1
|
Long Z, Yi Z, Yan W, Wang H. Trends in the immunotherapy for glioblastoma: A two-decade bibliometric analysis. Hum Vaccin Immunother 2025; 21:2466299. [PMID: 39950580 PMCID: PMC11834472 DOI: 10.1080/21645515.2025.2466299] [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: 08/30/2024] [Revised: 01/27/2025] [Accepted: 02/09/2025] [Indexed: 02/20/2025] Open
Abstract
Glioblastoma is a life-threatening primary malignant brain tumor with an unfavorable prognosis. Contributing factors to its poor outcome include tumor heterogeneity, low mutational burden, and immunosuppression within the tumor microenvironment. Recognizing these challenges, immunotherapeutic strategies have emerged as a promising avenue for glioblastoma treatment. Although several dynamic research and scientific trend have increasingly taken pace in the immunotherapeutic approaches to glioblastoma, systematic bibliometric studies on such trends are few. On this note, this study explores a bibliometric analysis of the research hotspots and trends in glioblastoma immunotherapy. We conducted a search in the Web of Science Core Collection database for articles on glioblastoma immunotherapy published between 2004 and 2024. Using VOSviewer and CiteSpace software, we analyzed collected articles to explore aspects such as country of origin, journal of publication, affiliated institute, authorship, keywords, and citation patterns. As of May 1, 2024, we retrieved 3,729 papers on Glioblastoma Immunotherapy. In the field of glioblastoma immunotherapy, the United States stands out as the leading contributor, with 1,708 publications and a substantial 90,590 citations. Following closely, China has made significant contributions through 926 publications, earning 17,533 citations, while Germany adds to the body of knowledge with 349 publications and 16,355 citations. Furthermore, Authoritative journals in this field include Clinical Cancer Research and Neuro-Oncology. The top five keywords during this period were temozolomide, radiotherapy, dendritic cell, cytotoxic T lymphocyte, and vaccination. Moreover, Hotspots in the field include immune checkpoint inhibitors and chimeric antigen receptor T cell therapy.
Collapse
Affiliation(s)
- Zhi Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Yan
- The First Department of General Surgery, Hunan Provincial People’s Hospital, Hunan Normal University, Changsha, China
| | - Hongxin Wang
- Department of Neurosurgery, The Affiliated Changsha Central Hospital, Hengyang Medical School,University of South China, Changsha, China
| |
Collapse
|
2
|
Mojica CV, Gutierrez KME, Mason WP. Advances in IDH-mutant glioma management: IDH inhibitors, clinical implications of INDIGO trial, and future perspectives. Future Oncol 2025:1-11. [PMID: 40424199 DOI: 10.1080/14796694.2025.2511587] [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: 10/31/2024] [Accepted: 05/23/2025] [Indexed: 05/29/2025] Open
Abstract
The discovery of isocitrate dehydrogenase (IDH) mutation in gliomas marked the new era of molecular classification of CNS tumors. Understanding the complex role of IDH mutation in oncogenesis led to the evaluation of novel small molecules targeting this enzyme as a potential therapeutic intervention. Vorasidenib, a brain-penetrant inhibitor of both IDH1 and IDH2-mutant enzymes, was one such agent. The phase 3 INDIGO trial evaluated vorasidenib and demonstrated its efficacy in IDH-mutant low-grade gliomas (LGG). This study established vorasidenib as an effective inhibitor of both IDH1 and IDH2-mutant enzymes, highlighting its great potential in advancing the therapeutic armamentarium for patients with LGG. While vorasidenib has been recently included in several treatment guidelines for CNS tumors, further research on the use of this novel agent, as monotherapy or in combination with other drugs, becomes imperative to exploit fully its potential in the management of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Christianne V Mojica
- Divisions of Neurology and Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Katrina Mari E Gutierrez
- Divisions of Neurology and Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Warren P Mason
- Divisions of Neurology and Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Skouras P, Giakoumettis G, Argyros C, Vavoulis G, Verigos EK, Giakoumettis D. Oligodendroglioma of the Hippocampus: A Case Report and Systematic Review on Therapeutic Approaches of Oligodendroglioma After WHO 2021 Classification. Pharmaceuticals (Basel) 2025; 18:349. [PMID: 40143126 PMCID: PMC11944404 DOI: 10.3390/ph18030349] [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: 01/26/2025] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Oligodendrogliomas are a molecularly distinct subtype of glioma according to the WHO 2021 tumor classification, defined as isocitrate dehydrogenase (IDH) mutations and 1p/19q co-deletion. This updated classification has changed the approach to glioma management by emphasizing the critical role of molecular diagnostics. This study explores current therapeutic strategies for adult oligodendrogliomas and contextualizes findings with a patient with a Grade 3 oligodendroglioma of the hippocampus. Methods: A systematic review was conducted, synthesizing evidence from 36 studies published between 2021 and 2024. The review focuses on surgical resection, PCV chemotherapy (procarbazine, lomustine, vincristine), and radiotherapy, with progression-free survival (PFS) and overall survival (OS) as primary outcomes. Moreover, a 45-year-old woman diagnosed with an IDH-mutant, 1p/19q-co-deleted Grade 3 oligodendroglioma is presented to illustrate clinical management. Results: The review highlights the significance of molecular profiling in personalizing treatment strategies. The findings highlight that maximal safe surgical resection combined with PCV chemotherapy and radiotherapy optimizes PFS and OS. However, our case underwent chemotherapy and radiotherapy after a multidisciplinary consultation, demonstrating favorable initial outcomes. These findings reaffirm the importance of integrating molecular insight into clinical decision-making. Conclusions: Advancements in molecular diagnostics have profoundly enhanced the personalization of therapy for oligodendrogliomas, yielding improved survival outcomes. Optimal management should entail a multidisciplinary approach incorporating surgery, chemotherapy, and radiotherapy, guided by molecular features. This study reinforces the necessity of molecular-driven strategies to improve survival and quality of care for patients with oligodendroglioma.
Collapse
Affiliation(s)
- Panagiotis Skouras
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Department of Social and Family Medicine, General Hospital of Karditsa, 43100 Thessaly, Greece
| | - Georgios Giakoumettis
- Medical Physics & Digital Innovation Laboratory, School of Medicine, Faculty of Health Sciences, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Charalampos Argyros
- Department of Neurosurgery, “Agios Savvas” General Anticancer-Oncological Hospital of Athens, 11522 Athens, Greece;
| | - George Vavoulis
- Department of Neurosurgery, “KAT” General Hospital of Athens, 14561 Kifissia, Greece;
| | - Emmanouil K. Verigos
- Department of Radiotherapy, “Agios Savvas” General Anticancer-Oncological Hospital of Athens, 11522 Athens, Greece;
| | - Dimitrios Giakoumettis
- Department of Neurosurgery, “Agios Savvas” General Anticancer-Oncological Hospital of Athens, 11522 Athens, Greece;
- Department of Neurosurgery, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
4
|
de la Fuente MI, Touat M, van den Bent MJ, Preusser M, Peters KB, Young RJ, Huang RY, Ellingson BM, Capper D, Phillips JJ, Halasz LM, Shih HA, Rudà R, Lim-Fat MJ, Blumenthal DT, Weller M, Arakawa Y, Whittle JR, Ducray F, Reardon DA, Bi WL, Minniti G, Rahman R, Hervey-Jumper S, Chang SM, Wen PY. The role of vorasidenib in the treatment of isocitrate dehydrogenase-mutant glioma. Neuro Oncol 2024:noae259. [PMID: 39723472 DOI: 10.1093/neuonc/noae259] [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] [Indexed: 12/28/2024] Open
Abstract
Isocitrate dehydrogenase (IDH)-mutant gliomas are the most common malignant primary brain tumors in young adults. This condition imposes a substantial burden on patients and their caregivers, marked by neurocognitive deficits and high mortality rates due to tumor progression, coupled with significant morbidity from current treatment modalities. Although surgery, radiation therapy, and chemotherapy improve survival, these treatments can adversely affect cognitive function, quality of life, finances, employment status, and overall independence. Consequently, there is an urgent need for innovative strategies that delay progression and the use of radiation therapy and chemotherapy. The recent Federal Drug Administration (FDA) approval of vorasidenib, a brain-penetrant small molecule targeting mutant IDH1/2 proteins, heralds a shift in the therapeutic landscape for IDH-mutant gliomas. In this review, we address the role of vorasidenib in the treatment of IDH-mutant gliomas, providing a roadmap for its incorporation into daily practice. We discuss ongoing clinical trials with vorasidenib and other IDH inhibitors, as single-agent or in combination with other therapies, as well as current challenges and future directions.
Collapse
Affiliation(s)
- Macarena I de la Fuente
- Department of Neurology, University of Miami, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Mehdi Touat
- Service de Neuro-oncologie, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, Paris Brain Institute, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Paris, France
- Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Martin J van den Bent
- Service de Neuro-oncologie, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, Paris Brain Institute, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Paris, France
- Department of Neurology, Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Katherine B Peters
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University, Durham, North Carolina, USA
| | - Robert J Young
- Service Neuroradiology, Department of Radiology, Memorial Sloan Kettering Cancer, New York, New York, USA
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - David Capper
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Berlin, Heidelberg, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Joanna J Phillips
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Lia M Halasz
- Department of Radiation Oncology, University of Washington/Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Helen A Shih
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience, University of Turin, Turin, Italy
| | - Mary Jane Lim-Fat
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Yoshiki Arakawa
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - James R Whittle
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Personalised Oncology Division, WEHI, Parkville, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - François Ducray
- Department of Neuro-Oncology, East Group Hospital, Hospices Civils de Lyon, Université de Lyon, Université Claude Bernard, Lyon, France
| | - David A Reardon
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Giuseppe Minniti
- IRCCS Neuromed, Pozzilli, Isernia, Italy
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Rifaquat Rahman
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shawn Hervey-Jumper
- Department of Neurological Surgery, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Susan M Chang
- Division of Neuro-Oncology, Department of Neurosurgery, University of California, San Francisco, California, USA
| | - Patrick Y Wen
- Center For Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Cicone F, Gnesin S, Santo G, Stokke C, Bartolomei M, Cascini GL, Minniti G, Paganelli G, Verger A, Cremonesi M. Do we need dosimetry for the optimization of theranostics in CNS tumors? Neuro Oncol 2024; 26:S242-S258. [PMID: 39351795 PMCID: PMC11631076 DOI: 10.1093/neuonc/noae200] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
Radiopharmaceutical theranostic treatments have grown exponentially worldwide, and internal dosimetry has attracted attention and resources. Despite some similarities with chemotherapy, radiopharmaceutical treatments are essentially radiotherapy treatments, as the release of radiation into tissues is the determinant of the observed clinical effects. Therefore, absorbed dose calculations are key to explaining dose-effect correlations and individualizing radiopharmaceutical treatments. The present article introduces the basic principles of internal dosimetry and provides an overview of available loco-regional and systemic radiopharmaceutical treatments for central nervous system (CNS) tumors. The specific characteristics of dosimetry as applied to these treatments are highlighted, along with their limitations and most relevant results. Dosimetry is performed with higher precision and better reproducibility than in the past, and dosimetric data should be systematically collected, as treatment planning and verification may help exploit the full potential of theranostic of CNS tumors.
Collapse
Affiliation(s)
- Francesco Cicone
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Silvano Gnesin
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giulia Santo
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Caroline Stokke
- Department of Physics, University of Oslo, Oslo, Norway
- Department of Diagnostic Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Department of Oncology and Haematology, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Giuseppe Lucio Cascini
- Nuclear Medicine Unit, Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, Catanzaro, Italy
| | - Giuseppe Minniti
- IRCCS Neuromed, Pozzilli (IS), Italy
- Radiation Oncology Unit, Department of Radiological Sciences, Oncology and Anatomical Pathology, “Sapienza” University of Rome, Rome, Italy
| | - Giovanni Paganelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori,”Meldola, Italy
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU-Nancy, IADI, INSERM, UMR 1254, Université de Lorraine, Nancy, France
| | - Marta Cremonesi
- Unit of Radiation Research, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
6
|
Baek C, Laurenge A, Touat M. Advances in the treatment of IDH-mutant gliomas. Curr Opin Neurol 2024; 37:708-716. [PMID: 39253756 DOI: 10.1097/wco.0000000000001316] [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: 09/11/2024]
Abstract
PURPOSE OF REVIEW Isocitrate dehydrogenase (IDH) mutation is a defining molecular driver of WHO grade 2-4 astrocytomas and oligodendrogliomas. In this article, we review the recent therapeutic approaches specifically targeting IDH-mutant gliomas and summarize ongoing clinical trials in this population. RECENT FINDINGS The IDH inhibitor vorasidenib recently demonstrated its efficacy after surgical resection in grade 2 IDH-mutated gliomas. Several studies in patients with IDH-mutant gliomas are currently exploring various strategies to target IDH mutations, including the use of small-molecule inhibitors, immunotherapies, peptide vaccines and agents targeting metabolic and epigenomic vulnerabilities. SUMMARY Mutant-IDH targeting holds significant promise in treating progressive or recurrent IDH-mutant gliomas. Recent results with IDH inhibitors will change practice and influence the existing guidelines in a near future.
Collapse
Affiliation(s)
- Chooyoung Baek
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
| | - Alice Laurenge
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
- Institut du Cerveau, Paris Brain Institute (ICM), Inserm, CNRS, Sorbonne Université, AP-HP, SIRIC CURAMUS, Paris, France
| | - Mehdi Touat
- Service de Neuro-oncologie, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, AP-HP, Sorbonne Université
- Institut du Cerveau, Paris Brain Institute (ICM), Inserm, CNRS, Sorbonne Université, AP-HP, SIRIC CURAMUS, Paris, France
- Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Yao L, Hatami M, Ma W, Skutella T. Vaccine-based immunotherapy and related preclinical models for glioma. Trends Mol Med 2024; 30:965-981. [PMID: 39013724 DOI: 10.1016/j.molmed.2024.06.009] [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: 02/19/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/18/2024]
Abstract
Glioma, the most common primary malignant tumor in the central nervous system (CNS), lacks effective treatments, and >60% of cases are glioblastoma (GBM), the most aggressive form. Despite advances in immunotherapy, GBM remains highly resistant. Approaches that target tumor antigens expedite the development of immunotherapies, including personalized tumor-specific vaccines, patient-specific target selection, dendritic cell (DC) vaccines, and chimeric antigen receptor (CAR) and T cell receptor (TCR) T cells. Recent studies show promising results in treating GBM and lower-grade glioma (LGG), fostering hope for future immunotherapy. This review discusses tumor vaccines against glioma, preclinical models in immunological research, and the role of CD4+ T cells in vaccine-induced antitumor immunity. We also summarize clinical approaches, challenges, and future research for creating more effective vaccines.
Collapse
Affiliation(s)
- Longping Yao
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Maryam Hatami
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Heidelberg Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
8
|
Oster C, Kessler L, Blau T, Keyvani K, Pabst KM, Fendler WP, Fragoso Costa P, Lazaridis L, Schmidt T, Feldheim J, Pierscianek D, Schildhaus HU, Sure U, Ahmadipour Y, Kleinschnitz C, Guberina N, Stuschke M, Deuschl C, Scheffler B, Herrmann K, Kebir S, Glas M. The Role of Fibroblast Activation Protein in Glioblastoma and Gliosarcoma: A Comparison of Tissue, 68Ga-FAPI-46 PET Data, and Survival Data. J Nucl Med 2024; 65:1217-1223. [PMID: 38960714 DOI: 10.2967/jnumed.123.267151] [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: 12/02/2023] [Accepted: 05/22/2024] [Indexed: 07/05/2024] Open
Abstract
Despite their unique histologic features, gliosarcomas belong to the group of glioblastomas and are treated according to the same standards. Fibroblast activation protein (FAP) is a component of a tumor-specific subpopulation of fibroblasts that plays a critical role in tumor growth and invasion. Some case studies suggest an elevated expression of FAP in glioblastoma and a particularly strong expression in gliosarcoma attributed to traits of predominant mesenchymal differentiation. However, the prognostic impact of FAP and its diagnostic and therapeutic potential remain unclear. Here, we investigate the clinical relevance of FAP expression in gliosarcoma and glioblastoma and how it correlates with 68Ga-FAP inhibitor (FAPI)-46 PET uptake. Methods: Patients diagnosed with gliosarcoma or glioblastoma without sarcomatous differentiation with an overall survival of less than 2.5 y were enrolled. Histologic examination included immunohistochemistry and semiquantitative scoring of FAP (0-3, with higher values indicating stronger expression). Additionally, 68Ga-FAPI-46 PET scans were performed in a subset of glioblastomas without sarcomatous differentiation patients. The clinical SUVs were correlated with FAP expression levels in surgically derived tumor tissue and relevant prognostic factors. Results: Of the 61 patients who were enrolled, 13 of them had gliosarcoma. Immunohistochemistry revealed significantly more FAP in gliosarcomas than in glioblastomas without sarcomatous differentiation of tumor tissue (P < 0.0001). In the latter, FAP expression was confined to the perivascular space, whereas neoplastic cells additionally expressed FAP in gliosarcoma. A significant correlation of immunohistochemical FAP with SUVmean and SUVpeak of 68Ga-FAPI-46 PET indicates that clinical tracer uptake represents FAP expression of the tumor. Although gliosarcomas express higher levels of FAP than do glioblastomas without sarcomatous differentiation, overall survival does not significantly differ between the groups. Conclusion: The analysis reveals a significant correlation between SUVmean and SUVpeak in 68Ga-FAPI-46 PET and immunohistochemical FAP expression. This study indicates that FAP expression is much more abundant in the gliosarcoma subgroup of glioblastomas. This could open not only a diagnostic but also a therapeutic gap, since FAP could be explored as a theranostic target to enhance survival in a distinct subgroup of high-risk brain tumor patients with poor survival prognosis.
Collapse
Affiliation(s)
- Christoph Oster
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen-Düsseldorf, Partnership Between DKFZ and University Hospital Essen, Essen, Germany; and DKFZ-Division of Translational Neurooncology at West German Cancer Center (WTZ), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Lukas Kessler
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; and National Center for Tumor Diseases (NCT), NCT West, Essen, Germany
| | - Tobias Blau
- Institute of Neuropathology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Kathy Keyvani
- Institute of Neuropathology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Kim M Pabst
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; and National Center for Tumor Diseases (NCT), NCT West, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; and National Center for Tumor Diseases (NCT), NCT West, Essen, Germany
| | - Pedro Fragoso Costa
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; and National Center for Tumor Diseases (NCT), NCT West, Essen, Germany
| | - Lazaros Lazaridis
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Teresa Schmidt
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Jonas Feldheim
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Daniela Pierscianek
- Department of Neurosurgery and Spine Surgery, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University Duisburg-Essen, Essen, Germany
- Department of Neurosurgery and Spine Surgery, St. Marienhospital Lünen, Lünen, Germany
| | - Hans Ulrich Schildhaus
- Institute of Pathology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
- Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany
| | - Ulrich Sure
- Department of Neurosurgery and Spine Surgery, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Yahya Ahmadipour
- Department of Neurosurgery and Spine Surgery, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Nika Guberina
- Department of Radiotherapy, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- Department of Radiotherapy, University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Cornelius Deuschl
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Björn Scheffler
- German Cancer Consortium (DKTK), Partner Site Essen-Düsseldorf, Partnership Between DKFZ and University Hospital Essen, Essen, Germany; and DKFZ-Division of Translational Neurooncology at West German Cancer Center (WTZ), University Medicine Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany; and
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; and National Center for Tumor Diseases (NCT), NCT West, Essen, Germany
- National Center for Tumor Diseases (NCT), NCT West, Heidelberg, Germany
| | - Sied Kebir
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen-Düsseldorf, Partnership Between DKFZ and University Hospital Essen, Essen, Germany; and DKFZ-Division of Translational Neurooncology at West German Cancer Center (WTZ), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Glas
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Division of Clinical Neurooncology, University Medicine Essen, University Duisburg-Essen, Essen, Germany;
- German Cancer Consortium (DKTK), Partner Site Essen-Düsseldorf, Partnership Between DKFZ and University Hospital Essen, Essen, Germany; and DKFZ-Division of Translational Neurooncology at West German Cancer Center (WTZ), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
9
|
Streibel Y, Breckwoldt MO, Hunger J, Pan C, Fischer M, Turco V, Boztepe B, Fels-Palesandro H, Scheck JG, Sturm V, Karimian-Jazi K, Agardy DA, Annio G, Mustapha R, Soni SS, Alasa A, Weidenfeld I, Rodell CB, Wick W, Heiland S, Winkler F, Platten M, Bendszus M, Sinkus R, Schregel K. Tumor biomechanics as a novel imaging biomarker to assess response to immunotherapy in a murine glioma model. Sci Rep 2024; 14:15613. [PMID: 38971907 PMCID: PMC11227492 DOI: 10.1038/s41598-024-66519-7] [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: 03/13/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
Glioblastoma is the most common and aggressive primary malignant brain tumor with poor prognosis. Novel immunotherapeutic approaches are currently under investigation. Even though magnetic resonance imaging (MRI) is the most important imaging tool for treatment monitoring, response assessment is often hampered by therapy-related tissue changes. As tumor and therapy-associated tissue reactions differ structurally, we hypothesize that biomechanics could be a pertinent imaging proxy for differentiation. Longitudinal MRI and magnetic resonance elastography (MRE) were performed to monitor response to immunotherapy with a toll-like receptor 7/8 agonist in orthotopic syngeneic experimental glioma. Imaging results were correlated to histology and light sheet microscopy data. Here, we identify MRE as a promising non-invasive imaging method for immunotherapy-monitoring by quantifying changes in response-related tumor mechanics. Specifically, we show that a relative softening of treated compared to untreated tumors is linked to the inflammatory processes following therapy-induced re-education of tumor-associated myeloid cells. Mechanistically, combined effects of myeloid influx and inflammation including extracellular matrix degradation following immunotherapy form the basis of treated tumors being softer than untreated glioma. This is a very early indicator of therapy response outperforming established imaging metrics such as tumor volume. The overall anti-tumor inflammatory processes likely have similar effects on human brain tissue biomechanics, making MRE a promising tool for gauging response to immunotherapy in glioma patients early, thereby strongly impacting patient pathway.
Collapse
Affiliation(s)
- Yannik Streibel
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Michael O Breckwoldt
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jessica Hunger
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Chenchen Pan
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Manuel Fischer
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Verena Turco
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medical Oncology, Heidelberg University Hospital, National Center for Tumor Diseases, Heidelberg, Germany
| | - Berin Boztepe
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Hannah Fels-Palesandro
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Jonas G Scheck
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Volker Sturm
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Kianush Karimian-Jazi
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dennis A Agardy
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
| | - Giacomo Annio
- INSERM UMRS1148-Laboratory for Vascular Translational Science, University Paris, Paris, France
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Rami Mustapha
- Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Shreya S Soni
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, USA
| | - Abdulrahman Alasa
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, USA
| | - Ina Weidenfeld
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Christopher B Rodell
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, USA
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Frank Winkler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Ralph Sinkus
- INSERM UMRS1148-Laboratory for Vascular Translational Science, University Paris, Paris, France
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Katharina Schregel
- Department of Neuroradiology, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DTK) within the German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
10
|
Ivanov S, Nano O, Hana C, Bonano-Rios A, Hussein A. Molecular Targeting of the Isocitrate Dehydrogenase Pathway and the Implications for Cancer Therapy. Int J Mol Sci 2024; 25:7337. [PMID: 39000443 PMCID: PMC11242572 DOI: 10.3390/ijms25137337] [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: 04/15/2024] [Revised: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The advent of comprehensive genomic profiling using next-generation sequencing (NGS) has unveiled an abundance of potentially actionable genetic aberrations that have shaped our understanding of the cancer biology landscape. Isocitrate dehydrogenase (IDH) is an enzyme present in the cytosol (IDH1) and mitochondria (IDH2 and IDH3). In the mitochondrion, it catalyzes the irreversible oxidative decarboxylation of isocitrate, yielding the production of α-ketoglutarate and nicotinamide adenine dinucleotide phosphate (NADPH) as well as carbon dioxide (CO2). In the cytosol, IDH catalyzes the decarboxylation of isocitrate to α-ketoglutarate as well as the reverse reductive carboxylation of α-ketoglutarate to isocitrate. These rate-limiting steps in the tricarboxylic acid cycle, as well as the cytoplasmic response to oxidative stress, play key roles in gene regulation, cell differentiation, and tissue homeostasis. Mutations in the genes encoding IDH1 and IDH2 and, less commonly, IDH3 have been found in a variety of cancers, most commonly glioma, acute myeloid leukemia (AML), chondrosarcoma, and intrahepatic cholangiocarcinoma. In this paper, we intend to elucidate the theorized pathophysiology behind IDH isomer mutation, its implication in cancer manifestation, and discuss some of the available clinical data regarding the use of novel IDH inhibitors and their role in therapy.
Collapse
Affiliation(s)
- Stanislav Ivanov
- Memorial Cancer Institute, Memorial Healthcare System, Pembroke Pines, FL 33028, USA; (O.N.); (A.B.-R.); (A.H.)
| | | | | | | | | |
Collapse
|
11
|
Rudà R, Horbinski C, van den Bent M, Preusser M, Soffietti R. IDH inhibition in gliomas: from preclinical models to clinical trials. Nat Rev Neurol 2024; 20:395-407. [PMID: 38760442 DOI: 10.1038/s41582-024-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
Gliomas are the most common malignant primary brain tumours in adults and cannot usually be cured with standard cancer treatments. Gliomas show intratumoural and intertumoural heterogeneity at the histological and molecular levels, and they frequently contain mutations in the isocitrate dehydrogenase 1 (IDH1) or IDH2 gene. IDH-mutant adult-type diffuse gliomas are subdivided into grade 2, 3 or 4 IDH-mutant astrocytomas and grade 2 or 3 IDH-mutant, 1p19q-codeleted oligodendrogliomas. The product of the mutated IDH genes, D-2-hydroxyglutarate (D-2-HG), induces global DNA hypermethylation and interferes with immunity, leading to stimulation of tumour growth. Selective inhibitors of mutant IDH, such as ivosidenib and vorasidenib, have been shown to reduce D-2-HG levels and induce cellular differentiation in preclinical models and to induce MRI-detectable responses in early clinical trials. The phase III INDIGO trial has demonstrated superiority of vorasidenib, a brain-penetrant pan-mutant IDH inhibitor, over placebo in people with non-enhancing grade 2 IDH-mutant gliomas following surgery. In this Review, we describe the pathway of development of IDH inhibitors in IDH-mutant low-grade gliomas from preclinical models to clinical trials. We discuss the practice-changing implications of the INDIGO trial and consider new avenues of investigation in the field of IDH-mutant gliomas.
Collapse
Affiliation(s)
- Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy.
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Martin van den Bent
- Brain Tumour Center at Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Riccardo Soffietti
- Division of Neuro-Oncology, Department of Neuroscience 'Rita Levi Montalcini', University of Turin, Turin, Italy
| |
Collapse
|
12
|
Thallner R, Gumbinger C, Hohmann A, Wick A, Wick W, Busetto L. Patient, Relative and Staff Experiences of Clinical Trial Participation in Neurooncology: "Maybe You Can Also Show the Positive, No Matter How It Ends". Cancer Manag Res 2024; 16:663-676. [PMID: 38919873 PMCID: PMC11197948 DOI: 10.2147/cmar.s447407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Purpose There is a lack of evidence regarding how patients with malignant brain tumor and their relatives experience participation in neurooncological clinical trials. Similarly, insights from the perspective of trial staff caring for this group of patients are missing. This study aims to investigate patient, relative and trial staff experiences regarding participation in clinical neurooncological trials. Methods Within a qualitative exploratory study, 29 semi-structured interviews with brain tumor patients, relatives and trial staff were conducted and analyzed using reflexive thematic analysis (RTA) by Braun and Clarke. A patient researcher and patient council were involved in data analysis and interpretation. Results Four themes were developed reflecting significant aspects of the trial experience: 1. "It all revolves around hope"; 2. "Trial participation: experiencing unique medical care"; 3. "Everyone's roles are changing"; 4. "Communication as a possible area of conflict". Experiencing trial participation and general medical treatment were found to be interconnected to such a degree that they were often not meaningfully distinguished by patients and relatives. Conclusion In addition to assessing traditional endpoints for patient outcomes, we recommend increased emphasis on investigating the impact of the "soft" components constituting trial participation. Due to the interconnectedness of medical treatment and trial participation, we recommend further investigation in comparison to experiences in regular care. A deeper understanding of trial participation is needed to inform improvements for patient experiences and staff satisfaction alongside medical and scientific progress.
Collapse
Affiliation(s)
- Ronja Thallner
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Gumbinger
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anja Hohmann
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Antje Wick
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Loraine Busetto
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
- Institute for Medical Virology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Zhao B, Kilian M, Bunse T, Platten M, Bunse L. Tumor-reactive T helper cells in the context of vaccination against glioma. Cancer Cell 2023; 41:1829-1834. [PMID: 37863064 DOI: 10.1016/j.ccell.2023.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/11/2023] [Accepted: 09/26/2023] [Indexed: 10/22/2023]
Abstract
With the advances in immunogenomics, the majority of tumor-specific antigens were found to be recognized by T helper cells (THCs). This observation led to the development of long epitope vaccines in various cancers. Mechanistically, we are still gaining a deeper understanding of the mode of action of THCs as precision antitumor agonists. Here, we discuss the specific cellular mechanisms of THC functions in glioma immunology and contextualize current advances in anti-glioma vaccination exploiting THCs.
Collapse
Affiliation(s)
- Binghao Zhao
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany; German Cancer Consortium (DKTK), DKFZ, core center, Heidelberg, Germany
| | - Michael Kilian
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany; German Cancer Consortium (DKTK), DKFZ, core center, Heidelberg, Germany
| | - Theresa Bunse
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany; German Cancer Consortium (DKTK), DKFZ, core center, Heidelberg, Germany
| | - Michael Platten
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Germany; Helmholtz Institute of Translational Oncology Mainz (HI-TRON Mainz) - a Helmholtz Institute of the DKFZ, Mainz, Germany; German Cancer Consortium (DKTK), DKFZ, core center, Heidelberg, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg, a partnership between DKFZ and University Hospital, Heidelberg, Germany
| | - Lukas Bunse
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Germany; German Cancer Consortium (DKTK), DKFZ, core center, Heidelberg, Germany.
| |
Collapse
|
14
|
Jucht A, Dumont S, Pooley C, Gonzalez Castro LN. Cancer vaccine strategies for the treatment of diffusely infiltrating gliomas. Ther Adv Vaccines Immunother 2023; 11:25151355231206163. [PMID: 37886714 PMCID: PMC10599115 DOI: 10.1177/25151355231206163] [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: 02/24/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023] Open
Abstract
Diffusely infiltrating gliomas - including glioblastoma (GBM), isocitrate dehydrogenase (IDH) mutant gliomas, and histone 3 (H3) altered gliomas - are primary brain tumors with an invariably fatal outcome. Despite advances in the understanding of their biology, standard, targeted and immune checkpoint inhibitor immunotherapies have proven ineffective in arresting their inexorable progression and associated morbidity and mortality. Recognizing the unique aspects of the immunogenicity of cancer cells, the last decade has seen the development and evaluation of vaccine-based therapies for the treatment of solid tumors, including gliomas. Here we review the current vaccine strategies for the treatment of GBM, IDH-mutant gliomas and diffuse midline glioma H3 K27M-altered. We discuss potential benefits and challenges of vaccine therapies in these specific patient populations.
Collapse
|
15
|
Grassl N, Sahm K, Süße H, Poschke I, Bunse L, Bunse T, Boschert T, Mildenberger I, Rupp AK, Ewinger MP, Lanz LM, Denk M, Tabatabai G, Ronellenfitsch MW, Herrlinger U, Glas M, Krex D, Vajkoczy P, Wick A, Harting I, Sahm F, von Deimling A, Bendszus M, Wick W, Platten M. INTERCEPT H3: a multicenter phase I peptide vaccine trial for the treatment of H3-mutated diffuse midline gliomas. Neurol Res Pract 2023; 5:55. [PMID: 37853454 PMCID: PMC10585906 DOI: 10.1186/s42466-023-00282-4] [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: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Diffuse midline gliomas (DMG) are universally lethal central nervous system tumors that carry almost unanimously the clonal driver mutation histone-3 K27M (H3K27M). The single amino acid substitution of lysine to methionine harbors a neoantigen that is presented in tumor tissue. The long peptide vaccine H3K27M-vac targeting this major histocompatibility complex class II (MHC class II)-restricted neoantigen induces mutation-specific immune responses that suppress the growth of H3K27M+ flank tumors in an MHC-humanized rodent model. METHODS INTERCEPT H3 is a non-controlled open label, single arm, multicenter national phase 1 trial to assess safety, tolerability and immunogenicity of H3K27M-vac in combination with standard radiotherapy and the immune checkpoint inhibitor atezolizumab (ATE). 15 adult patients with newly diagnosed K27M-mutant histone-3.1 (H3.1K27M) or histone-3.3 (H3.3K27M) DMG will be enrolled in this trial. The 27mer peptide vaccine H3K27M-vac will be administered concomitantly to standard radiotherapy (RT) followed by combinatorial treatment with the programmed death-ligand 1 (PD-L1) targeting antibody ATE. The first three vaccines will be administered bi-weekly (q2w) followed by a dose at the beginning of recovery after RT and six-weekly administrations of doses 5 to 11 thereafter. In a safety lead-in, the first three patients (pts. 1-3) will be enrolled sequentially. PERSPECTIVE H3K27M-vac is a neoepitope targeting long peptide vaccine derived from the clonal driver mutation H3K27M in DMG. The INTERCEPT H3 trial aims at demonstrating (1) safety and (2) immunogenicity of repeated fixed dose vaccinations of H3K27M-vac administered with RT and ATE in adult patients with newly diagnosed H3K27M-mutant DMG. TRIAL REGISTRATION NCT04808245.
Collapse
Affiliation(s)
- Niklas Grassl
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
| | - Katharina Sahm
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
| | - Heike Süße
- National Center for Tumor Diseases (NCT) Trial Center, NCT, Heidelberg, Germany
| | - Isabel Poschke
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Immune Monitoring Unit, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
| | - Theresa Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
| | - Tamara Boschert
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Helmholtz Institute for Translational Oncology (HI-TRON), Mainz, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Iris Mildenberger
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
| | - Anne-Kathleen Rupp
- National Center for Tumor Diseases (NCT) Trial Center, NCT, Heidelberg, Germany
| | - Max Philipp Ewinger
- National Center for Tumor Diseases (NCT) Trial Center, NCT, Heidelberg, Germany
| | - Lisa-Marie Lanz
- National Center for Tumor Diseases (NCT) Trial Center, NCT, Heidelberg, Germany
| | - Monika Denk
- Department of Peptide-based Immunotherapy, University and University Hospital Tübingen, Tübingen, Germany
- Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- Partner site Tübingen, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Tübingen, Germany
| | - Ghazaleh Tabatabai
- Department of Neurology & Neuro-Oncology, University Hospital Tübingen, Tübingen, Germany
- Center for Neuro-Oncology, Comprehensive Cancer Center, University of Tübingen, Tübingen, Germany
| | - Michael W Ronellenfitsch
- Dr. Senckenberg Institute for Neurooncology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Ulrich Herrlinger
- Division of Clinical Neurooncology, University Hospital Bonn, Bonn, Germany
| | - Martin Glas
- Division of Clinical Neurooncology, Department of Neurology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS) and West German Cancer Center, Partner Site Essen, University Hospital Essen, German Cancer Consortium, University Duisburg-Essen, Essen, Germany
| | - Dietmar Krex
- Clinic and Polyclinic for Neurosurgery, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité Berlin, Berlin, Germany
| | - Antje Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Inga Harting
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, DKTK, DKFZ, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany.
- DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- National Center for Tumor Diseases (NCT) Trial Center, NCT, Heidelberg, Germany.
- Helmholtz Institute for Translational Oncology (HI-TRON) Mainz, German Cancer Research Center, INF 280, D69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Tardito S, MacKay C. Rethinking our approach to cancer metabolism to deliver patient benefit. Br J Cancer 2023; 129:406-415. [PMID: 37340094 PMCID: PMC10403540 DOI: 10.1038/s41416-023-02324-9] [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: 02/28/2023] [Revised: 05/25/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
Altered cellular metabolism is a major mechanism by which tumours support nutrient consumption associated with increased cellular proliferation. Selective dependency on specific metabolic pathways provides a therapeutic vulnerability that can be targeted in cancer therapy. Anti-metabolites have been used clinically since the 1940s and several agents targeting nucleotide metabolism are now well established as standard of care treatment in a range of indications. However, despite great progress in our understanding of the metabolic requirements of cancer and non-cancer cells within the tumour microenvironment, there has been limited clinical success for novel agents targeting pathways outside of nucleotide metabolism. We believe that there is significant therapeutic potential in targeting metabolic processes within cancer that is yet to be fully realised. However, current approaches to identify novel targets, test novel therapies and select patient populations most likely to benefit are sub-optimal. We highlight recent advances in technologies and understanding that will support the identification and validation of novel targets, re-evaluation of existing targets and design of optimal clinical positioning strategies to deliver patient benefit.
Collapse
Affiliation(s)
- Saverio Tardito
- The Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Craig MacKay
- Cancer Research Horizons, The Cancer Research UK Beatson Institute, Glasgow, UK.
| |
Collapse
|
17
|
Picca A, Bruno F, Nichelli L, Sanson M, Rudà R. Advances in molecular and imaging biomarkers in lower-grade gliomas. Expert Rev Neurother 2023; 23:1217-1231. [PMID: 37982735 DOI: 10.1080/14737175.2023.2285472] [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: 08/07/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
INTRODUCTION Lower-grade (grade 2-3) gliomas (LGGs) constitutes a group of primary brain tumors with variable clinical behaviors and treatment responses. Recent advancements in molecular biology have redefined their classification, and novel imaging modalities emerged for the noninvasive diagnosis and follow-up. AREAS COVERED This review comprehensively analyses the current knowledge on molecular and imaging biomarkers in LGGs. Key molecular alterations, such as IDH mutations and 1p/19q codeletion, are discussed for their prognostic and predictive implications in guiding treatment decisions. Moreover, the authors explore theranostic biomarkers for the potential of tailored therapies. Additionally, they also describe the utility of advanced imaging modalities, including widely available techniques, as dynamic susceptibility contrast perfusion-weighted imaging and less validated, emerging approaches, for the noninvasive LGGs characterization and follow-up. EXPERT OPINION The integration of molecular markers enhanced the stratification of LGGs, leading to the new concept of integrated histomolecular classification. While the IDH mutation is an established key prognostic and predictive marker, recent results from IDH inhibitors trials showed its potential value as a theranostic marker. In this setting, advanced MRI techniques such as 2-D-hydroxyglutarate spectroscopy are very promising for the noninvasive diagnosis and monitoring of LGGs. This progress offers exciting prospects for personalized medicine and improved treatment outcomes in LGGs.
Collapse
Affiliation(s)
- Alberto Picca
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Francesco Bruno
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| | - Lucia Nichelli
- Service de Neuroradiologie, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marc Sanson
- Service de Neurologie 2 Mazarin, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
- Sorbonne Université, Inserm, CNRS, UMRS1127, Institut du Cerveau-Paris Brain Institute-ICM, AP-HP, Paris, France
| | - Roberta Rudà
- Division of Neuro-Oncology, Department of Neuroscience "Rita Levi Montalcini", University and City of Health and Science University Hospital, Turin, Italy
| |
Collapse
|
18
|
Pu Y, Zhou G, Zhao K, Chen Y, Shen S. Immunotherapy for Recurrent Glioma-From Bench to Bedside. Cancers (Basel) 2023; 15:3421. [PMID: 37444531 DOI: 10.3390/cancers15133421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/12/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Glioma is the most aggressive malignant tumor of the central nervous system, and most patients suffer from a recurrence. Unfortunately, recurrent glioma often becomes resistant to established chemotherapy and radiotherapy treatments. Immunotherapy, a rapidly developing anti-tumor therapy, has shown a potential value in treating recurrent glioma. Multiple immune strategies have been explored. The most-used ones are immune checkpoint blockade (ICB) antibodies, which are barely effective in monotherapy. However, when combined with other immunotherapy, especially with anti-angiogenesis antibodies, ICB has shown encouraging efficacy and enhanced anti-tumor immune response. Oncolytic viruses and CAR-T therapies have shown promising results in recurrent glioma through multiple mechanisms. Vaccination strategies and immune-cell-based immunotherapies are promising in some subgroups of patients, and multiple new tumor antigenic targets have been discovered. In this review, we discuss current applicable immunotherapies and related mechanisms for recurrent glioma, focusing on multiple preclinical models and clinical trials in the last 5 years. Through reviewing the current combination of immune strategies, we would like to provide substantive thoughts for further novel therapeutic regimes treating recurrent glioma.
Collapse
Affiliation(s)
- Yi Pu
- Laboratory of Mitochondria and Metabolism, Department of Burn and Reconstructive Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guanyu Zhou
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kejia Zhao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shensi Shen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
19
|
Current Trends in Neoantigen-Based Cancer Vaccines. Pharmaceuticals (Basel) 2023; 16:ph16030392. [PMID: 36986491 PMCID: PMC10056833 DOI: 10.3390/ph16030392] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/18/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer immunotherapies are treatments that use drugs or cells to activate patients’ own immune systems against cancer cells. Among them, cancer vaccines have recently been rapidly developed. Based on tumor-specific antigens referred to as neoantigens, these vaccines can be in various forms such as messenger (m)RNA and synthetic peptides to activate cytotoxic T cells and act with or without dendritic cells. Growing evidence suggests that neoantigen-based cancer vaccines possess a very promising future, yet the processes of immune recognition and activation to relay identification of a neoantigen through the histocompatibility complex (MHC) and T-cell receptor (TCR) remain unclear. Here, we describe features of neoantigens and the biological process of validating neoantigens, along with a discussion of recent progress in the scientific development and clinical applications of neoantigen-based cancer vaccines.
Collapse
|
20
|
Kciuk M, Kołat D, Kałuzińska-Kołat Ż, Gawrysiak M, Drozda R, Celik I, Kontek R. PD-1/PD-L1 and DNA Damage Response in Cancer. Cells 2023; 12:530. [PMID: 36831197 PMCID: PMC9954559 DOI: 10.3390/cells12040530] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
The application of immunotherapy for cancer treatment is rapidly becoming more widespread. Immunotherapeutic agents are frequently combined with various types of treatments to obtain a more durable antitumor clinical response in patients who have developed resistance to monotherapy. Chemotherapeutic drugs that induce DNA damage and trigger DNA damage response (DDR) frequently induce an increase in the expression of the programmed death ligand-1 (PD-L1) that can be employed by cancer cells to avoid immune surveillance. PD-L1 exposed on cancer cells can in turn be targeted to re-establish the immune-reactive tumor microenvironment, which ultimately increases the tumor's susceptibility to combined therapies. Here we review the recent advances in how the DDR regulates PD-L1 expression and point out the effect of etoposide, irinotecan, and platinum compounds on the anti-tumor immune response.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Mateusz Gawrysiak
- Department of Immunology and Allergy, Medical University of Lodz, Pomorska 251, 92-213 Lodz, Poland
| | - Rafał Drozda
- Department of Gastrointestinal Endoscopy, Wl. Bieganski Hospital, 91-347 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| |
Collapse
|
21
|
Sharma N, Mallela AN, Shi DD, Tang LW, Abou-Al-Shaar H, Gersey ZC, Zhang X, McBrayer SK, Abdullah KG. Isocitrate dehydrogenase mutations in gliomas: A review of current understanding and trials. Neurooncol Adv 2023; 5:vdad053. [PMID: 37287696 PMCID: PMC10243983 DOI: 10.1093/noajnl/vdad053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Isocitrate dehydrogenase (IDH) is a key enzyme in normal metabolism and homeostasis. However, mutant forms of IDH are also defining features of a subset of diffuse gliomas. In this review, we highlight current techniques targeting IDH-mutated gliomas and summarize current and completed clinical trials exploring these strategies. We discuss clinical data from peptide vaccines, mutant IDH (mIDH) inhibitors, and PARP inhibitors. Peptide vaccines have the unique advantage of targeting the specific epitope of a patient's tumor, inducing a highly tumor-specific CD4+ T-cell response. mIDH-inhibitors, on the other hand, specifically target mutant IDH proteins in cancer cell metabolism and thus help halt gliomagenesis. We also explore PARP inhibitors and their role in treating diffuse gliomas, which exploit IDH-mutant diffuse gliomas by allowing the persistence of unrepaired DNA complexes. We summarize various completed and current trials targeting IDH1 and IDH2 mutations in diffuse gliomas. Therapies targeting mutant IDH have significant promise in treating progressive or recurrent IDH-mutant gliomas and may significantly change treatment paradigms in the next decade.
Collapse
Affiliation(s)
- Nikhil Sharma
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Arka N Mallela
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diana D Shi
- Harvard Radiation Oncology Program, Harvard Medical School, Boston, Massachusetts, USA
| | - Lilly W Tang
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Hussam Abou-Al-Shaar
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zachary C Gersey
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Xiaoran Zhang
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Samuel K McBrayer
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
22
|
Kessler T, Ito J, Wick W, Wick A. Conventional and emerging treatments of astrocytomas and oligodendrogliomas. J Neurooncol 2022; 162:471-478. [PMID: 36566461 DOI: 10.1007/s11060-022-04216-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/13/2022] [Indexed: 12/26/2022]
Abstract
PURPOSE Astrocytomas and oligodendrogliomas are mainly diffuse primary brain tumors harboring a diagnostic and prognostically favorable isocitrate dehydrogenase mutation. They are still incurable besides growing molecular knowledge and therapy options. Circumscribed astrocytomas are also discussed here, although they represent a separate entity despite similarities in the nomenclature. METHODS We reviewed clinical trials, preclinical approaches as well as guideline recommendations form the major scientific Neuro-Oncology organizations for astrocytomas and oligodendrogliomas according to PRISMA guidelines. RESULTS After histopathological diagnosis and eventually a maximal safe resection, patients with good prognostic factors may be followed by magnetic resonance imaging (MRI). If further treatment is necessary, either after diagnosis or at progression, diffuse astrocytomas and oligodendrogliomas are mainly treated with combined radiochemotherapy or maximal safe resection followed by combined radiochemotherapy according to current guidelines based on randomized trials. Circumscribed gliomas like pilocytic astrocytomas, CNS WHO grade 1, or pleomorphic xanthoastrocytomas, CNS WHO grade 2, are often treated with surgery alone. Current approaches for therapy optimization include decision of the best chemotherapy regimen. The IDH mutation presents a rational target for small molecule inhibition and immune therapy in diffuse astrocytomas and oligodendrogliomas, while the BRAF pathway is frequently mutated and treatable in circumscribed gliomas. CONCLUSION Despite establishment of standard treatment approaches for gliomas that include resection, radio- and chemotherapy, there is a lack of effective treatments for progressive disease. Immune- and targeted therapies are currently investigated.
Collapse
Affiliation(s)
- Tobias Kessler
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany.
- Neurology Clinic and Neurooncology Program, National Center for Tumor Diseases & DKTK, DKFZ, Im Neuenheimer Feld 400, D-69120, Heidelberg, Germany.
| | - Jakob Ito
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang Wick
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Antje Wick
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
23
|
Wu F, Yin YY, Fan WH, Zhai Y, Yu MC, Wang D, Pan CQ, Zhao Z, Li GZ, Zhang W. Immunological profiles of human oligodendrogliomas define two distinct molecular subtypes. EBioMedicine 2022; 87:104410. [PMID: 36525723 PMCID: PMC9772571 DOI: 10.1016/j.ebiom.2022.104410] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human oligodendroglioma presents as a heterogeneous disease, primarily characterized by the isocitrate dehydrogenase (IDH) mutation and 1p/19q co-deletion. Therapy development for this tumor is hindered by incomplete knowledge of somatic driving alterations and suboptimal disease classification. We herein aim to identify intrinsic molecular subtypes through integrated analysis of transcriptome, genome and methylome. METHODS 137 oligodendroglioma patients from the Cancer Genome Atlas (TCGA) dataset were collected for unsupervised clustering analysis of immune gene expression profiles and comparative analysis of genome and methylome. Two independent datasets containing 218 patients were used for validation. FINDINGS We identified and independently validated two reproducible subtypes associated with distinct molecular characteristics and clinical outcomes. The proliferative subtype, named Oligo1, was characterized by more tumors of CNS WHO grade 3, as well as worse prognosis compared to the Oligo2 subtype. Besides the clinicopathologic features, Oligo1 exhibited enrichment of cell proliferation, regulation of cell cycle and Wnt signaling pathways, and significantly altered genes, such as EGFR, NOTCH1 and MET. In contrast, Oligo2, with favorable outcome, presented increased activation of immune response and metabolic process. Higher T cell/APC co-inhibition and inhibitory checkpoint levels were observed in Oligo2 tumors. Finally, multivariable analysis revealed our classification was an independent prognostic factor in oligodendrogliomas, and the robustness of these molecular subgroups was verified in the validation cohorts. INTERPRETATION This study provides further insights into patient stratification as well as presents opportunities for therapeutic development in human oligodendrogliomas. FUNDING The funders are listed in the Acknowledgement.
Collapse
Affiliation(s)
- Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China,Corresponding author. Nan Si Huan Xi Lu 119, Fengtai District, Beijing 100070, China.
| | - Yi-Yun Yin
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Wen-Hua Fan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Ming-Chen Yu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Di Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Chang-Qing Pan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Guan-Zhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, 100070, China,Corresponding author. Nan Si Huan Xi Lu 119, Fengtai District, Beijing 100070, China.
| |
Collapse
|
24
|
Bunse L, Bunse T, Krämer C, Chih YC, Platten M. Clinical and Translational Advances in Glioma Immunotherapy. Neurotherapeutics 2022; 19:1799-1817. [PMID: 36303101 PMCID: PMC9723056 DOI: 10.1007/s13311-022-01313-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 02/06/2023] Open
Abstract
Gliomas are highly treatment refractory against immune checkpoint blockade, an immunotherapeutic modality that revolutionized therapy for many tumors. At the same time, technological innovation has dramatically accelerated the development of immunotherapeutic approaches such as personalized tumor-specific vaccine production, dendritic cell vaccine manufacture, patient-individual target selection and chimeric antigen receptor, and T cell receptor T cell manufacture. Here we review recent clinical and translational advances in glioma immunotherapy with a focus on targets and their cognate immune receptor derivates as well as concepts to improve intratumoral T cell effector functions.
Collapse
Affiliation(s)
- Lukas Bunse
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
| | - Theresa Bunse
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany
| | - Christopher Krämer
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yu-Chan Chih
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Michael Platten
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, MCTN, Heidelberg University, Mannheim, Germany.
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg, Germany.
- Helmholtz Institute of Translational Oncology (HI-TRON), Mainz, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|