vaccine target in AML due to its overexpression in leukemic blast cells and near absence in normal hematopoietic cells. Maintenance therapy with galinpepimut-S, a multivalent heteroclitic WT1 peptide vaccine, holds promise in early phase trials, in patients with AML by inducing a strong innate immune response against the WT1 antigen, leading to the design of this international, open-label, randomized clinical trial, named REGAL. Clinical trial registration: https://clinicaltrials.gov/study/NCT04229979. The clinical trial identifier is NCT04229979.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- WT1 Proteins/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Remission Induction
- Randomized Controlled Trials as Topic
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/therapeutic use
- Maintenance Chemotherapy/methods
- Female
- Treatment Outcome
- Male
- Adult
- Middle Aged
- Hematopoietic Stem Cell Transplantation
- Salvage Therapy
Collapse
Affiliation(s)
- Omer Jamy
- Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dragan Cicic
- Department of Clinical Development, SELLAS Life Sciences Group, Inc., New York, NY, USA
| |
Collapse
|
20
|
Ogasawara M. Wilms' tumor 1 -targeting cancer vaccine: Recent advancements and future perspectives. Hum Vaccin Immunother 2024; 20:2296735. [PMID: 38148629 PMCID: PMC10760787 DOI: 10.1080/21645515.2023.2296735] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023] Open
Abstract
This mini-review explores recent advancements in cancer vaccines that target Wilms' tumor (WT1). Phase I/II trials of WT1 peptide vaccines have demonstrated their safety and efficacy against various cancers. Early trials employing HLA class I peptides evolved through their combination with HLA class II peptides, resulting in improved clinical outcomes. Additionally, WT1-targeted dendritic cell vaccines have exhibited favorable results. Studies focusing on hematological malignancies have revealed promising outcomes, including long-term remission and extended survival times. The combination of vaccines with immune checkpoint inhibitors has shown synergistic effects. Current preclinical developments are focused on enhancing the effectiveness of WT1 vaccines, underscoring the necessity for future large-scale Phase III trials to further elucidate their efficacy.
Collapse
Affiliation(s)
- Masahiro Ogasawara
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan
| |
Collapse
|
21
|
Wu E, Ellis A, Bell K, Moss DL, Landry SJ, Hristova K, Wimley WC. pH-Responsive Peptide Nanoparticles Deliver Macromolecules to Cells via Endosomal Membrane Nanoporation. ACS NANO 2024; 18:33922-33936. [PMID: 39651582 DOI: 10.1021/acsnano.4c07525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
The synthetically evolved pHD family of peptides is known to self-assemble into macromolecule-sized nanopores of 2-10 nm diameter in synthetic lipid bilayers, but only when the pH is below ∼6. Here, we show that a representative family member, pHD108, has the same pH-responsive nanopore-forming activity in the endosomal membranes of living human cells, which is triggered by endosomal acidification. This enables the cytosolic delivery of endocytosed proteins and other macromolecules. Acylation of either peptide terminus significantly decreases the concentration of peptide required for macromolecule delivery to the cell cytosol while not causing any measurable cytotoxicity. Longer acyl chains are more effective. The N-terminal palmitoylated C16-pHD108 is the most potent of all of the acyl-pHD108 variants and readily delivers a cytotoxic enzyme, fluorescent proteins, and a dye-labeled dextran to the cell cytosol. C16-pHD108 forms stable monodisperse micellar nanoparticles in a buffer at pH 7 with an average diameter of around 120 nm. These nanoparticles are not cytolytic or cytotoxic because the acylated pHD peptide does not partition from the nanoparticles into cell membranes at pH 7. At pH 5, the nanoparticles are unstable, driving acylated pHD108 to bind strongly to membranes. We hypothesize that passive endocytosis of macromolecular cargo and stable peptide nanoparticles, followed by endosomal acidification-dependent destabilization of the nanoparticles, triggers the nanopore-forming activity of acylated pHD peptides in the endosomal membrane, enabling internalized macromolecules to be delivered to the cytosol.
Collapse
Affiliation(s)
- Eric Wu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Ains Ellis
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Keynon Bell
- Chemistry-Biology Interface Program, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Daniel L Moss
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Samuel J Landry
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Kalina Hristova
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - William C Wimley
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
22
|
Mazziotta F, Martin LE, Eagan DN, Bar M, Kinsella S, Paulson KG, Voillet V, Lahman MC, Hunter D, Schmitt TM, Duerkopp N, Yeung C, Tang TH, Gottardo R, Asano Y, Wilcox EC, Lee B, Zhang T, Lopedote P, Penter L, Wu CJ, Milano F, Greenberg PD, Chapuis AG. Acute Myeloid Leukemia Skews Therapeutic WT1-specific CD8 TCR-T Cells Towards an NK-like Phenotype that Compromises Function and Persistence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.13.24318504. [PMID: 39763516 PMCID: PMC11702715 DOI: 10.1101/2024.12.13.24318504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Acute myeloid leukemia (AML) that is relapsed and/or refractory post-allogeneic hematopoietic cell transplantation (HCT) is usually fatal. In a prior study, we demonstrated that AML relapse in high-risk patients was prevented by post-HCT immunotherapy with Epstein-Barr virus (EBV)-specific donor CD8+ T cells engineered to express a high-affinity Wilms Tumor Antigen 1 (WT1)-specific T-cell receptor (TTCR-C4). However, in the present study, infusion of EBV- or Cytomegalovirus (CMV)-specific TTCR-C4 did not clearly improve outcomes in fifteen patients with active disease post-HCT. TCRC4-transduced EBV-specific T cells persisted longer post-transfer than CMV-specific T cells. Persisting TTCR-C4 skewed towards dysfunctional natural killer-like terminal differentiation, distinct from the dominant exhaustion programs reported for T-cell therapies targeting solid tumors. In one patient with active AML post-HCT, a sustained TTCR-C4 effector-memory profile correlated with long-term TTCR-C4 persistence and disease control. These findings reveal complex mechanisms underlying AML-induced T-cell dysfunction, informing future therapeutic strategies for addressing post-HCT relapse.
Collapse
Affiliation(s)
- Francesco Mazziotta
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Lauren E. Martin
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel N. Eagan
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Merav Bar
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Bristol Myers Squibb
| | - Sinéad Kinsella
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kelly G. Paulson
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Valentin Voillet
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, Cape Town, South Africa
| | - Miranda C. Lahman
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Daniel Hunter
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas M. Schmitt
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Natalie Duerkopp
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cecilia Yeung
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tzu-Hao Tang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Raphael Gottardo
- Biomedical Data Science Center, Lausanne University Hospital
- University of Lausanne, Lausanne, Switzerland
- Agora Translational Research Center, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yuta Asano
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elise C. Wilcox
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Bo Lee
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Tianzi Zhang
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Paolo Lopedote
- Department of Medicine, St. Elizabeth’s Medical Center, Boston University, Boston, MA, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Berlin, Germany
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Filippo Milano
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Philip D. Greenberg
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Departments of Immunology and Medicine, University of Washington, Seattle, WA, USA
| | - Aude G. Chapuis
- Program in Immunology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Translational Sciences and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Immunotherapy Integrated Research Center, Fred Hutch Cancer Center, Seattle, WA, USA
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Du JJ, Zhou SH, Liu J, Zhong XY, Zhang RY, Zhao WX, Wen Y, Su ZH, Lu Z, Guo J. Diphtheria Toxoid-Derived T-Helper Epitope and α-galactosylceramide Synergistically Enhance the Immunogenicity of Glycopeptide Antigen. ACS Pharmacol Transl Sci 2024; 7:3889-3901. [PMID: 39698257 PMCID: PMC11651215 DOI: 10.1021/acsptsci.4c00437] [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: 07/22/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 12/20/2024]
Abstract
The tumor-associated antigen MUC1 is an attractive target for immunotherapy, however, its weak immunogenicity limits the induction of antitumor immune responses. To overcome this limitation, in this study, MUC1 glycopeptide was covalently linked with a diphtheria toxin-derived T-helper epitope (DT331-345). Subsequently, the resulting DT-MUC1 glycopeptide was physically mixed with natural killer T cell agonist αGalCer to explore their immunomodulatory synergy. Biological results demonstrated that compared to MUC1+αGalCer and DT-MUC1 groups, the specific IgG antibody titer of DT-MUC1+αGalCer group increased by 189- and 3-fold, respectively, indicating that the diphtheria toxin-derived T-helper epitope synergistically enhanced MUC1 immunogenicity with αGalCer. Moreover, the DT-MUC1+αGalCer vaccine induced potent cellular immune responses and significantly inhibited the growth of B16-MUC1 tumors in vivo. Furthermore, it was found that the anti-MUC1 IgG antibody titer induced by DT-MUC1+αGalCer was equivalent to that induced by palmitoylated DT-MUC1+αGalCer (P1-DT-MUC1+αGalCer) and significantly higher than that induced by doubly palmitoylated DT-MUC1+αGalCer (P2-DT-MUC1+αGalCer), suggesting that the easily synthesized DT-MUC1 may not require lipid chain modification and already possess good amphiphilicity. This is the first time that a diphtheria toxin-derived helper T-helper epitope was covalently linked to a glycopeptide antigen to enhance its immunogenicity, and this study may provide an effective vaccine design strategy for MUC1-targeted antitumor vaccines and offer novel insights into the design of fully synthetic peptide vaccines.
Collapse
Affiliation(s)
- Jing-Jing Du
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Shi-Hao Zhou
- National
Key Laboratory of Green Pesticide, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Jin Liu
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Xing-Yuan Zhong
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Ru-Yan Zhang
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Wen-Xiang Zhao
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Yu Wen
- National
Key Laboratory of Green Pesticide, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| | - Zhen-Hong Su
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Zheng Lu
- Hubei
Key Laboratory of Kidney Disease Pathogenesis and Intervention, College
of Medicine, Hubei Polytechnic University, Huangshi 435003, China
| | - Jun Guo
- National
Key Laboratory of Green Pesticide, International Joint Research Center
for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
24
|
Nishat S, Hossain MK, Valentin G, Hossain F, Gamage S, Wall KA, Andreana PR. The Glycopeptide PV-PS A1 Immunogen Elicits Both CD4+ and CD8+ Responses. Vaccines (Basel) 2024; 12:1375. [PMID: 39772036 PMCID: PMC11680115 DOI: 10.3390/vaccines12121375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES The MHCII-dependent, CD4+ T-cell zwitterionic polysaccharide PS A1 has been investigated as a promising carrier for vaccine development because it can induce an MHCII-dependent CD4+ response towards a variety of tumor-associated carbohydrate antigens (TACAs). However, PS A1 cannot elicit cytotoxic T lymphocytes through MHCI, which may or may not hamper its potential clinical use in cancer, infectious and viral vaccine development. This paper addresses PS A1 MHCI independence through the introduction of an MHCI epitope, the poliovirus (PV) peptide, to establish an MHCI- and MHCII-dependent vaccine. METHODS We synthesized a glycopeptide construct targeting the Thomsen-nouveau TACA (Tn-PV-PS A1) and a control Tn-PV peptide. C57BL/6 mice were immunized with both constructs, and the resulting T-cells were extracted from spleens. RESULTS Through cell proliferation assays, we show that Tn-PV-PS A1 elicits a robust CD4+ and CD8+ immune response. The resulting cytotoxic T lymphocytes are specific towards Tn-PV and trigger cell lysis of Tn-expressing EL4 cells. CONCLUSIONS This study confirms PV-PS A1 as a robust MHCI- and MHCII-dependent carrier. This is the first report of MHCI dependence in a zwitterionic polysaccharide.
Collapse
Affiliation(s)
- Sharmeen Nishat
- Department of Chemistry, Bangladesh University of Engineering and Technology (BUET), Dhaka 1000, Bangladesh
| | - Md Kamal Hossain
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA (K.A.W.)
| | - Geraud Valentin
- Department of Chemistry and Biochemistry and School of Green Chemistry and Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA
| | - Farzana Hossain
- Department of Chemistry and Biochemistry and School of Green Chemistry and Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA
| | - Shanika Gamage
- Department of Chemistry and Biochemistry and School of Green Chemistry and Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA
| | - Katherine A. Wall
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA (K.A.W.)
| | - Peter R. Andreana
- Department of Chemistry and Biochemistry and School of Green Chemistry and Engineering, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA
| |
Collapse
|
25
|
Chen S, Cheng S, Cai J, Liu Z, Li H, Wang P, Li Y, Yang F, Chen K, Qiu M. The current therapeutic cancer vaccines landscape in non-small cell lung cancer. Int J Cancer 2024; 155:1909-1927. [PMID: 39109825 DOI: 10.1002/ijc.35088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/12/2024] [Accepted: 05/29/2024] [Indexed: 10/04/2024]
Abstract
Currently, conventional immunotherapies for the treatment of non-small cell lung cancer (NSCLC) have low response rates and benefit only a minority of patients, particularly those with advanced disease, so novel therapeutic strategies are urgent deeded. Therapeutic cancer vaccines, a form of active immunotherapy, harness potential to activate the adaptive immune system against tumor cells via antigen cross-presentation. Cancer vaccines can establish enduring immune memory and guard against recurrences. Vaccine-induced tumor cell death prompts antigen epitope spreading, activating functional T cells and thereby sustaining a cancer-immunity cycle. The success of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rendered cancer vaccines a promising avenue, especially when combined with immunotherapy or chemoradiotherapy for NSCLC. This review delves into the intricate antitumor immune mechanisms underlying therapeutic cancer vaccines, enumerates the tumor antigen spectrum of NSCLC, discusses different cancer vaccines progress and summarizes relevant clinical trials. Additionally, we analyze the combination strategies, current limitations, and future prospects of cancer vaccines in NSCLC treatment, aiming to offer fresh insights for their clinical application in managing NSCLC. Overall, cancer vaccines offer promising potential for NSCLC treatment, particularly combining with chemoradiotherapy or immunotherapy could further improve survival in advanced patients. Exploring inhaled vaccines or prophylactic vaccines represents a crucial research avenue.
Collapse
Affiliation(s)
- Shaoyi Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Sida Cheng
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Jingsheng Cai
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Zheng Liu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Haoran Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Peiyu Wang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Yun Li
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Fan Yang
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Kezhong Chen
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| | - Mantang Qiu
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, China
- Thoracic Oncology Institute, Peking University People's Hospital Thoracic Oncology Institute & Research Unit of Intelligence Diagnosis and Treatment in Early Non-small Cell Lung Cancer, Beijing, China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, China
| |
Collapse
|
26
|
Devaraja K, Singh M, Sharan K, Aggarwal S. Coley's Toxin to First Approved Therapeutic Vaccine-A Brief Historical Account in the Progression of Immunobiology-Based Cancer Treatment. Biomedicines 2024; 12:2746. [PMID: 39767654 PMCID: PMC11726767 DOI: 10.3390/biomedicines12122746] [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: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer immunobiology is one of the hot topics of discussion amongst researchers today, and immunotherapeutic modalities are among the selected few emerging approaches to cancer treatment that have exhibited a promising outlook. However, immunotherapy is not a new kid on the block; it has been around for centuries. The origin of cancer immunotherapy in modern medicine can be traced back to the initial reports of spontaneous regression of malignant tumors in some patients following an acute febrile infection, at the turn of the twentieth century. This review briefly revisits the historical accounts of immunotherapy, highlighting some of the significant developments in the field of cancer immunobiology, that have been instrumental in bringing back the immunotherapeutic approaches to the forefront of cancer research. Some of the topics covered are: Coley's toxin-the first immunotherapeutic; the genesis of the theory of immune surveillance; the discovery of T lymphocytes and dendritic cells and their roles; the role of tumor antigens; relevance of tumor microenvironment; the anti-tumor (therapeutic) ability of Bacillus Calmette- Guérin; Melacine-the first therapeutic vaccine engineered; theories of immunoediting and immunophenotyping of cancer; and Provenge-the first FDA-approved therapeutic vaccine. In this review, head and neck cancer has been taken as the reference tumor for narrating the progression of cancer immunobiology, particularly for highlighting the advent of immunotherapeutic agents.
Collapse
Affiliation(s)
- K. Devaraja
- Department of Head and Neck Surgery, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Manisha Singh
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Krishna Sharan
- Department of Radiation Oncology, K S Hegde Medical College, Nitte University, Mangalore 574110, India;
| | - Sadhna Aggarwal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
27
|
Zhang X, You W, Wang Y, Dejenie R, Wang C, Huang Y, Li J. Prospects of anti-GD2 immunotherapy for retinoblastoma. Front Immunol 2024; 15:1499700. [PMID: 39620227 PMCID: PMC11604707 DOI: 10.3389/fimmu.2024.1499700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 10/21/2024] [Indexed: 12/11/2024] Open
Abstract
Retinoblastoma is the most common type of eye tumor in infants and children. Current treatments for retinoblastoma include intravenous chemotherapy, intra-arterial chemotherapy, intravitreal chemotherapy, cryotherapy, radiotherapy, and surgery. However, these treatments come accompanied by adverse effects such as the toxic side effects of chemotherapeutic drugs, post-operative complications including blindness after surgery, or other complications caused by radiotherapy. Immunotherapy is more promising for its low toxicity on normal cells and effectively improves the quality of life of patients. Disialoganglioside (GD2), a sphingolipid expressed on the surface of retinoblastoma, is a potential therapeutic target for retinoblastoma. We summarized immunotherapeutic approaches for both preclinical studies and clinical trials of GD2. An anti-GD2 monoclonal antibody (Dinutuximab), which has been approved for the treatment of high-risk neuroblastomas, has shown promising efficacy in improving patients' prognosis. Additionally, chimeric antigen receptors (CAR)-T therapy, GD2 vaccines and nanoparticles are also potential therapeutics. Finally, we discuss the prospects and current limitations of these immunotherapeutic approaches for treating retinoblastoma, as well as how to address these problems.
Collapse
Affiliation(s)
- Xinlong Zhang
- Affiliated Hospital of Shandong Second Medical University,School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Jinming Yu Academician Workstation of Oncology, Shandong Second Medical University, Shandong, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Wulin You
- Department of Orthopedics, Wuxi Hospital Affiliated of Nanjing University of Chinese Medicine, Wuxi, China
- Medical Center, University of Chicago, Chicago, IL, United States
| | - Yuntao Wang
- Affiliated Hospital of Shandong Second Medical University,School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Jinming Yu Academician Workstation of Oncology, Shandong Second Medical University, Shandong, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Rebeka Dejenie
- Medical Center, University of Chicago, Chicago, IL, United States
- School of Medicine, University of California, Davis, Davis, CA, United States
| | - Chenhao Wang
- Department of Orthopedics, Wuxi Hospital Affiliated of Nanjing University of Chinese Medicine, Wuxi, China
| | - Yan Huang
- Affiliated Hospital of Shandong Second Medical University,School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Jinming Yu Academician Workstation of Oncology, Shandong Second Medical University, Shandong, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Jingjing Li
- Affiliated Hospital of Shandong Second Medical University,School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Jinming Yu Academician Workstation of Oncology, Shandong Second Medical University, Shandong, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Medical Center, University of Chicago, Chicago, IL, United States
| |
Collapse
|
28
|
Morimoto S, Tanaka Y, Nakata J, Fujiki F, Hasegawa K, Nakajima H, Nishida S, Tsuboi A, Hosen N, Kagawa N, Maruno M, Myoui A, Enomoto T, Izumoto S, Sekimoto M, Hashimoto N, Yoshimine T, Kumanogoh A, Oji Y, Oka Y, Sugiyama H. Spontaneous high clonal expansion of Wilms' tumor gene 1-specific cytotoxic T-lymphocytes in patients with Wilms' tumor gene 1-expressing solid tumor. Cancer Immunol Immunother 2024; 74:15. [PMID: 39509060 PMCID: PMC11543974 DOI: 10.1007/s00262-024-03862-8] [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: 05/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
Wilms' tumor protein 1 (WT1)-targeted immunotherapy has been used in patients with leukemia and solid tumors. However, the spontaneous WT1-specific immune response before WT1 peptide vaccination in patients with WT1-expressing tumors (PTs) remains unclear. Therefore, we investigated whether WT1-specific cytotoxic CD8+ T-lymphocytes (CTLs) are clonally expanded in the peripheral blood outside of tumor sites. Clonal expansion of WT1126 peptide (a.a.126-134)-specific CTLs (WT1126-CTLs) was compared between seven PTs and five healthy volunteers (HVs), and their T-cell receptors (TCRs) were analyzed at the single-cell level. Overall, 433 and 351 TCR β-chains of WT1126-CTLs were detected from PTs and HVs, respectively, and complementarity-determining region 3 was sequenced for clonality analysis. The frequencies of WT1126-CTLs were higher in human leukocyte antigen (HLA)-A*02:01+ PTs than in HLA-A*02:01+ HVs, although the difference was not statistically significant. WT1126-CTLs of differentiated types, including memory and effector, were higher in PTs than in HVs; whereas, those of the naïve type were higher in HVs than in PTs. WT1126-CTL clonality was significantly higher in PTs than in HVs. Furthermore, the frequency of effector WT1126-CTLs positively correlated with WT1126-CTL clonality in PTs; whereas, the frequency of naïve phenotype WT1126-CTLs tended to be negatively correlated with clonality. In conclusion, these results suggest that the WT1 protein in tumor cells is highly immunogenic, thereby stimulating endogenous naïve-type WT1126-CTLs and enabling them to clonally expand and differentiate into effector-type WT1126-CTLs.
Collapse
Affiliation(s)
- Soyoko Morimoto
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Yukie Tanaka
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Molecular Microbiology and Immunology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Jun Nakata
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kana Hasegawa
- Laboratory of Cellular Immunotherapy, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sumiyuki Nishida
- Strategic Global Partnership & X (Cross)-Innovation Initiative Graduate School of Medicine, Osaka University and Osaka University Hospital, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Osaka, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naoki Hosen
- Laboratory of Cellular Immunotherapy, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Naoki Kagawa
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motohiko Maruno
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Neurosurgery, Yukioka Hospital, Osaka, Japan
| | - Akira Myoui
- Medical Center for Translational Research, Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
- Center for Genetic Medicine, Itami City Hospital, Hyogo, Japan
| | - Shuichi Izumoto
- Department of Neurosurgery, Hyogo College of Medicine, Hyogo, Japan
- Department of Neurosurgery, Kindai University Nara Hospital, Nara, Japan
| | - Mitsugu Sekimoto
- Department of Gastroenterological Surgery Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Surgery, Minoh City Hospital, Osaka, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Neurosurgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiki Yoshimine
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Endowed Research Department of Clinical Neuroengineering, Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
- Iseikai Medical Corporation, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Oji
- Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
29
|
Yagawa Y, Kobayashi Y, Fujita I, Watanabe M, Koido S, Sugiyama H, Tanigawa K. Peritoneal Dissemination and Malignant Ascites in Duodenal Cancer Successfully Treated With Adoptive Cell Therapy Using WT1- and MUC1-Pulsed Dendritic Cells and Activated T Cells With No Adverse Effects: A Case Report. Cureus 2024; 16:e74834. [PMID: 39737308 PMCID: PMC11684412 DOI: 10.7759/cureus.74834] [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] [Accepted: 11/30/2024] [Indexed: 01/01/2025] Open
Abstract
A satisfactory treatment for the dissemination of duodenal cancer has not yet been established. We describe a case of peritoneal dissemination and malignant ascites in duodenal cancer that was successfully treated with adoptive cell therapy with no adverse effects. A 72-year-old Japanese male patient with primary duodenal cancer with distal lymph node metastases received chemotherapy with S-1, an oral pyrimidine fluoride-derived agent, and oxaliplatin after gastrojejunal bypass, which resulted in tumor shrinkage; however, peritoneal dissemination developed. Despite the administration of a second-line chemotherapy regimen comprising irinotecan, peritoneal dissemination, malignant ascites, and cachexia continued to progress, ultimately resulting in the failure of chemotherapy. He then received adoptive cell therapy with Wilms' tumor 1 (WT1)- and mucin 1 (MUC1) peptide-pulsed dendritic cells (WT1/MUC1-DC) and CD3-activated T lymphocytes (CAT). Following the administration of this treatment eight times per week, the patient's symptoms and malignant ascites surrounding his cancer disappeared. He developed no adverse effects from this treatment and was able to resume his usual activities without any symptoms. He has continued this treatment every few months as maintenance therapy and has been free of relapse for 54 months. This case suggests a possible beneficial effect of adoptive cell therapy with WT1/MUC1-DC and CAT for peritoneal dissemination and malignant ascites in duodenal cancer.
Collapse
Affiliation(s)
- Yohsuke Yagawa
- Department of Immunotherapy, Bio-Thera Clinic, Tokyo, JPN
| | | | - Izumi Fujita
- Department of Surgery, Ebara Hospital, Tokyo, JPN
| | - Manabu Watanabe
- Department of Surgery, Toho University Ohashi Medical Center, Tokyo, JPN
| | - Shigeo Koido
- Internal Medicine, The Jikei University School of Medicine, Tokyo, JPN
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medcine, Osaka, JPN
| | | |
Collapse
|
30
|
Frederico SC, Raphael I, Nisnboym M, Huq S, Schlegel BT, Sneiderman CT, Jackson SA, Jain A, Olin MR, Rood BR, Pollack IF, Hwang EI, Rajasundaram D, Kohanbash G. Transcriptomic observations of intra and extracellular immunotherapy targets for pediatric brain tumors. Expert Rev Clin Immunol 2024; 20:1411-1420. [PMID: 39114885 DOI: 10.1080/1744666x.2024.2390023] [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: 05/14/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
OBJECTIVES Despite surgical resection, chemoradiation, and targeted therapy, brain tumors remain a leading cause of cancer-related death in children. Immunotherapy has shown some promise and is actively being investigated for treating childhood brain tumors. However, a critical step in advancing immunotherapy for these patients is to uncover targets that can be effectively translated into therapeutic interventions. METHODS In this study, our team performed a transcriptomic analysis across pediatric brain tumor types to identify potential targets for immunotherapy. Additionally, we assessed components that may impact patient response to immunotherapy, including the expression of genes essential for antigen processing and presentation, inhibitory ligands and receptors, interferon signature, and overall predicted T cell infiltration. RESULTS We observed distinct expression patterns across tumor types. These included elevated expression of antigen genes and antigen processing machinery in some tumor types while other tumors had elevated inhibitory checkpoint receptors, known to be associated with response to checkpoint inhibitor immunotherapy. CONCLUSION These findings suggest that pediatric brain tumors exhibit distinct potential for specific immunotherapies. We believe our findings can guide investigators in their assessment of appropriate immunotherapy classes and targets in pediatric brain tumors.
Collapse
Affiliation(s)
- Stephen C Frederico
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michal Nisnboym
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
- Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Sakibul Huq
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brent T Schlegel
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sydney A Jackson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anya Jain
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael R Olin
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Brian R Rood
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | - Ian F Pollack
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugene I Hwang
- Division of Oncology, Children's National Medical Center, Washington, DC, USA
| | | | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Han H, Santos HA. Nano- and Micro-Platforms in Therapeutic Proteins Delivery for Cancer Therapy: Materials and Strategies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409522. [PMID: 39263818 DOI: 10.1002/adma.202409522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Proteins have emerged as promising therapeutics in oncology due to their great specificity. Many treatment strategies are developed based on protein biologics, such as immunotherapy, starvation therapy, and pro-apoptosis therapy, while some protein biologics have entered the clinics. However, clinical translation is severely impeded by instability, short circulation time, poor transmembrane transportation, and immunogenicity. Micro- and nano-particles-based drug delivery platforms are designed to solve those problems and enhance protein therapeutic efficacy. This review first summarizes the different types of therapeutic proteins in clinical and research stages, highlighting their administration limitations. Next, various types of micro- and nano-particles are described to demonstrate how they can overcome those limitations. The potential of micro- and nano-particles are then explored to enhance the therapeutic efficacy of proteins by combinational therapies. Finally, the challenges and future directions of protein biologics carriers are discussed for optimized protein delivery.
Collapse
Affiliation(s)
- Huijie Han
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| |
Collapse
|
32
|
Nielsen AJ, Albert GK, Sanchez A, Chen J, Liu J, Davalos AS, Geng D, Bradeen X, Hintzsche JD, Robinson W, McCarter M, Amato C, Tobin R, Couts K, Wilky BA, Davila E. DNA-PK inhibition enhances neoantigen diversity and increases T cell responses to immunoresistant tumors. J Clin Invest 2024; 134:e180278. [PMID: 39436696 PMCID: PMC11645140 DOI: 10.1172/jci180278] [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: 02/13/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Effective antitumor T cell activity relies on the expression and MHC presentation of tumor neoantigens. Tumor cells can evade T cell detection by silencing the transcription of antigens or by altering MHC machinery, resulting in inadequate neoantigen-specific T cell activation. We identified the DNA-protein kinase inhibitor (DNA-PKi) NU7441 as a promising immunomodulator that reduced immunosuppressive proteins, while increasing MHC-I expression in a panel of human melanoma cell lines. In tumor-bearing mice, combination therapy using NU7441 and the immune adjuvants stimulator of IFN genes (STING) ligand and the CD40 agonist NU-SL40 substantially increased and diversified the neoantigen landscape, antigen-presenting machinery, and, consequently, substantially increased both the number and repertoire of neoantigen-reactive, tumor-infiltrating lymphocytes (TILs). DNA-PK inhibition or KO promoted transcription and protein expression of various neoantigens in human and mouse melanomas and induced sensitivity to immune checkpoint blockade (ICB) in resistant tumors. In patients, protein kinase, DNA-activated catalytic subunit (PRKDC) transcript levels were inversely correlated with MHC-I expression and CD8+ TILs but positively correlated with increased neoantigen loads and improved responses to ICB. These studies suggest that inhibition of DNA-PK activity can restore tumor immunogenicity by increasing neoantigen expression and presentation and broadening the neoantigen-reactive T cell population.
Collapse
Affiliation(s)
- Allison J. Nielsen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Gabriella K. Albert
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Amelia Sanchez
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jiangli Chen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Jing Liu
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Andres S. Davalos
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Degui Geng
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
| | - Xander Bradeen
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - William Robinson
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Martin McCarter
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Carol Amato
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Richard Tobin
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kasey Couts
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Breelyn A. Wilky
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| | - Eduardo Davila
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs, Research Service, Rocky Mountain Regional Veterans Affairs, Aurora, Colorado, USA
- University of Colorado Comprehensive Cancer Center and
| |
Collapse
|
33
|
Hao J, Dong S, Li J. Estimating weight for multidimensional health poverty using Delphi method and analytic hierarchy process: a case of China. BMC Public Health 2024; 24:2908. [PMID: 39434048 PMCID: PMC11494899 DOI: 10.1186/s12889-024-20406-y] [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: 12/28/2023] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Traditionally, poverty assessment has relied on a single income criterion, which is inadequate in contexts where absolute poverty has been mitigated. This study developed a weighting scheme for the Multidimensional Health Poverty Index (MHPI) based on sustainable livelihood framework (SLF), aimed at enhancing the accuracy of poverty identification in China. METHODS A two-round Delphi process was used to determine the indicators of the index system. The weight of each indicator was calculated using the analytic hierarchy process (AHP) method. The multidimensional poverty index incorporates SLF's five dimensions: financial, natural, physical, social, and human capital. RESULTS The initial 46 indicators formed an indicator pool for the Delphi questionnaire. Based on the final consensus of the expert panel, the Delphi consultation resulted in an index system comprising six first-level and 23 second-level indicators. The weight values of the first-level indicators (economic security, health status, education, social capital, healthcare utilisation, and living conditions) were 0.2715, 0.2593, 0.0855, 0.0657, 0.1812, and 0.1363, respectively. CONCLUSION This study established a scientific and effective index to evaluate generate weight for estimating multidimensional health poverty in China. Economic security, health status, and healthcare utilisation are the most crucial aspects of multidimensional health poverty. Moreover, the results indicated that vocational training and social capital should be emphasised.
Collapse
Affiliation(s)
- Jin Hao
- School of Public Health, Centre for Health Management and Policy Research, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Shiju Dong
- School of Public Health, Centre for Health Management and Policy Research, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China
| | - Jiajia Li
- School of Public Health, Centre for Health Management and Policy Research, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- NHC Key Lab of Health Economics and Policy Research, Shandong University, Jinan, 250012, China.
| |
Collapse
|
34
|
Rosenblum LT, Sever RE, Gilbert R, Guerrero D, Vincze SR, Menendez DM, Birikorang PA, Rodgers MR, Jaswal AP, Vanover AC, Latoche JD, Cortez AG, Day KE, Foley LM, Sneiderman CT, Raphael I, Hitchens TK, Nedrow JR, Kohanbash G, Edwards WB, Malek MM. Dual-labeled anti-GD2 targeted probe for intraoperative molecular imaging of neuroblastoma. J Transl Med 2024; 22:940. [PMID: 39407274 PMCID: PMC11476241 DOI: 10.1186/s12967-024-05728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Surgical resection is integral for the treatment of neuroblastoma, the most common extracranial solid malignancy in children. Safely locating and resecting primary tumor and remote deposits of disease remains a significant challenge, resulting in high rates of complications and incomplete surgery, worsening outcomes. Intraoperative molecular imaging (IMI) uses targeted radioactive or fluorescent tracers to identify and visualize tumors intraoperatively. GD2 was selected as an IMI target, as it is highly overexpressed in neuroblastoma and minimally expressed in normal tissue. METHODS GD2 expression in neuroblastoma cell lines was measured by flow cytometry. DTPA and IRDye® 800CW were conjugated to anti-GD2 antibody to generate DTPA-αGD2-IR800. Binding affinity (Kd) of the antibody and the non-radiolabeled tracer were then measured by ELISA assay. Human neuroblastoma SK-N-BE(2) cells were surgically injected into the left adrenal gland of 3.5-5-week-old nude mice and the orthotopic xenograft tumors grew for 5 weeks. 111In-αGD2-IR800 or isotype control tracer was administered via tail vein injection. After 4 and 6 days, mice were euthanized and gamma and fluorescence biodistributions were measured using a gamma counter and ImageJ analysis of acquired SPY-PHI fluorescence images of resected organs (including tumor, contralateral adrenal, kidneys, liver, muscle, blood, and others). Organ uptake was compared by one-way ANOVA (with a separate analysis for each tracer/day combination), and if significant, Sidak's multiple comparison test was used to compare the uptake of each organ to the tumor. Handheld tools were also used to detect and visualize tumor in situ, and to assess for residual disease following non-guided resection. RESULTS 111In-αGD2-IR800 was successfully synthesized with 0.75-2.0 DTPA and 2-3 IRDye® 800CW per antibody and retained adequate antigen-binding (Kd = 2.39 nM for aGD2 vs. 21.31 nM for DTPA-aGD2-IR800). The anti-GD2 tracer demonstrated antigen-specific uptake in mice with human neuroblastoma xenografts (gamma biodistribution tumor-to-blood ratios of 3.87 and 3.88 on days 4 and 6 with anti-GD2 tracer), while isotype control tracer did not accumulate (0.414 and 0.514 on days 4 and 6). Probe accumulation in xenografts was detected and visualized using widely available operative tools (Neoprobe® and SPY-PHI camera) and facilitated detection ofputative residual disease in the resection cavity following unguided resection. CONCLUSIONS We have developed a dual-labeled anti-GD2 antibody-based tracer that incorporates In-111 and IRDye® 800CW for radio- and fluorescence-guided surgery, respectively. The tracer adequately binds to GD2, specifically accumulates in GD2-expressing xenograft tumors, and enables tumor visualization with a hand-held NIR camera. These results encourage the development of 111In-αGD2-IR800 for future use in children with neuroblastoma, with the goal of improving patient safety, completeness of resection, and overall patient outcomes.
Collapse
Affiliation(s)
- Lauren Taylor Rosenblum
- Department of General Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - ReidAnn E Sever
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA
| | - Ryan Gilbert
- University of Pittsburgh School of Medicine, 3550 Terrace Street, Pittsburgh, PA, 15213, USA
| | - David Guerrero
- University of Pittsburgh School of Medicine, 3550 Terrace Street, Pittsburgh, PA, 15213, USA
| | - Sarah R Vincze
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA
| | - Dominic M Menendez
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Peggy A Birikorang
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Mikayla R Rodgers
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Ambika Parmar Jaswal
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA
| | - Alexander C Vanover
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Joseph D Latoche
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Angel G Cortez
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Kathryn E Day
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Lesley M Foley
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Chaim T Sneiderman
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA
| | - Itay Raphael
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA
| | - T Kevin Hitchens
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Jessie R Nedrow
- In Vivo Imaging Facility Core, Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Avenue, Pittsburgh, PA, 15232, USA
| | - Gary Kohanbash
- Department of Neurological Surgery, Children's Hospital of Pittsburgh of UPMC, 7131 Rangos Research Building, 530 45th Street, Pittsburgh, PA, 15201, USA.
- Department of Immunology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
| | - W Barry Edwards
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| | - Marcus M Malek
- Department of General Surgery, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.
- Department of Pediatric General Surgery, University of Pittsburgh Medical Center, One Children's Hospital Drive, 4401 Penn Ave., Faculty Pavilion 7th Floor, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
35
|
Sennikov S, Volynets M, Alrhmoun S, Perik-Zavodskii R, Perik-Zavodskaia O, Fisher M, Lopatnikova J, Shevchenko J, Nazarov K, Philippova J, Alsalloum A, Kurilin V, Silkov A. Modified Dendritic cell-based T-cell expansion protocol and single-cell multi-omics allow for the selection of the most expanded and in vitro-effective clonotype via profiling of thousands of MAGE-A3-specific T-cells. Front Immunol 2024; 15:1470130. [PMID: 39450161 PMCID: PMC11499154 DOI: 10.3389/fimmu.2024.1470130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Adoptive cell therapy using TCR-engineered T-cells is one of the most effective strategies against tumor cells. The TCR T-cell approach has been well tested against a variety of blood neoplasms but is yet to be deeply tested against solid tumors. Among solid tumors, cancer-testis antigens are the most prominent targets for tumor-specific therapy, as they are usually found on cells that lie behind blood-tissue barriers. Methods We have employed a novel efficient protocol for MAGE-A3-specific T-cell clonal expansion, performed single-cell multi-omic analysis of the expanded T-cells via BD Rhapsody, engineered a selected T-cell receptor into a lentiviral construct, and tested it in an in vitro LDH-cytotoxicity test. Results and discussion We have observed a 191-fold increase in the MAGE-A3-specific T-cell abundance, obtained a dominant T-cell receptor via single-cell multi-omic BD Rhapsody data analysis in the TCRscape bioinformatics tool, and observed potent cytotoxicity of the dominant-clonotype transduced TCR T-cells against a MAGE-A3-positive tumor. We have demonstrated the efficiency of our T-cell enrichment protocol in obtaining potent anti-tumor T-cells and their T-cell receptors, especially when paired with the modern single-cell analysis methods.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Humans
- Neoplasm Proteins/immunology
- Neoplasm Proteins/genetics
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Immunotherapy, Adoptive/methods
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Single-Cell Analysis/methods
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Cell Line, Tumor
- Clone Cells
- Cell Proliferation
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Cytotoxicity, Immunologic
- Multiomics
Collapse
Affiliation(s)
- Sergey Sennikov
- Laboratory of Molecular Immunology, Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chi WY, Hu Y, Huang HC, Kuo HH, Lin SH, Kuo CTJ, Tao J, Fan D, Huang YM, Wu AA, Hung CF, Wu TC. Molecular targets and strategies in the development of nucleic acid cancer vaccines: from shared to personalized antigens. J Biomed Sci 2024; 31:94. [PMID: 39379923 PMCID: PMC11463125 DOI: 10.1186/s12929-024-01082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Recent breakthroughs in cancer immunotherapies have emphasized the importance of harnessing the immune system for treating cancer. Vaccines, which have traditionally been used to promote protective immunity against pathogens, are now being explored as a method to target cancer neoantigens. Over the past few years, extensive preclinical research and more than a hundred clinical trials have been dedicated to investigating various approaches to neoantigen discovery and vaccine formulations, encouraging development of personalized medicine. Nucleic acids (DNA and mRNA) have become particularly promising platform for the development of these cancer immunotherapies. This shift towards nucleic acid-based personalized vaccines has been facilitated by advancements in molecular techniques for identifying neoantigens, antigen prediction methodologies, and the development of new vaccine platforms. Generating these personalized vaccines involves a comprehensive pipeline that includes sequencing of patient tumor samples, data analysis for antigen prediction, and tailored vaccine manufacturing. In this review, we will discuss the various shared and personalized antigens used for cancer vaccine development and introduce strategies for identifying neoantigens through the characterization of gene mutation, transcription, translation and post translational modifications associated with oncogenesis. In addition, we will focus on the most up-to-date nucleic acid vaccine platforms, discuss the limitations of cancer vaccines as well as provide potential solutions, and raise key clinical and technical considerations in vaccine development.
Collapse
Affiliation(s)
- Wei-Yu Chi
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Hu
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hsin-Che Huang
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hui-Hsuan Kuo
- Pharmacology PhD Program, Weill Cornell Medicine, New York, NY, USA
| | - Shu-Hong Lin
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas Graduate School of Biomedical Sciences at Houston and MD Anderson Cancer Center, Houston, TX, USA
| | - Chun-Tien Jimmy Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Julia Tao
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Darrell Fan
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Yi-Min Huang
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Annie A Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T-C Wu
- Department of Pathology, Johns Hopkins School of Medicine, 1550 Orleans St, CRB II Room 309, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Obstetrics and Gynecology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
37
|
Koido S, Taguchi J, Shimabuku M, Kan S, Bito T, Misawa T, Ito Z, Uchiyama K, Saruta M, Tsukinaga S, Suka M, Yanagisawa H, Sato N, Ohkusa T, Shimodaira S, Sugiyama H. Dendritic cells pulsed with multifunctional Wilms' tumor 1 (WT1) peptides combined with multiagent chemotherapy modulate the tumor microenvironment and enable conversion surgery in pancreatic cancer. J Immunother Cancer 2024; 12:e009765. [PMID: 39384197 PMCID: PMC11474828 DOI: 10.1136/jitc-2024-009765] [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: 05/30/2024] [Accepted: 09/08/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND We aimed to develop a chemoimmunotherapy regimen consisting of a novel Wilms' tumor 1 (WT1) peptide-pulsed dendritic cell (WT1-DC) vaccine and multiagent chemotherapy and to investigate the safety, clinical outcomes, and WT1-specific immune responses of patients with unresectable advanced pancreatic ductal adenocarcinoma (UR-PDAC) who received this treatment. METHODS Patients with UR-PDAC with stage III disease (locally advanced (LA-PDAC; n=6)), stage IV disease (metastatic (M-PDAC; n=3)), or recurrent disease after surgery (n=1) were enrolled in this phase I study. The patients received one cycle of nab-paclitaxel plus gemcitabine alone followed by 15 doses of the WT1-DC vaccine independent of chemotherapy. The novel WT1 peptide cocktail was composed of a multifunctional helper peptide specific for major histocompatibility complex class II, human leukocyte antigen (HLA)-A*02:01, or HLA-A*02:06 and a killer peptide specific for HLA-A*24:02. RESULTS The chemoimmunotherapy regimen was well tolerated. In the nine patients for whom a prognostic analysis was feasible, the clinical outcomes of long-term WT1 peptide-specific delayed-type hypersensitivity (WT1-DTH)-positive patients (n=4) were significantly superior to those of short-term WT1-DTH-positive patients (n=5). During chemoimmunotherapy, eight patients were deemed eligible for conversion surgery and underwent R0 resection (four patients with LA-PDAC, one patient with M-PDAC, and one recurrence) or R1 resection (one patient with M-PDAC), and one patient with LA-PDAC was determined to be unresectable. Long-term WT1-DTH positivity was observed in three of the four patients with R0-resected LA-PDAC. These three patients exhibited notable infiltration of T cells and programmed cell death protein-1+ cells within the pancreatic tumor microenvironment (TME). All patients with long-term WT1-DTH positivity were alive for at least 4.5 years after starting therapy. In patients with long-term WT1-DTH positivity, the percentage of WT1-specific circulating CD4+ or CD8+ T cells that produced IFN-γ or TNF-α was significantly greater than that in patients with short-term WT1-DTH positivity after two vaccinations. Moreover, after 12 vaccinations, the percentages of both circulating regulatory T cells and myeloid-derived suppressor cells were significantly lower in patients with long-term WT1-DTH-positive PDAC than in short-term WT1-DTH-positive patients. CONCLUSIONS Potent activation of WT1-specific immune responses through a combination chemoimmunotherapy regimen including the WT1-DC vaccine in patients with UR-PDAC may modulate the TME and enable conversion surgery, resulting in clinical benefits (Online supplemental file 1). TRIAL REGISTRATION NUMBER jRCTc030190195.
Collapse
Affiliation(s)
- Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | | | | | - Shin Kan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Tuuse Bito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Takeyuki Misawa
- Department of Surgery, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Zensho Ito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Kan Uchiyama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Minato-ku, Japan
| | - Shintaro Tsukinaga
- Department of Endoscopy, The Jikei University Kashiwa Hospital, Kashiwa, Japan
| | - Machi Suka
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Minato-ku, Japan
| | - Hiroyuki Yanagisawa
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Minato-ku, Japan
| | - Nobuhiro Sato
- Department of Microbiota Research, Juntendo University, Bunkyo-ku, Japan
| | - Toshifumi Ohkusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University Kashiwa Hospital, Kashiwa, Japan
- Department of Microbiota Research, Juntendo University, Bunkyo-ku, Japan
| | | | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University, Suita, Japan
| |
Collapse
|
38
|
Kheirkhah AH, Habibi S, Yousefi MH, Mehri S, Ma B, Saleh M, Kavianpour M. Finding potential targets in cell-based immunotherapy for handling the challenges of acute myeloid leukemia. Front Immunol 2024; 15:1460437. [PMID: 39411712 PMCID: PMC11474923 DOI: 10.3389/fimmu.2024.1460437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Acute myeloid leukemia (AML) is a hostile hematological malignancy under great danger of relapse and poor long-term survival rates, despite recent therapeutic advancements. To deal with this unfulfilled clinical necessity, innovative cell-based immunotherapies have surfaced as promising approaches to improve anti-tumor immunity and enhance patient outcomes. In this comprehensive review, we provide a detailed examination of the latest developments in cell-based immunotherapies for AML, including chimeric antigen receptor (CAR) T-cell therapy, T-cell receptor (TCR)-engineered T-cell therapy, and natural killer (NK) cell-based therapies. We critically evaluate the unique mechanisms of action, current challenges, and evolving strategies to improve the efficacy and safety of these modalities. The review emphasizes how promising these cutting-edge immune-based strategies are in overcoming the inherent complexities and heterogeneity of AML. We discuss the identification of optimal target antigens, the importance of mitigating on-target/off-tumor toxicity, and the need to enhance the persistence and functionality of engineered immune effector cells. All things considered, this review offers a thorough overview of the rapidly evolving field of cell-based immunotherapy for AML, underscoring the significant progress made and the ongoing efforts to translate these innovative approaches into more effective and durable treatments for this devastating disease.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Killer Cells, Natural/immunology
- Immunotherapy/methods
- Antigens, Neoplasm/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Amir Hossein Kheirkhah
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biotechnology, School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Bin Ma
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mahshid Saleh
- Wisconsin National Primate Research Center, University of Wisconsin Graduate School, Madison, WI, United States
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
39
|
Lu C, Zou L, Wang Q, Sun M, Shi T, Xu S, Meng F, Du J. Potent antitumor activity of a bispecific T-cell engager antibody targeting the intracellular antigen KRAS G12V. BIOMOLECULES & BIOMEDICINE 2024; 24:1424-1434. [PMID: 38752985 PMCID: PMC11379025 DOI: 10.17305/bb.2024.10431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 09/07/2024]
Abstract
Kirsten Rat Sarcoma viral oncogene homolog (KRAS) is one of the most frequent oncogenes. However, there are limited treatment options due to its intracellular expression. To address this, we developed a novel bispecific T-cell engager (BiTE) antibody targeting HLA-A2/KRAS G12V complex and CD3 (HLA-G12V/CD3 BiTE). We examined its specific binding to tumor cells and T cells, as well as its anti-tumor effects in vivo. HLA-G12V/CD3 BiTE was expressed in Escherichia coli and its binding affinities to CD3 and HLA-A2/KRAS G12V were measured by flow cytometry, along with T-cell activation. In a xenograft pancreatic tumor model, the HLA-G12V/CD3 BiTE's anti-tumor effects were assessed through tumor growth, survival time, and safety. Our results demonstrated specific binding of HLA-G12V/CD3 BiTE to tumor cells with an HLA-A2/KRAS G12V mutation and T cells. The HLA-G12V/CD3 BiTE also activated T-cells in the presence of tumor cells in vitro. HLA-G12V/CD3 BiTE in vivo testing showed delayed tumor growth without severe toxicity to major organs and prolonged mouse survival. This study highlights the potential of constructing BiTEs recognizing an HLA-peptide complex and providing a novel therapy for cancer treatment targeting the intracellular tumor antigen.
Collapse
Affiliation(s)
- Changchang Lu
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Lu Zou
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiaoli Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mengna Sun
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianyu Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shuang Xu
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fanyan Meng
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Juan Du
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
40
|
Dong L, Cao Z, Chen M, Liu Y, Ma X, Lu Y, Zhang Y, Feng K, Zhang Y, Meng Z, Yang Q, Wang Y, Wu Z, Han W. Inhibition of glycosphingolipid synthesis with eliglustat in combination with immune checkpoint inhibitors in advanced cancers: preclinical evidence and phase I clinical trial. Nat Commun 2024; 15:6970. [PMID: 39138212 PMCID: PMC11322526 DOI: 10.1038/s41467-024-51495-3] [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: 05/14/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
Glycosphingolipids (GSLs) are abundantly expressed in cancer cells. The effects of GSL-targeted immunotherapies are not fully understood. Here, we show that the inhibition of GSL synthesis with the UDP-glucose ceramide glucosyltransferase inhibitor eliglustat can increase the exposure of the major histocompatibility complex (MHC) and tumour antigen peptides, enhancing the antitumour response of CD8+ T cells in a range of tumour models. We therefore conducted a proof-of-concept phase I trial on the combination of eliglustat and an anti-PD-1 antibody for the treatment of advanced cancers (NCT04944888). The primary endpoints were safety and feasibility, and the secondary endpoint was antitumor activity. All prespecified endpoints were met. Among the 31 enrolled patients, only 1 patient experienced a grade 3 adverse event (AE), and no grade 4 AEs were observed. The objective response rate was 22.6% and the disease control rate reached 71%. Of the 8 patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) colorectal cancer, one achieved complete response and two each had partial response and stable disease. In summary, inhibiting the synthesis of GSLs might represent an effective immunotherapy approach.
Collapse
Affiliation(s)
- Liang Dong
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhi Cao
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Meixia Chen
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yang Liu
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xinran Ma
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Yuting Lu
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yan Zhang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Kaichao Feng
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yang Zhang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zhenzhen Meng
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Qingming Yang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yao Wang
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Zhiqiang Wu
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Weidong Han
- Department of Bio-therapeutic, the First Medical Centre, Chinese PLA General Hospital, Beijing, China.
- School of Medicine, Nankai University, Tianjin, China.
- Changping Laboratory, Beijing, China.
- National Clinical Research Centre for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
41
|
Ito M, Koido S, Iwamoto T, Morimoto S, Fujiki F, Sugiyama H, Matsumoto S, Effenberger C, Kiyotani K, Shiba K. Enhancing the immunogenicity of Wilms tumor 1 epitope in mesothelioma cells with immunoproteasome inhibitors. PLoS One 2024; 19:e0308330. [PMID: 39116074 PMCID: PMC11309442 DOI: 10.1371/journal.pone.0308330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
The immunogenicity of cancer cells is influenced by several factors, including the expression of the major histocompatibility complex class I (MHC-I), antigen expression, and the repertoire of proteasome-produced epitope peptides. The malignant pleural mesothelioma cell line ACC-MEOS-4 (MESO-4) expresses high levels of MHC-I and Wilms tumor 1 (WT1) tumor antigens. Using a functional T cell reporter assay specific for the HLA-A*24:02 restricted WT1 epitope (WT1235, CMTWNQMNL), we searched for factors that augmented the immunogenicity of MESO-4, focusing on proteasomes, which have a central role in the antigen processing machinery. ONX-0914, a selective inhibitor of the immunoproteasome subunit β5i, enhanced immunogenicity dose-dependently at low concentrations without cytotoxicity. In addition, CD8+ T lymphocytes recognizing WT1 showed greater cytotoxicity against MESO-4 pre-treated with ONX-0914. MESO-4 expresses a standard proteasome (SP) and immunoproteasome (IP). Notably, IP has distinct catalytic activity from SP, favoring the generation of antigenic peptides with high affinity for MHC-I in antigen-presenting cells and cancer cells. In vitro, immunoproteasome digestion assay and mass spectrometry analysis showed that IP cleaved WT1235 internally after the hydrophobic residues. Importantly, this internal cleavage of the WT1235 epitope was mitigated by ONX-0914. These results suggest that ONX-0914 prevents the internal destructive cleavage of WT1235 by IP, thereby promoting the specific presentation of the WT1 epitope by MESO-4. In conclusion, selective IP inhibitors might offer a means to modulate cancer cell immunogenicity by directing the presentation of particular tumor epitopes.
Collapse
Affiliation(s)
- Masaki Ito
- Institute of Clinical Medicine and Research, Research Center for Medical Sciences, The Jikei University School of Medicine, Chiba, Japan
- Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigeo Koido
- The Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Chiba, Japan
| | - Takeo Iwamoto
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Soyoko Morimoto
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Fumihiro Fujiki
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Saki Matsumoto
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Clara Effenberger
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kiyotaka Shiba
- Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
42
|
Forchhammer S, Pop OT, Hahn M, Aebischer V, Seitz CM, Schroeder C, Liebmann A, Abele M, Wild H, Bien E, Kunc M, Schneider DT, Cuk K, Büttel I, Flemmig C, Peters M, Laible M, Brück P, Türeci Ö, Sahin U, Flatz L, Brecht IB. Expression of the tumor antigens NY-ESO-1, tyrosinase, MAGE-A3, and TPTE in pediatric and adult melanoma: a retrospective case control study. Virchows Arch 2024; 485:335-346. [PMID: 38890171 PMCID: PMC11329550 DOI: 10.1007/s00428-024-03846-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Tumor-associated antigens (TAAs) are potential targets for T cell-based immunotherapy approaches in cutaneous melanoma. BNT111, an investigational lipoplex-formulated mRNA-based therapeutic cancer vaccine encoding melanoma TAAs NY-ESO-1, tyrosinase, MAGE-A3, and TPTE, is undergoing clinical testing in adults. Expression of these TAAs in pediatric melanoma is unclear but is a prerequisite for feasibility of this treatment approach in children with melanoma. Our main objective was to characterize expression of those TAAs in pediatric melanomas compared to control cohorts. In this retrospective case control study, protein and transcript expression of NY-ESO-1, tyrosinase, MAGE-A3, and TPTE were analyzed in a cohort of 25 pediatric melanomas, 31 melanomas of young adults, 29 adult melanomas, and 30 benign melanocytic nevi in children using immunohistochemical staining and digital pathology (QuPath) and reverse transcription quantitative PCR. Based on IHC analysis, pediatric melanomas expressed tyrosinase (100.0%), TPTE (44.0%), MAGE-A3 (12.0%), and NY-ESO-1 (8.0%). Young adult melanomas expressed tyrosinase (96.8%), NY-ESO-1 (19.4%), MAGE-A3 (19.4%), and TPTE (3.2%). Adult melanomas expressed tyrosinase (86.2%), MAGE-A3 (75.9%), NY-ESO-1 (48.3%), and TPTE (48.3%). Childhood melanocytic nevi only expressed tyrosinase (93.3%). Expression prevalence of individual TAAs did not differ between subtypes of pediatric melanoma, and no association with prognosis was found. All four TAAs were expressed in pediatric melanoma, albeit NY-ESO-1 and MAGE-A3 to a lesser extent than in adult melanoma. These data support the possibility of investigating vaccines targeting these TAAs for the treatment of pediatric melanoma.
Collapse
Affiliation(s)
- Stephan Forchhammer
- Department of Dermatology, Eberhard Karls University of Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany.
| | - Oltin Tiberiu Pop
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Matthias Hahn
- Department of Dermatology, Eberhard Karls University of Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Valentin Aebischer
- Department of Dermatology, Eberhard Karls University of Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Christian M Seitz
- Pediatric Hematology and Oncology, Children's Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Alexandra Liebmann
- Institute of Medical Genetics and Applied Genomics, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Michael Abele
- Pediatric Hematology and Oncology, Children's Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Hannah Wild
- Pediatric Hematology and Oncology, Children's Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Ewa Bien
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdansk, Gdansk, Poland
| | - Michal Kunc
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Dominik T Schneider
- Clinic of Pediatrics, Dortmund Municipal Hospital, University Witten/Herdecke, Dortmund, Germany
| | | | | | | | | | | | | | | | | | - Lukas Flatz
- Department of Dermatology, Eberhard Karls University of Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Department of Dermatology, Venereology, and Allergology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Ines B Brecht
- Pediatric Hematology and Oncology, Children's Hospital, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
43
|
Yao X, Matosevic S. Generation and evaluation of cancer binding capacity of HLA-A2-WT1 complex-targeting antibody. Immunol Lett 2024; 268:106881. [PMID: 38810886 DOI: 10.1016/j.imlet.2024.106881] [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: 12/28/2023] [Revised: 05/03/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
Wilms' tumor (WT1), a transcription factor highly expressed in various leukemias and solid tumors, is a highly specific intracellular tumor antigen, requiring presentation through complexation with HLA-restricted peptides.. WT1-derived epitopes are able to assemble with MHC-I and thereby be recognized by T cell receptors (TCR). Identification of new targetable epitopes derived from WT1 on solid tumors is a challenge, but meaningful for the development of therapeutics that could in this way target intracellular oncogenic proteins. In this study, we developed and comprehensively describe methods to validate the formation of the complex of WT1126-134 and HLA-A2. Subsequently, we developed an antibody fragment able to recognize the extracellular complex on the surface of cancer cells. The single chain variable fragment (scFv) of an established TCR-mimic antibody, specifically recognizing the WT1-derived peptide presented by the HLA-A2 complex, was expressed, purified, and functionally validated using a T2 cell antigen presentation model. Furthermore, we evaluated the potential of the WT1-derived peptide as a targetable extracellular antigen in multiple solid tumor cell lines. Our study describes methodology for the evaluation of WT1-derived peptides as tumor-specific antigen on solid tumors, and may facilitate the selection of potential candidates for future immunotherapy targeting WT1 epitopes.
Collapse
MESH Headings
- Humans
- WT1 Proteins/immunology
- WT1 Proteins/metabolism
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/metabolism
- Cell Line, Tumor
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Protein Binding
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Antigen Presentation/immunology
- Epitopes/immunology
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Peptides/immunology
- Peptides/chemistry
- Peptides/metabolism
Collapse
Affiliation(s)
- Xue Yao
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, USA
| | - Sandro Matosevic
- Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, IN, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
44
|
Jiang Y, Zhang G, Zhu J, Wang X, Tao Z, Yu P. Development and validation of a TAAbs and TAAs based non-invasive model for diagnosing lung cancer. Heliyon 2024; 10:e33888. [PMID: 39027487 PMCID: PMC11255565 DOI: 10.1016/j.heliyon.2024.e33888] [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: 11/16/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
Background Single Tumor-associated autoantibodies (TAAbs) and tumor-associated antigens (TAAs) have been found to have lower diagnostic efficacy in lung cancer. Our objective is to develop and validate a lung cancer prediction model that utilizes TAAbs and TAAs and to enhance the accuracy of lung cancer detection. Methods 1830 subjects were randomly divided into training and validation sets at a 7:3 ratio for this study. Lasso regression analysis was used to remove collinear variables, whereas univariate logistic regression analysis was employed to identify potential independent risk factors for lung cancer. A diagnostic model was constructed using multivariate logistic analysis. The results were presented as a nomogram and assessed for various performance measures, including area under the curve, calibration curve, and decision curve analysis. Results The diagnostic model was developed using gender, age, GAGE7, MAGE-A1, CA125, and CEA as variables. The training set had an AUC of 0.787, while the validation set had an AUC of 0.750. The calibration curves of the training and validation sets showed a strong agreement between anticipated and observed values. The nomogram performed better than any individual variable in both the training and validation sets in terms of net benefits for lung cancer detection, according to DCA analysis. Conclusions This study proposes a diagnostic model for lung cancer that uses TAAbs and TAAs and incorporates individual characteristics. This model can be easily applied to personalized diagnosis.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Gong Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Jiayi Zhu
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xuchu Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Pan Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| |
Collapse
|
45
|
Zhang J, Terreni M, Liu F, Sollogoub M, Zhang Y. Ganglioside GM3-based anticancer vaccines: Reviewing the mechanism and current strategies. Biomed Pharmacother 2024; 176:116824. [PMID: 38820973 DOI: 10.1016/j.biopha.2024.116824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Ganglioside GM3 is one of the most common membrane-bound glycosphingolipids. The over-expression of GM3 on tumor cells makes it defined as a tumor-associated carbohydrate antigen (TACA). The specific expression property in cancers, especially in melanoma, make it become an important target to develop anticancer vaccines or immunotherapies. However, in the manner akin to most TACAs, GM3 is an autoantigen facing with problems of low immunogenicity and easily inducing immunotolerance, which means itself only cannot elicit a powerful enough immune response to prevent or treat cancer. With a comparative understanding of the mechanisms that how immune system responses to the carbohydrate vaccines, this review summarizes the studies on the recent efforts to development GM3-based anticancer vaccines.
Collapse
Affiliation(s)
- Jiaxu Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Marco Terreni
- Drug Sciences Department, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Fang Liu
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Matthieu Sollogoub
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France
| | - Yongmin Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, Paris 75005, France; College of Life Sciences, Northwest University, Xi'an 710069, China.
| |
Collapse
|
46
|
Nagai H, Chen H, Karube R, Koitabashi Y, Numata O, Yamahara K. Combination of Radiation Therapy, Wim's Tumor 1 (WT1) Dendritic Cell Vaccine Therapy, and α-Galactosylceramide-Pulsed Dendritic Cell Vaccine Therapy for End-Stage Small Bowel Cancer. Cureus 2024; 16:e64972. [PMID: 39035592 PMCID: PMC11259906 DOI: 10.7759/cureus.64972] [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] [Accepted: 07/20/2024] [Indexed: 07/23/2024] Open
Abstract
There is no established treatment for terminal cancer patients who no longer respond to surgery, radiotherapy, or chemotherapy, and palliative care is the standard worldwide. We performed intensity-modulated radiation therapy for pain relief in a 40-year-old male patient with end-stage small intestinal cancer who had been diagnosed with a life expectancy of two months after chemotherapy had been ineffective. Subsequent administration of seven doses of dendritic cell vaccine recognizing Wim's tumor 1 (WT1) and α-galactosylceramide antigens resulted in significant shrinkage of the cancer and marked improvement of the patient's general condition. The combination therapy of radiotherapy and dendritic cell vaccine therapy may suppress cancer progression and prolong survival, even in patients with chemotherapy-refractory terminal cancer. In particular, double dendritic cell vaccine therapy with WT1 and α-galactosylceramide-pulsed dendritic cell may provide an anti-tumor immune effect that is superior to that of the respective monotherapy.
Collapse
Affiliation(s)
- Hisashi Nagai
- Department of Human Development - Environment and Resources, Graduate School of Human and Environmental Studies, Tokai University, Kanagawa, JPN
- Department of Regenerative Medicine, Ginza Phoenix Clinic, Tokyo, JPN
| | - Hao Chen
- Department of Respiratory Medicine, Yokohama City University Hospital, Yokohama, JPN
| | - Ryusuke Karube
- Department of Regenerative Medicine, Ginza Phoenix Clinic, Tokyo, JPN
| | - Yusuke Koitabashi
- Department of Regenerative Medicine, Ginza Phoenix Clinic, Tokyo, JPN
| | - Ouka Numata
- Department of Regenerative Medicine, Ginza Phoenix Clinic, Tokyo, JPN
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, Hyogo, JPN
| |
Collapse
|
47
|
Farhangnia P, Ghods R, Falak R, Zarnani AH, Delbandi AA. Identification of placenta-specific protein 1 (PLAC-1) expression on human PC-3 cell line-derived prostate cancer stem cells compared to the tumor parental cells. Discov Oncol 2024; 15:251. [PMID: 38943028 PMCID: PMC11213845 DOI: 10.1007/s12672-024-01121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024] Open
Abstract
Placenta-specific protein 1 (PLAC-1) is a gene primarily expressed in the placenta and the testis. Interestingly, it is also found to be expressed in many solid tumors, and it is involved in malignant cell features. However, no evidence has been reported regarding the relationship between PLAC-1 and cancer stem cells (CSCs). In the current research, we explored the expression of the PLAC-1 molecule in prostate cancer stem cells (PCSCs) derived from the human PC-3 cell line. The enrichment of PCSCs was achieved using a three-dimensional cell culture technique known as the sphere-formation assay. To confirm the identity of PCSCs, we examined the expression of genes associated with stemness and pluripotency, such as SOX2, OCT4, Nanog, C-Myc, and KLF-4, as well as stem cell differentiation molecules like CD44 and CD133. These evaluations were conducted in both the PCSCs and the original tumor cells (parental cells) using real-time PCR and flow cytometry. Subsequently, we assessed the expression of the PLAC-1 molecule in both enriched cells and parental tumor cells at the gene and protein levels using the same techniques. The tumor cells from the PC-3 cell line formed spheroids with CSC characteristics in a non-adherent medium. The expression of SOX2, OCT4, Nanog, and C-Myc genes (p < 0.01), and the molecules CD44 and CD133 (p < 0.05) were significantly elevated in PCSCs compared to the parental cells. The expression of the PLAC-1 molecule in PCSCs showed a significant increase compared to the parental cells at both gene (p < 0.01) and protein (p < 0.001) levels. In conclusion, it was indicated for the first time that PLAC-1 is up-regulated in PCSCs derived from human PC-3 cell line. This study may propose PLAC-1 as a potential target in targeted therapies, which should be confirmed through further studies.
Collapse
Affiliation(s)
- Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali-Akbar Delbandi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Reproductive Sciences and Technology Research Center, Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Yuan DY, McKeague ML, Raghu VK, Schoen RE, Finn OJ, Benos PV. Cellular and transcriptional profiles of peripheral blood mononuclear cells pre-vaccination predict immune response to preventative MUC1 vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598031. [PMID: 38948837 PMCID: PMC11212910 DOI: 10.1101/2024.06.14.598031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
A single arm trial (NCT007773097) and a double-blind, placebo controlled randomized trial ( NCT02134925 ) were conducted in individuals with a history of advanced colonic adenoma to test the safety and immunogenicity of the MUC1 tumor antigen vaccine and its potential to prevent new adenomas. These were the first two trials of a non-viral cancer vaccine administered in the absence of cancer. The vaccine was safe and strongly immunogenic in 43% (NCT007773097) and 25% ( NCT02134925 ) of participants. The lack of response in a significant number of participants suggested, for the first time, that even in a premalignant setting, the immune system may have already been exposed to some level of suppression previously reported only in cancer. Single-cell RNA-sequencing (scRNA-seq) on banked pre-vaccination peripheral blood mononuclear cells (PBMCs) from 16 immune responders and 16 non-responders identified specific cell types, genes, and pathways of a productive vaccine response. Responders had a significantly higher percentage of CD4+ naive T cells pre-vaccination, but a significantly lower percentage of CD8+ T effector memory (TEM) cells and CD16+ monocytes. Differential gene expression (DGE) and transcription factor inference analysis showed a higher level of expression of T cell activation genes, such as Fos and Jun, in CD4+ naive T cells, and pathway analysis showed enriched signaling activity in responders. Furthermore, Bayesian network analysis suggested that these genes were mechanistically connected to response. Our analyses identified several immune mechanisms and candidate biomarkers to be further validated as predictors of immune responses to a preventative cancer vaccine that could facilitate selection of individuals likely to benefit from a vaccine or be used to improve vaccine responses.
Collapse
|
49
|
Emens LA, Romero PJ, Anderson AC, Bruno TC, Capitini CM, Collyar D, Gulley JL, Hwu P, Posey AD, Silk AW, Wargo JA. Challenges and opportunities in cancer immunotherapy: a Society for Immunotherapy of Cancer (SITC) strategic vision. J Immunother Cancer 2024; 12:e009063. [PMID: 38901879 PMCID: PMC11191773 DOI: 10.1136/jitc-2024-009063] [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: 04/24/2024] [Indexed: 06/22/2024] Open
Abstract
Cancer immunotherapy has flourished over the last 10-15 years, transforming the practice of oncology and providing long-term clinical benefit to some patients. During this time, three distinct classes of immune checkpoint inhibitors, chimeric antigen receptor-T cell therapies specific for two targets, and two distinct classes of bispecific T cell engagers, a vaccine, and an oncolytic virus have joined cytokines as a standard of cancer care. At the same time, scientific progress has delivered vast amounts of new knowledge. For example, advances in technologies such as single-cell sequencing and spatial transcriptomics have provided deep insights into the immunobiology of the tumor microenvironment. With this rapid clinical and scientific progress, the field of cancer immunotherapy is currently at a critical inflection point, with potential for exponential growth over the next decade. Recognizing this, the Society for Immunotherapy of Cancer convened a diverse group of experts in cancer immunotherapy representing academia, the pharmaceutical and biotechnology industries, patient advocacy, and the regulatory community to identify current opportunities and challenges with the goal of prioritizing areas with the highest potential for clinical impact. The consensus group identified seven high-priority areas of current opportunity for the field: mechanisms of antitumor activity and toxicity; mechanisms of drug resistance; biomarkers and biospecimens; unique aspects of novel therapeutics; host and environmental interactions; premalignant immunity, immune interception, and immunoprevention; and clinical trial design, endpoints, and conduct. Additionally, potential roadblocks to progress were discussed, and several topics were identified as cross-cutting tools for optimization, each with potential to impact multiple scientific priority areas. These cross-cutting tools include preclinical models, data curation and sharing, biopsies and biospecimens, diversification of funding sources, definitions and standards, and patient engagement. Finally, three key guiding principles were identified that will both optimize and maximize progress in the field. These include engaging the patient community; cultivating diversity, equity, inclusion, and accessibility; and leveraging the power of artificial intelligence to accelerate progress. Here, we present the outcomes of these discussions as a strategic vision to galvanize the field for the next decade of exponential progress in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Ana Carrizosa Anderson
- The Gene Lay Institute of Immunology and Inflammation, Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Tullia C Bruno
- Department of Immunology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christian M Capitini
- Department of Pediatrics and Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Deborah Collyar
- Patient Advocates in Research (PAIR), Danville, California, USA
| | - James L Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Avery D Posey
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
50
|
Dąbrowski A, Nowicki M, Budzyńska A, Suchodolski J, Ogórek R, Chabowski M, Przywara K. Analysis of CYP1B1 Polymorphisms in Lung Cancer Patients Using Novel, Quick and Easy Methods Based on CAPS and ACRS-PCR Techniques. Int J Mol Sci 2024; 25:6676. [PMID: 38928381 PMCID: PMC11203417 DOI: 10.3390/ijms25126676] [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: 05/07/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Within the sequence of the CYP1B1 gene, more than 50 polymorphisms, resulting from single-nucleotide polymorphisms (SNPs), have been described. Some of them play an important role as specific genetic markers in the process of carcinogenesis and for therapeutic purposes. In this publication, we present methods we have developed that enable the specific and unambiguous identification of four polymorphisms that result in amino acid changes: c. 142C > G, c. 355G > T, c. 1294C > G, and c. 1358A > G. Our studies are based on cleaved amplified polymorphic sequences (CAPSs) and artificially created restriction site (ACRS) PCR techniques; therefore, they require only basic laboratory equipment and low financial outlays. Utilizing the described methods allows for the reduction of research time and cost, and the minimization of errors. Their effectiveness and efficiency depend on the careful design of appropriate primers and the precise selection of suitable restriction enzymes. As a result, further confirmation by sequencing is not necessary. Using the developed method, we examined 63 patients diagnosed with lung cancer and observed a 1.5 to 2.1 times higher frequency of the analyzed single-nucleotide polymorphisms compared to the frequency in the European population.
Collapse
Affiliation(s)
- Adam Dąbrowski
- Laboratory of Molecular Diagnostics “Bio-Genetik” NZOZ, 50-525 Wrocław, Poland;
| | - Maciej Nowicki
- Department of Surgery, 4th Military Teaching Hospital, 53-114 Wroclaw, Poland; (M.N.); (M.C.)
| | - Aleksandra Budzyńska
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, 51-148 Wrocław, Poland; (A.B.); (J.S.); (R.O.)
| | - Jakub Suchodolski
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, 51-148 Wrocław, Poland; (A.B.); (J.S.); (R.O.)
| | - Rafał Ogórek
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, 51-148 Wrocław, Poland; (A.B.); (J.S.); (R.O.)
| | - Mariusz Chabowski
- Department of Surgery, 4th Military Teaching Hospital, 53-114 Wroclaw, Poland; (M.N.); (M.C.)
- Department of Nursing and Obstetrics, Division of Anesthesiological and Surgical Nursing, Faculty of Health Science, Wroclaw Medical University, 50-556 Wroclaw, Poland
- Department of Clinical Surgical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, 51-377 Wroclaw, Poland
| | - Katarzyna Przywara
- Department of Mycology and Genetics, Faculty of Biological Sciences, University of Wrocław, 51-148 Wrocław, Poland; (A.B.); (J.S.); (R.O.)
| |
Collapse
|