1
|
Regmi M, Wang Y, Liu W, Dai Y, Liu S, Ma K, Lin G, Yang J, Liu H, Wu J, Yang C. From glioma gloom to immune bloom: unveiling novel immunotherapeutic paradigms-a review. J Exp Clin Cancer Res 2024; 43:47. [PMID: 38342925 PMCID: PMC10860318 DOI: 10.1186/s13046-024-02973-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/04/2024] [Indexed: 02/13/2024] Open
Abstract
In tumor therapeutics, the transition from conventional cytotoxic drugs to targeted molecular therapies, such as those targeting receptor tyrosine kinases, has been pivotal. Despite this progress, the clinical outcomes have remained modest, with glioblastoma patients' median survival stagnating at less than 15 months. This underscores the urgent need for more specialized treatment strategies. Our review delves into the progression toward immunomodulation in glioma treatment. We dissect critical discoveries in immunotherapy, such as spotlighting the instrumental role of tumor-associated macrophages, which account for approximately half of the immune cells in the glioma microenvironment, and myeloid-derived suppressor cells. The complex interplay between tumor cells and the immune microenvironment has been explored, revealing novel therapeutic targets. The uniqueness of our review is its exhaustive approach, synthesizing current research to elucidate the intricate roles of various molecules and receptors within the glioma microenvironment. This comprehensive synthesis not only maps the current landscape but also provides a blueprint for refining immunotherapy for glioma, signifying a paradigm shift toward leveraging immune mechanisms for improved patient prognosis.
Collapse
Affiliation(s)
- Moksada Regmi
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China
| | - Yingjie Wang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Weihai Liu
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Yuwei Dai
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Shikun Liu
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
- Peking University Health Science Center, Beijing, 100191, China
| | - Ke Ma
- Peking University Health Science Center, Beijing, 100191, China
| | - Guozhong Lin
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Hongyi Liu
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China
- National Engineering Research Center for Ophthalmology, Beijing, 100730, China
- Engineering Research Center of Ophthalmic Equipment and Materials, Ministry of Education, Beijing, 100730, China
- Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100730, China
| | - Jian Wu
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China.
- National Engineering Research Center for Ophthalmology, Beijing, 100730, China.
- Engineering Research Center of Ophthalmic Equipment and Materials, Ministry of Education, Beijing, 100730, China.
- Beijing Key Laboratory of Ophthalmology and Visual Sciences, Beijing, 100730, China.
| | - Chenlong Yang
- Department of Neurosurgery, Peking University Third Hospital, Peking University, Beijing, 100191, China.
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Peking University, Beijing, 100191, China.
- Henan Academy of Innovations in Medical Science (AIMS), Zhengzhou, 450003, China.
| |
Collapse
|
2
|
Gubser C, Pascoe RD, Chang J, Chiu C, Solomon A, Cao R, Rasmussen TA, Lewin SR. GITR activation ex vivo impairs CD8 T cell function in people with HIV on antiretroviral therapy. iScience 2023; 26:108165. [PMID: 38026168 PMCID: PMC10660494 DOI: 10.1016/j.isci.2023.108165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/15/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Glucocorticoid-induced tumor necrosis factor related protein (GITR) is a co-stimulatory immune checkpoint molecule constitutively expressed on regulatory T cells (Tregs) and on activated T conventional cells (Tconv). In blood collected from PWH on suppressive ART, GITR expression was reduced in multiple activated CD4 and CD8 T cell subsets but was increased in Tregs. HIV specific CD8 T cells expressed higher levels of GITR and programmed cell death protein 1 (PD-1) compared to total CD8 T cells. Following stimulation with HIV peptides and GITR-ligand (L), we demonstrated a significant decrease in killing by HIV specific CD8 T cells and an increased exhausted profile. T cell receptor co-stimulation with GITR-L abrogated Treg suppression and induced expansion of CD4 Tconv. We conclude that GITR activation is an additional factor contributing to an impaired HIV immune response in PWH on ART and that GITR agonist antibodies should not be pursued for HIV cure strategies.
Collapse
Affiliation(s)
- Céline Gubser
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Rachel D. Pascoe
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Judy Chang
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Chris Chiu
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Ajantha Solomon
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Rosalyn Cao
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thomas A. Rasmussen
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
4
|
Huang X, Shi S, Wang H, Zhao T, Wang Y, Huang S, Su Y, Zhao C, Yang M. Advances in antibody-based drugs and their delivery through the blood-brain barrier for targeted therapy and immunotherapy of gliomas. Int Immunopharmacol 2023; 117:109990. [PMID: 37012874 DOI: 10.1016/j.intimp.2023.109990] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023]
Abstract
Gliomas are highly invasive and are the most common type of primary malignant brain tumor. The routine treatments for glioma include surgical resection, radiotherapy, and chemotherapy. However, glioma recurrence and patient survival remain unsatisfactory after employing these traditional treatment approaches. With the rapid development of molecular immunology, significant breakthroughs have been made in targeted glioma therapy and immunotherapy. Antibody-based therapy has excellent advantages in treating gliomas due to its high specificity and sensitivity. This article reviewed various targeted antibody drugs for gliomas, including anti-glioma surface marker antibodies, anti-angiogenesis antibodies, and anti-immunosuppressive signal antibodies. Notably, many antibodies have been validated clinically, such as bevacizumab, cetuximab, panitumumab, and anti-PD-1 antibodies. These antibodies can improve the targeting of glioma therapy, enhance anti-tumor immunity, reduce the proliferation and invasion of glioma, and thus prolong the survival time of patients. However, the existence of the blood-brain barrier (BBB) has caused significant difficulties in drug delivery for gliomas. Therefore, this paper also summarized drug delivery methods through the BBB, including receptor-mediated transportation, nano-based carriers, and some physical and chemical methods for drug delivery. With these exciting advancements, more antibody-based therapies will likely enter clinical practice and allow more successful control of malignant gliomas.
Collapse
Affiliation(s)
- Xin Huang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Shuyou Shi
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Hongrui Wang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Tiesuo Zhao
- Department of Immunology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yibo Wang
- The College of Clinical College, Jilin University, Changchun, China
| | - Sihua Huang
- The College of Clinical College, Jilin University, Changchun, China
| | - Yingying Su
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Chunyan Zhao
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China.
| | - Ming Yang
- College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China.
| |
Collapse
|
5
|
Bausart M, Bozzato E, Joudiou N, Koutsoumpou X, Manshian B, Préat V, Gallez B. Mismatch between Bioluminescence Imaging (BLI) and MRI When Evaluating Glioblastoma Growth: Lessons from a Study Where BLI Suggested "Regression" while MRI Showed "Progression". Cancers (Basel) 2023; 15:cancers15061919. [PMID: 36980804 PMCID: PMC10047859 DOI: 10.3390/cancers15061919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Orthotopic glioblastoma xenografts are paramount for evaluating the effect of innovative anti-cancer treatments. In longitudinal studies, tumor growth (or regression) of glioblastoma can only be monitored by noninvasive imaging. For this purpose, bioluminescence imaging (BLI) has gained popularity because of its low cost and easy access. In the context of the development of new nanomedicines for treating glioblastoma, we were using luciferase-expressing GL261 cell lines. Incidentally, using BLI in a specific GL261 glioblastoma model with cells expressing both luciferase and the green fluorescent protein (GL261-luc-GFP), we observed an apparent spontaneous regression. By contrast, the magnetic resonance imaging (MRI) analysis revealed that the tumors were actually growing over time. For other models (GL261 expressing only luciferase and U87 expressing both luciferase and GFP), data from BLI and MRI correlated well. We found that the divergence in results coming from different imaging modalities was not due to the tumor localization nor the penetration depth of light but was rather linked to the instability in luciferase expression in the viral construct used for the GL261-luc-GFP model. In conclusion, the use of multi-modality imaging prevents possible errors in tumor growth evaluation, and checking the stability of luciferase expression is mandatory when using BLI as the sole imaging modality.
Collapse
Affiliation(s)
- Mathilde Bausart
- Advanced Drug Delivery and Biomaterials (ADDB) Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Elia Bozzato
- Advanced Drug Delivery and Biomaterials (ADDB) Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Nicolas Joudiou
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Xanthippi Koutsoumpou
- Department of Imaging and Pathology, Translational Cell and Tissue Research Unit, Katholiek Universiteit Leuven (KULeuven), 3000 Leuven, Belgium
| | - Bella Manshian
- Department of Imaging and Pathology, Translational Cell and Tissue Research Unit, Katholiek Universiteit Leuven (KULeuven), 3000 Leuven, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials (ADDB) Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance (REMA) Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
6
|
Awada H, Paris F, Pecqueur C. Exploiting radiation immunostimulatory effects to improve glioblastoma outcome. Neuro Oncol 2023; 25:433-446. [PMID: 36239313 PMCID: PMC10013704 DOI: 10.1093/neuonc/noac239] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer treatment protocols depend on tumor type, localization, grade, and patient. Despite aggressive treatments, median survival of patients with Glioblastoma (GBM), the most common primary brain tumor in adults, does not exceed 18 months, and all patients eventually relapse. Thus, novel therapeutic approaches are urgently needed. Radiotherapy (RT) induces a multitude of alterations within the tumor ecosystem, ultimately modifying the degree of tumor immunogenicity at GBM relapse. The present manuscript reviews the diverse effects of RT radiotherapy on tumors, with a special focus on its immunomodulatory impact to finally discuss how RT could be exploited in GBM treatment through immunotherapy targeting. Indeed, while further experimental and clinical studies are definitively required to successfully translate preclinical results in clinical trials, current studies highlight the therapeutic potential of immunotherapy to uncover novel avenues to fight GBM.
Collapse
Affiliation(s)
- Hala Awada
- Nantes Université, CRCI2NA, INSERM, CNRS, F-44000 Nantes, France.,Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - François Paris
- Nantes Université, CRCI2NA, INSERM, CNRS, F-44000 Nantes, France.,Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Claire Pecqueur
- Nantes Université, CRCI2NA, INSERM, CNRS, F-44000 Nantes, France
| |
Collapse
|
7
|
Cao TQ, Wainwright DA, Lee-Chang C, Miska J, Sonabend AM, Heimberger AB, Lukas RV. Next Steps for Immunotherapy in Glioblastoma. Cancers (Basel) 2022; 14:4023. [PMID: 36011015 PMCID: PMC9406905 DOI: 10.3390/cancers14164023] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Outcomes for glioblastoma (GBM) patients undergoing standard of care treatment remain poor. Here we discuss the portfolio of previously investigated immunotherapies for glioblastoma, including vaccine therapy and checkpoint inhibitors, as well as novel emerging therapeutic approaches. In addition, we explore the factors that potentially influence response to immunotherapy, which should be considered in future research aimed at improving immunotherapy efficacy.
Collapse
Affiliation(s)
- Toni Q. Cao
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Derek A. Wainwright
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
- Department of Medicine, Division of Hematology/Oncology, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Department of Microbiology-Immunology, Northwestern University, Chicago, IL 60611, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| | - Rimas V. Lukas
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
- Lou & Jean Malnati Brain Tumor Institute, Chicago, IL 60611, USA
| |
Collapse
|
8
|
Gao R, Shi GP, Wang J. Functional Diversities of Regulatory T Cells in the Context of Cancer Immunotherapy. Front Immunol 2022; 13:833667. [PMID: 35371055 PMCID: PMC8969660 DOI: 10.3389/fimmu.2022.833667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/28/2022] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are a subset of CD4+ T cells with their immunosuppressive activities to block abnormal or excessive immune responses to self and non-autoantigens. Tregs express the transcription factor Foxp3, maintain the immune homeostasis, and prevent the initiation of anti-tumor immune effects in various ways as their mechanisms to modulate tumor development. Recognition of different phenotypes and functions of intratumoral Tregs has offered the possibilities to develop therapeutic strategies by selectively targeting Tregs in cancers with the aim of alleviating their immunosuppressive activities from anti-tumor immune responses. Several Treg-based immunotherapeutic approaches have emerged to target cytotoxic T lymphocyte antigen-4, glucocorticoid-induced tumor necrosis factor receptor, CD25, indoleamine-2, 3-dioxygenase-1, and cytokines. These immunotherapies have yielded encouraging outcomes from preclinical studies and early-phase clinical trials. Further, dual therapy or combined therapy has been approved to be better choices than single immunotherapy, radiotherapy, or chemotherapy. In this short review article, we discuss our current understanding of the immunologic characteristics of Tregs, including Treg differentiation, development, therapeutic efficacy, and future potential of Treg-related therapies among the general cancer therapy.
Collapse
Affiliation(s)
- Ran Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Bausart M, Préat V, Malfanti A. Immunotherapy for glioblastoma: the promise of combination strategies. J Exp Clin Cancer Res 2022; 41:35. [PMID: 35078492 PMCID: PMC8787896 DOI: 10.1186/s13046-022-02251-2] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) treatment has remained almost unchanged for more than 20 years. The current standard of care involves surgical resection (if possible) followed by concomitant radiotherapy and chemotherapy. In recent years, immunotherapy strategies have revolutionized the treatment of many cancers, increasing the hope for GBM therapy. However, mostly due to the high, multifactorial immunosuppression occurring in the microenvironment, the poor knowledge of the neuroimmune system and the presence of the blood-brain barrier, the efficacy of immunotherapy in GBM is still low. Recently, new strategies for GBM treatments have employed immunotherapy combinations and have provided encouraging results in both preclinical and clinical studies. The lessons learned from clinical trials highlight the importance of tackling different arms of immunity. In this review, we aim to summarize the preclinical evidence regarding combination immunotherapy in terms of immune and survival benefits for GBM management. The outcomes of recent studies assessing the combination of different classes of immunotherapeutic agents (e.g., immune checkpoint blockade and vaccines) will be discussed. Finally, future strategies to ameliorate the efficacy of immunotherapy and facilitate clinical translation will be provided to address the unmet medical needs of GBM.
Collapse
Affiliation(s)
- Mathilde Bausart
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| |
Collapse
|
10
|
Wang EJ, Chen JS, Jain S, Morshed RA, Haddad AF, Gill S, Beniwal AS, Aghi MK. Immunotherapy Resistance in Glioblastoma. Front Genet 2021; 12:750675. [PMID: 34976006 PMCID: PMC8718605 DOI: 10.3389/fgene.2021.750675] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults. Despite treatment consisting of surgical resection followed by radiotherapy and adjuvant chemotherapy, survival remains poor at a rate of 26.5% at 2 years. Recent successes in using immunotherapies to treat a number of solid and hematologic cancers have led to a growing interest in harnessing the immune system to target glioblastoma. Several studies have examined the efficacy of various immunotherapies, including checkpoint inhibitors, vaccines, adoptive transfer of lymphocytes, and oncolytic virotherapy in both pre-clinical and clinical settings. However, these therapies have yielded mixed results at best when applied to glioblastoma. While the initial failures of immunotherapy were thought to reflect the immunoprivileged environment of the brain, more recent studies have revealed immune escape mechanisms created by the tumor itself and adaptive resistance acquired in response to therapy. Several of these resistance mechanisms hijack key signaling pathways within the immune system to create a protumoral microenvironment. In this review, we discuss immunotherapies that have been trialed in glioblastoma, mechanisms of tumor resistance, and strategies to sensitize these tumors to immunotherapies. Insights gained from the studies summarized here may help pave the way for novel therapies to overcome barriers that have thus far limited the success of immunotherapy in glioblastoma.
Collapse
Affiliation(s)
- Elaina J. Wang
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Jia-Shu Chen
- Department of Neurological Surgery, The Warren Alpert School of Medicine, Brown University, Providence, RI, United States
| | - Saket Jain
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Ramin A. Morshed
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander F. Haddad
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Sabraj Gill
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Angad S. Beniwal
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Manish K. Aghi
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
11
|
Sharpe MA, Baskin DS, Jenson AV, Baskin AM. Hijacking Sexual Immuno-Privilege in GBM-An Immuno-Evasion Strategy. Int J Mol Sci 2021; 22:10983. [PMID: 34681642 PMCID: PMC8536168 DOI: 10.3390/ijms222010983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/28/2021] [Accepted: 10/05/2021] [Indexed: 01/12/2023] Open
Abstract
Regulatory T-cells (Tregs) are immunosuppressive T-cells, which arrest immune responses to 'Self' tissues. Some immunosuppressive Tregs that recognize seminal epitopes suppress immune responses to the proteins in semen, in both men and women. We postulated that GBMs express reproductive-associated proteins to manipulate reproductive Tregs and to gain immune privilege. We analyzed four GBM transcriptome databases representing ≈900 tumors for hypoxia-responsive Tregs, steroidogenic pathways, and sperm/testicular and placenta-specific genes, stratifying tumors by expression. In silico analysis suggested that the presence of reproductive-associated Tregs in GBM tumors was associated with worse patient outcomes. These tumors have an androgenic signature, express male-specific antigens, and attract reproductive-associated Related Orphan Receptor C (RORC)-Treg immunosuppressive cells. GBM patient sera were interrogated for the presence of anti-sperm/testicular antibodies, along with age-matched controls, utilizing monkey testicle sections. GBM patient serum contained anti-sperm/testicular antibodies at levels > six-fold that of controls. Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are associated with estrogenic tumors which appear to mimic placental tissue. We demonstrate that RORC-Tregs drive poor patient outcome, and Treg infiltration correlates strongly with androgen levels. Androgens support GBM expression of sperm/testicular proteins allowing Tregs from the patient's reproductive system to infiltrate the tumor. In contrast, estrogen appears responsible for MDSC/TAM immunosuppression.
Collapse
MESH Headings
- Androgens/metabolism
- Brain Neoplasms/immunology
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Databases, Factual
- Estrogens/metabolism
- Female
- Glioblastoma/immunology
- Glioblastoma/mortality
- Glioblastoma/pathology
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kaplan-Meier Estimate
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Microglia/immunology
- Microglia/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Microenvironment
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Amanda V. Jenson
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| | - Alexandra M. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA; (D.S.B.); (A.V.J.); (A.M.B.)
| |
Collapse
|
12
|
Vázquez Cervantes GI, González Esquivel DF, Gómez-Manzo S, Pineda B, Pérez de la Cruz V. New Immunotherapeutic Approaches for Glioblastoma. J Immunol Res 2021; 2021:3412906. [PMID: 34557553 PMCID: PMC8455182 DOI: 10.1155/2021/3412906] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor with a high mortality rate. The current treatment consists of surgical resection, radiation, and chemotherapy; however, the median survival rate is only 12-18 months despite these alternatives, highlighting the urgent need to find new strategies. The heterogeneity of GBM makes this tumor difficult to treat, and the immunotherapies result in an attractive approach to modulate the antitumoral immune responses favoring the tumor eradication. The immunotherapies for GMB including monoclonal antibodies, checkpoint inhibitors, vaccines, and oncolytic viruses, among others, have shown favorable results alone or as a multimodal treatment. In this review, we summarize and discuss promising immunotherapies for GBM currently under preclinical investigation as well as in clinical trials.
Collapse
Affiliation(s)
- Gustavo Ignacio Vázquez Cervantes
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio A, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, C.P. 04510 Distrito Federal, Mexico
| | - Dinora F. González Esquivel
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, México City 04530, Mexico
| | - Benjamín Pineda
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Verónica Pérez de la Cruz
- Neurochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| |
Collapse
|
13
|
Kelly WJ, Giles AJ, Gilbert M. T lymphocyte-targeted immune checkpoint modulation in glioma. J Immunother Cancer 2021; 8:jitc-2019-000379. [PMID: 32051289 PMCID: PMC7057419 DOI: 10.1136/jitc-2019-000379] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
Immunomodulatory therapies targeting inhibitory checkpoint molecules have revolutionized the treatment of solid tumor malignancies. Concerns about whether systemic administration of an immune checkpoint inhibitor could impact primary brain tumors were answered with the observation of definitive responses in pediatric patients harboring hypermutated gliomas. Although initial clinical results in patients with glioblastoma (GBM) were disappointing, recently published results have demonstrated a potential survival benefit in patients with recurrent GBM treated with neoadjuvant programmed cell death protein 1 blockade. While these findings necessitate verification in subsequent studies, they support the possibility of achieving clinical meaningful immune responses in malignant primary brain tumors including GBM, a disease in dire need of additional therapeutic options. There are several challenges involved in treating glioma with immune checkpoint modulators including the immunosuppressive nature of GBM itself with high inhibitory checkpoint expression, the immunoselective blood brain barrier impairing the ability for peripheral lymphocytes to traffic to the tumor microenvironment and the high prevalence of corticosteroid use which suppress lymphocyte activation. However, by simultaneously targeting multiple costimulatory and inhibitory pathways, it may be possible to achieve an effective antitumoral immune response. To this end, there are now several novel agents targeting more recently uncovered “second generation” checkpoint molecules. Given the multiplicity of drugs being considered for combination regimens, an increased understanding of the mechanisms of action and resistance combined with more robust preclinical and early clinical testing will be needed to be able to adequately test these agents. This review summarizes our current understanding of T lymphocyte-modulating checkpoint molecules as it pertains to glioma with the hope for a renewed focus on the most promising therapeutic strategies.
Collapse
Affiliation(s)
| | - Amber Jin Giles
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Karachi A, Dastmalchi F, Nazarian S, Huang J, Sayour EJ, Jin L, Yang C, Mitchell DA, Rahman M. Optimizing T Cell-Based Therapy for Glioblastoma. Front Immunol 2021; 12:705580. [PMID: 34421912 PMCID: PMC8374079 DOI: 10.3389/fimmu.2021.705580] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/20/2021] [Indexed: 11/30/2022] Open
Abstract
Evading T cell surveillance is a hallmark of cancer. Patients with solid tissue malignancy, such as glioblastoma (GBM), have multiple forms of immune dysfunction, including defective T cell function. T cell dysfunction is exacerbated by standard treatment strategies such as steroids, chemotherapy, and radiation. Reinvigoration of T cell responses can be achieved by utilizing adoptively transferred T cells, including CAR T cells. However, these cells are at risk for depletion and dysfunction as well. This review will discuss adoptive T cell transfer strategies and methods to avoid T cell dysfunction for the treatment of brain cancer.
Collapse
Affiliation(s)
- Aida Karachi
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Farhad Dastmalchi
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Saina Nazarian
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Jianping Huang
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Elias J Sayour
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Linchun Jin
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Changlin Yang
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| | - Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, University of Florida (UF) Brain Tumor Immunotherapy Program, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Desbaillets N, Hottinger AF. Immunotherapy in Glioblastoma: A Clinical Perspective. Cancers (Basel) 2021; 13:3721. [PMID: 34359621 PMCID: PMC8345081 DOI: 10.3390/cancers13153721] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most frequent and the most aggressive brain tumor. It is notoriously resistant to current treatments, and the prognosis remains dismal. Immunotherapies have revolutionized the treatment of numerous cancer types and generate great hope for glioblastoma, alas without success until now. In this review, the rationale underlying immune targeting of glioblastoma, as well as the challenges faced when targeting these highly immunosuppressive tumors, are discussed. Innovative immune-targeting strategies including cancer vaccines, oncolytic viruses, checkpoint blockade inhibitors, adoptive cell transfer, and CAR T cells that have been investigated in glioblastoma are reviewed. From a clinical perspective, key clinical trial findings and ongoing trials are discussed for each approach. Finally, limitations, either biological or arising from trial designs are analyzed, and strategies to overcome them are presented. Proof of efficacy for immunotherapy approaches remains to be demonstrated in glioblastoma, but our rapidly expanding understanding of its biology, its immune microenvironment, and the emergence of novel promising combinatorial approaches might allow researchers to finally fulfill the medical need for GBM patients.
Collapse
Affiliation(s)
- Nicolas Desbaillets
- Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois & Université de Lausanne, 1011 Lausanne, Switzerland;
| | - Andreas Felix Hottinger
- Department of Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois & Université de Lausanne, 1011 Lausanne, Switzerland;
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| |
Collapse
|
16
|
De Martino M, Padilla O, Daviaud C, Wu CC, Gartrell RD, Vanpouille-Box C. Exploiting Radiation Therapy to Restore Immune Reactivity of Glioblastoma. Front Oncol 2021; 11:671044. [PMID: 34094969 PMCID: PMC8173136 DOI: 10.3389/fonc.2021.671044] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is among the most aggressive of brain tumors and confers a dismal prognosis despite advances in surgical technique, radiation delivery methods, chemotherapy, and tumor-treating fields. While immunotherapy (IT) has improved the care of several adult cancers with previously dismal prognoses, monotherapy with IT in GBM has shown minimal response in first recurrence. Recent discoveries in lymphatics and evaluation of blood brain barrier offer insight to improve the use of ITs and determine the best combinations of therapies, including radiation. We highlight important features of the tumor immune microenvironment in GBM and potential for combining radiation and immunotherapy to improve prognosis in this devastating disease.
Collapse
Affiliation(s)
- Mara De Martino
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Oscar Padilla
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY, United States
| | - Camille Daviaud
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States
| | - Cheng-Chia Wu
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY, United States.,Herbert Irving Comprehensive Cancer Center, New York, NY, United States
| | - Robyn D Gartrell
- Department of Pediatrics, Pediatric Hematology/Oncology/SCT, Columbia University Irving Medical Center, New York, NY, United States
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, United States.,Sandra and Edward Meyer Cancer Center, New York, NY, United States
| |
Collapse
|
17
|
Amoozgar Z, Kloepper J, Ren J, Tay RE, Kazer SW, Kiner E, Krishnan S, Posada JM, Ghosh M, Mamessier E, Wong C, Ferraro GB, Batista A, Wang N, Badeaux M, Roberge S, Xu L, Huang P, Shalek AK, Fukumura D, Kim HJ, Jain RK. Targeting Treg cells with GITR activation alleviates resistance to immunotherapy in murine glioblastomas. Nat Commun 2021; 12:2582. [PMID: 33976133 PMCID: PMC8113440 DOI: 10.1038/s41467-021-22885-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint blockers (ICBs) have failed in all phase III glioblastoma (GBM) trials. Here, we show that regulatory T (Treg) cells play a key role in GBM resistance to ICBs in experimental gliomas. Targeting glucocorticoid-induced TNFR-related receptor (GITR) in Treg cells using an agonistic antibody (αGITR) promotes CD4 Treg cell differentiation into CD4 effector T cells, alleviates Treg cell-mediated suppression of anti-tumor immune response, and induces potent anti-tumor effector cells in GBM. The reprogrammed GBM-infiltrating Treg cells express genes associated with a Th1 response signature, produce IFNγ, and acquire cytotoxic activity against GBM tumor cells while losing their suppressive function. αGITR and αPD1 antibodies increase survival benefit in three experimental GBM models, with a fraction of cohorts exhibiting complete tumor eradication and immune memory upon tumor re-challenge. Moreover, αGITR and αPD1 synergize with the standard of care treatment for newly-diagnosed GBM, enhancing the cure rates in these GBM models.
Collapse
Affiliation(s)
- Zohreh Amoozgar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Jonas Kloepper
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Jun Ren
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Rong En Tay
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute (DFCI) and Harvard Medical School, Boston, MA, USA
| | - Samuel W Kazer
- Department of Chemistry, Institute for Medical Engineering & Science, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Ragon Institute of MGH, MIT & Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Evgeny Kiner
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Shanmugarajan Krishnan
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Jessica M Posada
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Mitrajit Ghosh
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Emilie Mamessier
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Christina Wong
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Gino B Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Ana Batista
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Nancy Wang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Mark Badeaux
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Sylvie Roberge
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Lei Xu
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Peigen Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Alex K Shalek
- Department of Chemistry, Institute for Medical Engineering & Science, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Ragon Institute of MGH, MIT & Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Dai Fukumura
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA
| | - Hye-Jung Kim
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute (DFCI) and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH) and Harvard Medical School (HMS), Boston, MA, USA.
| |
Collapse
|
18
|
Marciscano AE, Haimovitz-Friedman A, Lee P, Tran PT, Tomé WA, Guha C, (Spring) Kong FM, Sahgal A, El Naqa I, Rimner A, Marks LB, Formenti SC, DeWeese TL. Immunomodulatory Effects of Stereotactic Body Radiation Therapy: Preclinical Insights and Clinical Opportunities. Int J Radiat Oncol Biol Phys 2021; 110:35-52. [DOI: 10.1016/j.ijrobp.2019.02.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 02/11/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
|
19
|
de Groot J, Penas-Prado M, Alfaro-Munoz K, Hunter K, Pei BL, O'Brien B, Weathers SP, Loghin M, Kamiya Matsouka C, Yung WKA, Mandel J, Wu J, Yuan Y, Zhou S, Fuller GN, Huse J, Rao G, Weinberg JS, Prabhu SS, McCutcheon IE, Lang FF, Ferguson SD, Sawaya R, Colen R, Yadav SS, Blando J, Vence L, Allison J, Sharma P, Heimberger AB. Window-of-opportunity clinical trial of pembrolizumab in patients with recurrent glioblastoma reveals predominance of immune-suppressive macrophages. Neuro Oncol 2021; 22:539-549. [PMID: 31755915 DOI: 10.1093/neuonc/noz185] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We sought to ascertain the immune effector function of pembrolizumab within the glioblastoma (GBM) microenvironment during the therapeutic window. METHODS In an open-label, single-center, single-arm phase II "window-of-opportunity" trial in 15 patients with recurrent (operable) GBM receiving up to 2 pembrolizumab doses before surgery and every 3 weeks afterward until disease progression or unacceptable toxicities occurred, immune responses were evaluated within the tumor. RESULTS No treatment-related deaths occurred. Overall median follow-up time was 50 months. Of 14 patients monitored, 10 had progressive disease, 3 had a partial response, and 1 had stable disease. Median progression-free survival (PFS) was 4.5 months (95% CI: 2.27, 6.83), and the 6-month PFS rate was 40%. Median overall survival (OS) was 20 months, with an estimated 1-year OS rate of 63%. GBM patients' recurrent tumors contained few T cells that demonstrated a paucity of immune activation markers, but the tumor microenvironment was markedly enriched for CD68+ macrophages. CONCLUSIONS Immune analyses indicated that pembrolizumab anti-programmed cell death 1 (PD-1) monotherapy alone can't induce effector immunologic response in most GBM patients, probably owing to a scarcity of T cells within the tumor microenvironment and a CD68+ macrophage preponderance.
Collapse
Affiliation(s)
- John de Groot
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marta Penas-Prado
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kristin Alfaro-Munoz
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathy Hunter
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Be Lian Pei
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara O'Brien
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiao-Pei Weathers
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monica Loghin
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos Kamiya Matsouka
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W K Alfred Yung
- Departments of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jacob Mandel
- Department of Neurology, Baylor College of Medicine, Houston, Texas
| | - Jimin Wu
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ying Yuan
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shouhao Zhou
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Huse
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ganesh Rao
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey S Weinberg
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sujit S Prabhu
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ian E McCutcheon
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherise D Ferguson
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Raymond Sawaya
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rivka Colen
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shalini S Yadav
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis Vence
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James Allison
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
20
|
Genoud V, Migliorini D. Challenging Hurdles of Current Targeting in Glioblastoma: A Focus on Immunotherapeutic Strategies. Int J Mol Sci 2021; 22:3493. [PMID: 33800593 PMCID: PMC8036548 DOI: 10.3390/ijms22073493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/23/2023] Open
Abstract
Glioblastoma is the most frequent primary neoplasm of the central nervous system and still suffers from very poor therapeutic impact. No clear improvements over current standard of care have been made in the last decade. For other cancers, but also for brain metastasis, which harbors a very distinct biology from glioblastoma, immunotherapy has already proven its efficacy. Efforts have been pursued to allow glioblastoma patients to benefit from these new approaches, but the road is still long for broad application. Here, we aim to review key glioblastoma immune related characteristics, current immunotherapeutic strategies being explored, their potential caveats, and future directions.
Collapse
Affiliation(s)
- Vassilis Genoud
- Department of Oncology, University Hospital of Geneva, 1205 Geneva, Switzerland;
- Center for Translational Research in Onco-Haematology, University of Geneva, 1205 Geneva, Switzerland
| | - Denis Migliorini
- Department of Oncology, University Hospital of Geneva, 1205 Geneva, Switzerland;
- Center for Translational Research in Onco-Haematology, University of Geneva, 1205 Geneva, Switzerland
- Brain Tumor and Immune Cell Engineering Laboratory, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, 1205 Geneva, Switzerland
| |
Collapse
|
21
|
Neuroinflammatory changes of the normal brain tissue in cured mice following combined radiation and anti-PD-1 blockade therapy for glioma. Sci Rep 2021; 11:5057. [PMID: 33658642 PMCID: PMC7930115 DOI: 10.1038/s41598-021-84600-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
The efficacy of combining radiation therapy with immune checkpoint inhibitor blockade to treat brain tumors is currently the subject of multiple investigations and holds significant therapeutic promise. However, the long-term effects of this combination therapy on the normal brain tissue are unknown. Here, we examined mice that were intracranially implanted with murine glioma cell line and became long-term survivors after treatment with a combination of 10 Gy cranial irradiation (RT) and anti-PD-1 checkpoint blockade (aPD-1). Post-mortem analysis of the cerebral hemisphere contralateral to tumor implantation showed complete abolishment of hippocampal neurogenesis, but neural stem cells were well preserved in subventricular zone. In addition, we observed a drastic reduction in the number of mature oligodendrocytes in the subcortical white matter. Importantly, this observation was evident specifically in the combined (RT + aPD-1) treatment group but not in the single treatment arm of either RT alone or aPD-1 alone. Elimination of microglia with a small molecule inhibitor of colony stimulated factor-1 receptor (PLX5622) prevented the loss of mature oligodendrocytes. These results identify for the first time a unique pattern of normal tissue changes in the brain secondary to combination treatment with radiotherapy and immunotherapy. The results also suggest a role for microglia as key mediators of the adverse treatment effect.
Collapse
|
22
|
Sasidharan Nair V, Saleh R, Toor SM, Cyprian FS, Elkord E. Metabolic reprogramming of T regulatory cells in the hypoxic tumor microenvironment. Cancer Immunol Immunother 2021; 70:2103-2121. [PMID: 33532902 PMCID: PMC8289790 DOI: 10.1007/s00262-020-02842-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022]
Abstract
Metabolic dysregulation in the hypoxic tumor microenvironment (TME) is considered as a hallmark of solid tumors, leading to changes in biosynthetic pathways favoring onset, survival and proliferation of malignant cells. Within the TME, hypoxic milieu favors metabolic reprogramming of tumor cells, which subsequently affects biological properties of tumor-infiltrating immune cells. T regulatory cells (Tregs), including both circulating and tissue-resident cells, are particularly susceptible to hypoxic metabolic signaling that can reprogram their biological and physicochemical properties. Furthermore, metabolic reprogramming modifies Tregs to utilize alternative substrates and undergo a plethora of metabolic events to meet their energy demands. Major impact of this metabolic reprogramming can result in differentiation, survival, excessive secretion of immunosuppressive cytokines and proliferation of Tregs within the TME, which in turn dampen anti-tumor immune responses. Studies on fine-tuning of Treg metabolism are challenging due to heterogenicity of tissue-resident Tregs and their dynamic functions. In this review, we highlight tumor intrinsic and extrinsic factors, which can influence Treg metabolism in the hypoxic TME. Moreover, we focus on metabolic reprogramming of Tregs that could unveil potential regulatory networks favoring tumorigenesis/progression, and provide novel insights, including inhibitors against acetyl-coA carboxylase 1 and transforming growth factor beta into targeting Treg metabolism for therapeutic benefits.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Farhan S Cyprian
- Department of Basic Medical Sciences, College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, M5 4WT, UK.
| |
Collapse
|
23
|
Romano E, Honeychurch J, Illidge TM. Radiotherapy-Immunotherapy Combination: How Will We Bridge the Gap Between Pre-Clinical Promise and Effective Clinical Delivery? Cancers (Basel) 2021; 13:457. [PMID: 33530329 PMCID: PMC7865752 DOI: 10.3390/cancers13030457] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is highly effective at directly killing tumor cells and plays an important part in cancer treatments being delivered to around 50% of all cancer patients. The additional immunomodulatory properties of RT have been investigated, and if exploited effectively, have the potential to further improve the efficacy of RT and cancer outcomes. The initial results of combining RT with immunomodulatory agents have generated promising data in pre-clinical studies, which has in turn led to a large number of RT and immunotherapy clinical trials. The overarching aim of these combinations is to enhance anti-tumor immune responses and improve responses rates and patient outcomes. In order to maximize this undoubted opportunity, there remain a number of important questions that need to be addressed, including: (i) the optimal RT dose and fractionation schedule; (ii) the optimal RT target volume; (iii) the optimal immuno-oncology (IO) agent(s) to partner with RT; (iv) the optimal site(s)/route(s) of administration of IO agents; and finally, the optimal RT schedule. In this review, we will summarize progress to date and identify current gaps in knowledge that need to be addressed in order to facilitate effective clinical translation of RT and IO agent combinations.
Collapse
Affiliation(s)
- Erminia Romano
- Division of Cancer Sciences, Faculty of Biology, School of Medical Sciences, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (E.R.); (J.H.)
| | - Jamie Honeychurch
- Division of Cancer Sciences, Faculty of Biology, School of Medical Sciences, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (E.R.); (J.H.)
| | - Timothy M. Illidge
- Division of Cancer Sciences, Faculty of Biology, School of Medical Sciences, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (E.R.); (J.H.)
- Manchester Academic Health Science Centre, NIHR Biomedical Research Centre, The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| |
Collapse
|
24
|
Wouters R, Bevers S, Riva M, De Smet F, Coosemans A. Immunocompetent Mouse Models in the Search for Effective Immunotherapy in Glioblastoma. Cancers (Basel) 2020; 13:E19. [PMID: 33374542 PMCID: PMC7793150 DOI: 10.3390/cancers13010019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive intrinsic brain tumor in adults. Despite maximal therapy consisting of surgery and radio/chemotherapy, GBM remains largely incurable with a median survival of less than 15 months. GBM has a strong immunosuppressive nature with a multitude of tumor and microenvironment (TME) derived factors that prohibit an effective immune response. To date, all clinical trials failed to provide lasting clinical efficacy, despite the relatively high success rates of preclinical studies to show effectivity of immunotherapy. Various factors may explain this discrepancy, including the inability of a single mouse model to fully recapitulate the complexity and heterogeneity of GBM. It is therefore critical to understand the features and limitations of each model, which should probably be combined to grab the full spectrum of the disease. In this review, we summarize the available knowledge concerning immune composition, stem cell characteristics and response to standard-of-care and immunotherapeutics for the most commonly available immunocompetent mouse models of GBM.
Collapse
Affiliation(s)
- Roxanne Wouters
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Oncoinvent, A.S., 0484 Oslo, Norway
| | - Sien Bevers
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - Matteo Riva
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
- Department of Neurosurgery, Mont-Godinne Hospital, UCL Namur, 5530 Yvoir, Belgium
| | - Frederik De Smet
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium;
| | - An Coosemans
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000 Leuven, Belgium; (R.W.); (S.B.); (M.R.)
| |
Collapse
|
25
|
Immunotherapy for Glioblastoma: Current State, Challenges, and Future Perspectives. Cancers (Basel) 2020; 12:cancers12092334. [PMID: 32824974 PMCID: PMC7565291 DOI: 10.3390/cancers12092334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/10/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is the most lethal intracranial primary malignancy by no optimal treatment option. Cancer immunotherapy has achieved remarkable survival benefits against various advanced tumors, such as melanoma and non-small-cell lung cancer, thus triggering great interest as a new therapeutic strategy for glioblastoma. Moreover, the central nervous system has been rediscovered recently as a region for active immunosurveillance. There are vibrant investigations for successful glioblastoma immunotherapy despite the fact that initial clinical trial results are somewhat disappointing with unique challenges including T-cell dysfunction in the patients. This review will explore the potential of current immunotherapy modalities for glioblastoma treatment, especially focusing on major immune checkpoint inhibitors and the future strategies with novel targets and combo therapies. Immune-related adverse events and clinical challenges in glioblastoma immunotherapy are also summarized. Glioblastoma provides persistent difficulties for immunotherapy with a complex state of patients’ immune dysfunction and a variety of constraints in drug delivery to the central nervous system. However, rational design of combinational regimens and new focuses on myeloid cells and novel targets to circumvent current limitations hold promise to advent truly viable immunotherapy for glioblastoma.
Collapse
|
26
|
Repolarized macrophages, induced by intermediate stereotactic dose radiotherapy and immune checkpoint blockade, contribute to long-term survival in glioma-bearing mice. J Neurooncol 2020; 147:547-555. [PMID: 32215786 DOI: 10.1007/s11060-020-03459-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is a deadly brain tumor with a short expected median survival, despite current standard-of-care treatment. We explored the combination of intermediate stereotactic dose radiation therapy and immune checkpoint inhibitor therapy as a novel treatment strategy for GBM. METHODS Glioma xenograft-bearing mice were exposed to high dose brain-directed radiation (10 Gy single exposure) as well as mouse anti-PD-1 antibody. The tumor-bearing animals were randomized to four groups: no treatment, radiation alone, anti-PD-1 alone, and radiation + anti-PD-1. Survival was followed, and tumor growth was monitored using MRI. Immunohistochemistry, gene expression arrays, and flow cytometry were used to characterize the treatment-induced effects. Pharmacologic inhibitors of T-lymphocytes, bone marrow derived macrophages, and microglia were used to assess the respective roles of different immune populations in observed treatment effects. RESULTS We found the combined treatment with high dose radiation and immunotherapy to be highly effective with a 75% complete pathologic response and dramatically improved survival outcomes. We found both CD8+ T-cells and macrophages to be necessary for the full effect of combined therapy, with T lymphocytes appearing to play a role early on and macrophages mediating a later phase of the combined treatment effect. Radiation treatment appeared to trigger macrophage repolarization, increasing M1/M2 ratio. CONCLUSIONS These findings point to a novel immunologic mechanism underlying the interaction between radiotherapy and immunotherapy. They also provide the basis for clinical investigation of immunogenic dose radiation in combination with immune checkpoint blockade as a potential treatment approach for newly diagnosed high grade gliomas.
Collapse
|
27
|
Mohme M, Neidert MC. Tumor-Specific T Cell Activation in Malignant Brain Tumors. Front Immunol 2020; 11:205. [PMID: 32117316 PMCID: PMC7031483 DOI: 10.3389/fimmu.2020.00205] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/27/2020] [Indexed: 12/17/2022] Open
Abstract
Due to their delicate locations as well as aggressive and infiltrative behavior, malignant brain tumors remain a therapeutic challenge. Harnessing the efficacy and specificity of the T-cell response to counteract malignant brain tumor progression and recurrence, represents an attractive treatment option. With the tremendous advances in the current era of immunotherapy, ongoing studies aim to determine the best treatment strategies for mounting a tumor-specific immune response against malignant brain tumors. However, immunosuppression in the local tumor environment, molecular and cellular heterogeneity as well as a lack of suitable targets for tumor-specific vaccination impede the successful implementation of immunotherapeutic treatment strategies in neuro-oncology. In this review, we therefore discuss the role of T cell exhaustion, the genetic and antigenic landscape, potential pitfalls and ongoing efforts to overcome the individual challenges in order to elicit a tumor-specific T cell response.
Collapse
Affiliation(s)
- Malte Mohme
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marian Christoph Neidert
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.,Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.,Broad Institute of Harvard and MIT, Cambridge, MA, United States
| |
Collapse
|
28
|
Pombo Antunes AR, Scheyltjens I, Duerinck J, Neyns B, Movahedi K, Van Ginderachter JA. Understanding the glioblastoma immune microenvironment as basis for the development of new immunotherapeutic strategies. eLife 2020; 9:52176. [PMID: 32014107 PMCID: PMC7000215 DOI: 10.7554/elife.52176] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy by immune checkpoint blockade has proven its great potential by saving the lives of a proportion of late stage patients with immunogenic tumor types. However, even in these sensitive tumor types, the majority of patients do not sufficiently respond to the therapy. Furthermore, other tumor types, including glioblastoma, remain largely refractory. The glioblastoma immune microenvironment is recognized as highly immunosuppressive, posing a major hurdle for inducing immune-mediated destruction of cancer cells. Scattered information is available about the presence and activity of immunosuppressive or immunostimulatory cell types in glioblastoma tumors, including tumor-associated macrophages, tumor-infiltrating dendritic cells and regulatory T cells. These cell types are heterogeneous at the level of ontogeny, spatial distribution and functionality within the tumor immune compartment, providing insight in the complex cellular and molecular interplay that determines the immune refractory state in glioblastoma. This knowledge may also yield next generation molecular targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ana Rita Pombo Antunes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Scheyltjens
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Bart Neyns
- Department of Medical Oncology, UZ Brussels, Brussels, Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
29
|
Verma A, Mathur R, Farooque A, Kaul V, Gupta S, Dwarakanath BS. T-Regulatory Cells In Tumor Progression And Therapy. Cancer Manag Res 2019; 11:10731-10747. [PMID: 31920383 PMCID: PMC6935360 DOI: 10.2147/cmar.s228887] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022] Open
Abstract
Regulatory T cells (Tregs) are important members of the immune system regulating the host responses to infection and neoplasms. Tregs prevent autoimmune disorders by protecting the host-cells from an immune response, related to the peripheral tolerance. However, tumor cells use Tregs as a shield to protect themselves against anti-tumor immune response. Thus, Tregs are a hurdle in achieving the complete potential of anti-cancer therapies including immunotherapy. This has prompted the development of novel adjuvant therapies that obviate their negative effects thereby enhancing the therapeutic efficacy. Our earlier studies have shown the efficacy of the glycolytic inhibitor, 2-deoxy-D-glucose (2-DG) by reducing the induced Tregs pool and enhance immune stimulation as well as local tumor control. These findings have suggested its potential for enhancing the efficacy of immunotherapy, besides radiotherapy and chemotherapy. This review provides a brief account of the current status of Tregs as a component of the immune-biology of tumors and various preclinical and clinical strategies pursued to obviate the limitations imposed by them in achieving therapeutic efficacy.
Collapse
Affiliation(s)
- Amit Verma
- Armed Forces Radiobiology Research Institute, Uniformed Services University, Bethesda, MD, USA
| | - Rohit Mathur
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Vandana Kaul
- Division of Abdominal Transplantation, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | |
Collapse
|
30
|
Deng Z, Xu X, Garzon-Muvdi T, Xia Y, Kim E, Belcaid Z, Luksik A, Maxwell R, Choi J, Wang H, Yu J, Iordachita I, Lim M, Wong JW, Wang KKH. In Vivo Bioluminescence Tomography Center of Mass-Guided Conformal Irradiation. Int J Radiat Oncol Biol Phys 2019; 106:612-620. [PMID: 31738948 DOI: 10.1016/j.ijrobp.2019.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 01/21/2023]
Abstract
PURPOSE The cone-beam computed tomography (CBCT)-guided small animal radiation research platform (SARRP) has provided unique opportunities to test radiobiologic hypotheses. However, CBCT is less adept to localize soft tissue targets growing in a low imaging contrast environment. Three-dimensional bioluminescence tomography (BLT) provides strong image contrast and thus offers an attractive solution. We introduced a novel and efficient BLT-guided conformal radiation therapy and demonstrated it in an orthotopic glioblastoma (GBM) model. METHODS AND MATERIALS A multispectral BLT system was integrated with SARRP for radiation therapy (RT) guidance. GBM growth curve was first established by contrast CBCT/magnetic resonance imaging (MRI) to derive equivalent sphere as approximated gross target volume (aGTV). For BLT, mice were subject to multispectral bioluminescence imaging, followed by SARRP CBCT imaging and optical reconstruction. The CBCT image was acquired to generate anatomic mesh for the reconstruction and RT planning. To ensure high accuracy of the BLT-reconstructed center of mass (CoM) for target localization, we optimized the optical absorption coefficients μa by minimizing the distance between the CoMs of BLT reconstruction and contrast CBCT/MRI-delineated GBM volume. The aGTV combined with the uncertainties of BLT CoM localization and target volume determination was used to generate estimated target volume (ETV). For conformal irradiation procedure, the GBM was first localized by the predetermined ETV centered at BLT-reconstructed CoM, followed by SARRP radiation. The irradiation accuracy was qualitatively confirmed by pathologic staining. RESULTS Deviation between CoMs of BLT reconstruction and contrast CBCT/MRI-imaged GBM is approximately 1 mm. Our derived ETV centered at BLT-reconstructed CoM covers >95% of the tumor volume. Using the second-week GBM as an example, the ETV-based BLT-guided irradiation can cover 95.4% ± 4.7% tumor volume at prescribed dose. The pathologic staining demonstrated the BLT-guided irradiated area overlapped well with the GBM location. CONCLUSIONS The BLT-guided RT enables 3-dimensional conformal radiation for important orthotopic tumor models, which provides investigators a new preclinical research capability.
Collapse
Affiliation(s)
- Zijian Deng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xiangkun Xu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Neurosurgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eileen Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew Luksik
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John Choi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hailun Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jingjing Yu
- School of Physics and Information Technology, Shaanxi Normal University, Shanxi, China
| | - Iulian Iordachita
- Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John W Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ken Kang-Hsin Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
31
|
Rajani KR, Carlstrom LP, Parney IF, Johnson AJ, Warrington AE, Burns TC. Harnessing Radiation Biology to Augment Immunotherapy for Glioblastoma. Front Oncol 2019; 8:656. [PMID: 30854331 PMCID: PMC6395389 DOI: 10.3389/fonc.2018.00656] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common adult primary brain tumor and carries a dismal prognosis. Radiation is a standard first-line therapy, typically deployed following maximal safe surgical debulking, when possible, in combination with cytotoxic chemotherapy. For other systemic cancers, standard of care is being transformed by immunotherapies, including checkpoint-blocking antibodies targeting CTLA-4 and PD-1/PD-L1, with potential for long-term remission. Ongoing studies are evaluating the role of immunotherapies for GBM. Despite dramatic responses in some cases, randomized trials to date have not met primary outcomes. Challenges have been attributed in part to the immunologically "cold" nature of glioblastoma relative to other malignancies successfully treated with immunotherapy. Radiation may serve as a mechanism to improve tumor immunogenicity. In this review, we critically evaluate current evidence regarding radiation as a synergistic facilitator of immunotherapies through modulation of both the innate and adaptive immune milieu. Although current preclinical data encourage efforts to harness synergistic biology between radiation and immunotherapy, several practical and scientific challenges remain. Moreover, insights from radiation biology may unveil additional novel opportunities to help mobilize immunity against GBM.
Collapse
Affiliation(s)
- Karishma R. Rajani
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Lucas P. Carlstrom
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ian F. Parney
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | | | - Terry C. Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
32
|
Abstract
High-grade glioma is the most common primary brain tumor, with glioblastoma multiforme (GBM) accounting for 52% of all brain tumors. The current standard of care (SOC) of GBM involves surgery followed by adjuvant fractionated radiotherapy and chemotherapy. However, little progress has been made in extending overall survival, progression-free survival, and quality of life. Attempts to characterize and customize treatment of GBM have led to mitigating the deleterious effects of radiotherapy using hypofractionated radiotherapy, as well as various immunotherapies as a promising strategy for the incurable disease. A combination of radiotherapy and immunotherapy may prove to be even more effective than either alone, and preclinical evidence suggests that hypofractionated radiotherapy can actually prime the immune system to make immunotherapy more effective. This review addresses the complications of the current radiotherapy regimen, various methods of immunotherapy, and preclinical and clinical data from combined radioimmunotherapy trials.
Collapse
|
33
|
Ma Q, Long W, Xing C, Chu J, Luo M, Wang HY, Liu Q, Wang RF. Cancer Stem Cells and Immunosuppressive Microenvironment in Glioma. Front Immunol 2018; 9:2924. [PMID: 30619286 PMCID: PMC6308128 DOI: 10.3389/fimmu.2018.02924] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Glioma is one of the most common malignant tumors of the central nervous system and is characterized by extensive infiltrative growth, neovascularization, and resistance to various combined therapies. In addition to heterogenous populations of tumor cells, the glioma stem cells (GSCs) and other nontumor cells present in the glioma microenvironment serve as critical regulators of tumor progression and recurrence. In this review, we discuss the role of several resident or peripheral factors with distinct tumor-promoting features and their dynamic interactions in the development of glioma. Localized antitumor factors could be silenced or even converted to suppressive phenotypes, due to stemness-related cell reprogramming and immunosuppressive mediators in glioma-derived microenvironment. Furthermore, we summarize the latest knowledge on GSCs and key microenvironment components, and discuss the emerging immunotherapeutic strategies to cure this disease.
Collapse
Affiliation(s)
- Qianquan Ma
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Wenyong Long
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Changsheng Xing
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Junjun Chu
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Mei Luo
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China.,Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Helen Y Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, China
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, United States.,Institute of Biosciences and Technology, College of Medicine, Texas A&M University, Houston, TX, United States.,Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, NY, United States
| |
Collapse
|
34
|
Khalife E, Khodadadi A, Talaeizadeh A, Rahimian L, Nemati M, Jafarzadeh A. Overexpression of Regulatory T Cell-Related Markers (FOXP3, CTLA-4 and GITR) by Peripheral Blood Mononuclear Cells from Patients with Breast Cancer. Asian Pac J Cancer Prev 2018; 19:3019-3025. [PMID: 30484986 PMCID: PMC6318404 DOI: 10.31557/apjcp.2018.19.11.3019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Regulatory T (Treg) cells are immunosuppressor lymphocytes that play a critical role in the establishment and progression of cancers. A number of markers, especially FOXP3, CTLA-4 and GITR influence the function of Treg cells. This investigation aimed to evaluate the expression of a number of important Treg cell-related markers by peripheral blood mononuclear cells (PBMCs) from newly-diagnosed women with breast cancer. Methods: The fresh PBMCs were obtained from 20 women with breast cancer and 20 healthy individuals. The PBMCs from both groups were cultured for 32 hours in the presence or absence of PHA (10 μg/ml). After total RNA extraction from cultured PBMCs, the expression of the FOXP3, CTLA-4 and GITR transcripts was assessed using real time-PCR. Results: The mRNA expression of FOXP3, CTLA-4 and GITR in unstimulated PBMCs from patients with breast cancer were significantly higher than healthy control group (P<0.05, P<0.03 and P<0.04, respectively). Similarly, the expression of FOXP3, CTLA-4 and GITR transcripts in PHA-stimulated PBMCs from patients with breast cancer were significantly increased in comparison with healthy individuals (P<0.01, P<0.005 and P<0.01, respectively). Conclusion: The increased expression of FOXP3, CTLA-4 and GITR represent higher activity of Treg cells in patients with breast cancer that may play an important role in the tumor establishment and development.
Collapse
Affiliation(s)
- Esmat Khalife
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | | | | | | | | | | |
Collapse
|
35
|
Irradiation to Improve the Response to Immunotherapeutic Agents in Glioblastomas. Adv Radiat Oncol 2018; 4:268-282. [PMID: 31011672 PMCID: PMC6460102 DOI: 10.1016/j.adro.2018.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose Glioblastoma (GBM) remains an incurable disease despite extensive treatment with surgical resection, irradiation, and temozolomide. In line with many other forms of aggressive cancers, GBM is currently under consideration as a target for immunotherapy. However, GBM tends to be nonimmunogenic and exhibits a microenvironment with few or no effector T cells, a relatively low nonsynonymous somatic mutational load, and a low predicted neoantigen burden. GBM also exploits a multitude of immunosuppressive strategies. Methods and Materials A number of immunotherapeutic approaches have been tested with disappointing results. A rationale exists to combine immunotherapy and radiation therapy, which can induce an immunogenic form of cell death with T-cell activation and tumor infiltration. Results Various immunotherapy agents, including immune checkpoint modulators, transforming growth factor beta receptor inhibitors, and indoleamine-2,3-dioxygenase inhibitors, have been evaluated with irradiation in preclinical GBM models, with promising results, and are being further tested in clinical trials. Conclusions This review aims to present the basic rationale behind this emerging complementary therapeutic approach in GBM, appraise the current preclinical and clinical data, and discuss the future challenges in improving the antitumor immune response.
Collapse
|
36
|
RNA-seq for identification of therapeutically targetable determinants of immune activation in human glioblastoma. J Neurooncol 2018; 141:95-102. [PMID: 30353265 DOI: 10.1007/s11060-018-03010-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 09/20/2018] [Indexed: 12/31/2022]
Abstract
INTRODUCTION We sought to determine which therapeutically targetable immune checkpoints, costimulatory signals, and other tumor microenvironment (TME) factors are independently associated with immune cytolytic activity (CYT), a gene expression signature of activated effector T cells, in human glioblastoma (GBM). METHODS GlioVis was accessed for RNA-seq data from The Cancer Genome Atlas (TCGA). For subjects with treatment-naïve, primary GBM, we quantified mRNA expression of 28 therapeutically targetable TME factors. CYT (geometric mean of GZMA and PRF1 expression) was calculated for each tumor. Multiple linear regression was performed to determine the relationship between the dependent variable (CYT) and mRNA expression of each of the 28 factors. Variables associated with CYT in multivariate analysis were subsequently evaluated for this association in an independent cohort of newly diagnosed GBMs from the Chinese Glioma Cooperative Group (CGCG). RESULTS 109 TCGA tumors were analyzed. The final multiple linear regression model included the following variables, each positively associated with CYT except VEGF-A (negative association): CSF-1 (p = 0.003), CD137 (p = 0.042), VEGF-A (p < 0.001), CTLA4 (p = 0.028), CD40 (p = 0.023), GITR (p = 0.020), IL6 (p = 0.02), and OX40 (p < 0.001). In CGCG (n = 52), each of these variables remained significantly associated with CYT in univariate analysis except for VEGF-A. In multivariate analysis, only CTLA4 and CD40 remained statistically significant. CONCLUSIONS Using multivariate modeling of RNA-seq gene expression data, we identified therapeutically targetable TME factors that are independently associated with intratumoral cytolytic T-cell activity in human GBM. As a myriad of systemic immunotherapies are now available for investigation, our results could inform rational combinations for evaluation in GBM.
Collapse
|
37
|
Harris-Bookman S, Mathios D, Martin AM, Xia Y, Kim E, Xu H, Belcaid Z, Polanczyk M, Barberi T, Theodros D, Kim J, Taube JM, Burger PC, Selby M, Taitt C, Korman A, Ye X, Drake CG, Brem H, Pardoll DM, Lim M. Expression of LAG-3 and efficacy of combination treatment with anti-LAG-3 and anti-PD-1 monoclonal antibodies in glioblastoma. Int J Cancer 2018; 143:3201-3208. [PMID: 30248181 DOI: 10.1002/ijc.31661] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 02/03/2023]
Abstract
Like in many tumor types, immunotherapy is currently under investigation to assess its potential efficacy in glioblastoma patients. Trials are under way to assess the efficacy of new immune checkpoint inhibitors including anti-PD-1 or CTLA4. We here investigate the expression and efficacy of a novel immune-checkpoint inhibitor, called LAG-3. We show that LAG-3 is expressed in human glioblastoma samples and in a mouse glioblastoma model we show that knock out or LAG-3 inhibition with a blocking antibody is efficacious against glioblastoma and can be used in combination with other immune checkpoint inhibitors toward complete eradication of the model glioblastoma tumors. From a mechanistic standpoint we show that LAG-3 expression is an early marker of T cell exhaustion and therefore early treatment with LAG-3 blocking antibody is more efficacious than later treatment. These data provide insight and support the design of trials that incorporate LAG-3 in the treatment of glioblastoma.
Collapse
MESH Headings
- Aged
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/therapeutic use
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Cell Line, Tumor
- Female
- Flow Cytometry
- Glioblastoma/immunology
- Glioblastoma/therapy
- Humans
- Immunohistochemistry
- Immunologic Memory
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Programmed Cell Death 1 Receptor/immunology
- Survival Analysis
- Xenograft Model Antitumor Assays
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Sarah Harris-Bookman
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allison M Martin
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eileen Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Haiying Xu
- Department of Dermatology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Magdalena Polanczyk
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Theresa Barberi
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Debebe Theodros
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Janis M Taube
- Department of Dermatology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Peter C Burger
- Department of Neuropathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mark Selby
- Bristol-Myers Squibb Company, New York, NY
| | | | | | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Charles G Drake
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Drew M Pardoll
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Division Pediatric Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
38
|
Schoenhals JE, Cushman TR, Barsoumian HB, Li A, Cadena AP, Niknam S, Younes AI, Caetano MDS, Cortez MA, Welsh JW. Anti-glucocorticoid-induced Tumor Necrosis Factor-Related Protein (GITR) Therapy Overcomes Radiation-Induced Treg Immunosuppression and Drives Abscopal Effects. Front Immunol 2018; 9:2170. [PMID: 30294332 PMCID: PMC6158365 DOI: 10.3389/fimmu.2018.02170] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/03/2018] [Indexed: 12/15/2022] Open
Abstract
Despite the potential to cure metastatic disease, immunotherapy on its own often fails outright or early on due to tumor immune evasion. To address this obstacle, we investigated combinations of anti-GITR, anti-PD1 and radiation therapy (XRT) in our previously developed anti-PD1 resistant 344SQ non-small cell lung adenocarcinoma preclinical tumor model. We hypothesized that targeting multiple mechanisms of immune evasion with this triple therapy would lead to an enhanced tumor-specific immune response and improve survival more so than any mono- or dual therapy. In a two tumor 344SQR murine model, treatment with anti-GITR, anti-PD1, and XRT led to significantly improved survival and an abscopal response, with half of the mice becoming tumor free. These mice showed durable response and increased CD4+ and CD8+ effector memory on tumor rechallenge. Regulatory T cells (Tregs) expressed the highest level of GITR at the tumor site and anti-GITR therapy drastically diminished Tregs at the tumor site. Anti-tumor effects were largely dependent on CD4+ T cells and partially dependent on CD8+ T cells. Anti-GITR IgG2a demonstrated superior efficacy to anti-GITR IgG1 in driving antitumor effects. Collectively, these results suggest that combinatorial strategies targeting multiple points of tumor immune evasion may lead to a robust and lasting antitumor response.
Collapse
Affiliation(s)
- Jonathan E Schoenhals
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Taylor R Cushman
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hampartsoum B Barsoumian
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ailin Li
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexandra P Cadena
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sharareh Niknam
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ahmed I Younes
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mauricio da Silva Caetano
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Angelica Cortez
- Experimental Radiation Oncology Departments, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - James W Welsh
- Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
39
|
The Imbalance of FOXP3/GATA3 in Regulatory T Cells from the Peripheral Blood of Asthmatic Patients. J Immunol Res 2018; 2018:3096183. [PMID: 30013989 PMCID: PMC6022336 DOI: 10.1155/2018/3096183] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/02/2018] [Indexed: 12/15/2022] Open
Abstract
Background Treg cells play an important role in the pathogenic progress of asthma. Objective To address the alterations of Treg cells in asthma. Methods Proliferation-and function-associated markers of Treg cells along with the percentage of Treg cells producing some cytokine from asthmatics and healthy subjects were analyzed by flow cytometry. Besides, the expressions of USP21 and PIM2 in Treg cells were measured by cell immunochemistry after Treg cells were sorted. Results Treg cells from asthmatic patients showed lower proliferation activity and were more likely to be apoptotic. These cells expressed lower levels of GITR, CTLA-4, Nrp-1, and IL-10 compared to those from the healthy control. Th2-like Treg cells increased in asthmatic patients, while the percentage of IFN-r+ Treg cells was similar between two groups. Moreover, the percentage of IL-4+ Treg cells is related to the asthma control. Treg cells from asthmatic patients expressed more FOXP3 as well as GATA3; the expression level of GATA3 negatively correlated with FEV1%pred. Increased expressions of USP21 and PIM2 in Treg cells from asthmatic patients were found. Conclusion Treg cells decreased in asthmatic patients, with an impaired immunosupression function and a Th2-like phenotype, which may be due to overexpression of GATA3 and FOXP3, regulated by USP21 and PIM2, respectively.
Collapse
|
40
|
Lim M, Jackson CM. Fire in the Smoke: Battling Brain Tumors. CEREBRUM : THE DANA FORUM ON BRAIN SCIENCE 2018; 2018:cer-07-18. [PMID: 30746035 PMCID: PMC6353124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Therapeutic vaccines, drugs, and modified human cells that activate the immune system against cancer have improved outcomes and prolonged lives in some types of cancer in the past few years. For patients with glioblastoma, the most common primary brain tumor in adults, immunotherapy is still struggling to overcome this lethal malignancy.
Collapse
|
41
|
|
42
|
Abstract
PURPOSE OF REVIEW Immunotherapy has emerged as a cornerstone of modern oncology with regulatory approvals for a variety of immunotherapeutics being achieved for a spectrum of cancer indications. Nonetheless the role of these approaches for patients with glioblastoma (GBM), the most common and deadliest primary malignant brain neoplasm, remains unknown. In this review, we summarize the current status of clinical development for the major types of immunotherapeutics, including vaccines, cell-based therapies, and immune checkpoint modulators for GBM. We also highlight potential challenges confronting the development of these agents. RECENT FINDINGS Growing preclinical and clinical data is emerging regarding the potential of immunotherapy strategies for GBM. In parallel, growing data demonstrating that historical dogma classifying the brain as immunoprivileged is inaccurate but that many tumors, including GBM evoke myriad mechanisms to suppress antitumor immune responses. SUMMARY Ongoing initial trials will provide preliminary data on the role of immunotherapy for GBM patients. Subsequent clinical development steps will likely require rationally designed combinatorial regimens.
Collapse
|
43
|
Luksik AS, Maxwell R, Garzon-Muvdi T, Lim M. The Role of Immune Checkpoint Inhibition in the Treatment of Brain Tumors. Neurotherapeutics 2017; 14:1049-1065. [PMID: 28258545 PMCID: PMC5722751 DOI: 10.1007/s13311-017-0513-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The standard of care for malignant gliomas of the brain has changed very little over the last few decades, and does not offer a cure for these rare, but fatal, tumors. The field of immunotherapy has brought potent new drugs into the oncological armamentarium, and is becoming recognized as a potentially important arm in the treatment of glioblastoma for adults. Immune checkpoints are inhibitory receptors found on immune cells that, when stimulated, cause those immune cells to become quiescent. While this is a natural mechanism to prevent excessive inflammatory damage and autoimmunity in otherwise healthy tissues, cancer cells may utilize this process to grow in the absence of targeted immune destruction. Antibodies derived to block the stimulation of these negative checkpoints, allowing immune cells to remain activated and undergo effector function, are a growing area of immunotherapy. These therapies have seen much success in both the preclinical and clinical arenas for various tumors, particularly melanoma and nonsmall-cell lung cancer. Multiple clinical trials are underway to determine if these drugs have efficacy in glioblastoma. Here, we review the current evidence, from early preclinical data to lessons learned from clinical trials outside of glioblastoma, to assess the potential of immune checkpoint inhibition in the treatment of brain tumors and discuss how this therapy may be implemented with the present standard of care.
Collapse
Affiliation(s)
- Andrew S Luksik
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Razpotnik R, Novak N, Čurin Šerbec V, Rajcevic U. Targeting Malignant Brain Tumors with Antibodies. Front Immunol 2017; 8:1181. [PMID: 28993773 PMCID: PMC5622144 DOI: 10.3389/fimmu.2017.01181] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/06/2017] [Indexed: 12/31/2022] Open
Abstract
Antibodies have been shown to be a potent therapeutic tool. However, their use for targeting brain diseases, including neurodegenerative diseases and brain cancers, has been limited, particularly because the blood–brain barrier (BBB) makes brain tissue hard to access by conventional antibody-targeting strategies. In this review, we summarize new antibody therapeutic approaches to target brain tumors, especially malignant gliomas, as well as their potential drawbacks. Many different brain delivery platforms for antibodies have been studied such as liposomes, nanoparticle-based systems, cell-penetrating peptides (CPPs), and cell-based approaches. We have already shown the successful delivery of single-chain fragment variable (scFv) with CPP as a linker between two variable domains in the brain. Antibodies normally face poor penetration through the BBB, with some variants sufficiently passing the barrier on their own. A “Trojan horse” method allows passage of biomolecules, such as antibodies, through the BBB by receptor-mediated transcytosis (RMT). Such examples of therapeutic antibodies are the bispecific antibodies where one binding specificity recognizes and binds a BBB receptor, enabling RMT and where a second binding specificity recognizes an antigen as a therapeutic target. On the other hand, cell-based systems such as stem cells (SCs) are a promising delivery system because of their tumor tropism and ability to cross the BBB. Genetically engineered SCs can be used in gene therapy, where they express anti-tumor drugs, including antibodies. Different types and sources of SCs have been studied for the delivery of therapeutics to the brain; both mesenchymal stem cells (MSCs) and neural stem cells (NSCs) show great potential. Following the success in treatment of leukemias and lymphomas, the adoptive T-cell therapies, especially the chimeric antigen receptor-T cells (CAR-Ts), are making their way into glioma treatment as another type of cell-based therapy using the antibody to bind to the specific target(s). Finally, the current clinical trials are reviewed, showing the most recent progress of attractive approaches to deliver therapeutic antibodies across the BBB aiming at the specific antigen.
Collapse
Affiliation(s)
- Rok Razpotnik
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Neža Novak
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Vladka Čurin Šerbec
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Uros Rajcevic
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
45
|
Curley CT, Sheybani ND, Bullock TN, Price RJ. Focused Ultrasound Immunotherapy for Central Nervous System Pathologies: Challenges and Opportunities. Theranostics 2017; 7:3608-3623. [PMID: 29109764 PMCID: PMC5667336 DOI: 10.7150/thno.21225] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023] Open
Abstract
Immunotherapy is rapidly emerging as the cornerstone for the treatment of several forms of metastatic cancer, as well as for a host of other pathologies. Meanwhile, several new high-profile studies have uncovered remarkable linkages between the central nervous and immune systems. With these recent developments, harnessing the immune system for the treatment of brain pathologies is a promising strategy. Here, we contend that MR image-guided focused ultrasound (FUS) represents a noninvasive approach that will allow for favorable therapeutic immunomodulation in the setting of the central nervous system. One obstacle to effective immunotherapeutic drug delivery to the brain is the blood brain barrier (BBB), which refers to the specialized structure of brain capillaries that prevents transport of most therapeutics from the blood into brain tissue. When applied in the presence of circulating microbubbles, FUS can safely and transiently open the BBB to facilitate the delivery of immunotherapeutic agents into the brain parenchyma. Furthermore, it has been demonstrated that physical perturbations of the tissue microenvironment via FUS can modulate immune response in both normal and diseased tissue. In this review article, we provide an overview of FUS energy regimens and corresponding tissue bioeffects, followed by a review of the literature pertaining to FUS for therapeutic antibody delivery in normal brain and preclinical models of brain disease. We provide an overview of studies that demonstrate FUS-mediated immune modulation in both the brain and peripheral settings. Finally, we provide remarks on challenges facing FUS immunotherapy and opportunities for future expansion in this area.
Collapse
Affiliation(s)
- Colleen T. Curley
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | - Natasha D. Sheybani
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| | | | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| |
Collapse
|
46
|
Immunotherapy and radiation in glioblastoma. J Neurooncol 2017; 134:531-539. [DOI: 10.1007/s11060-017-2413-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/03/2017] [Indexed: 02/06/2023]
|
47
|
Hung AL, Garzon-Muvdi T, Lim M. Biomarkers and Immunotherapeutic Targets in Glioblastoma. World Neurosurg 2017; 102:494-506. [PMID: 28300714 DOI: 10.1016/j.wneu.2017.03.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is an aggressive central nervous system cancer with poor prognosis despite maximal therapy. The recent advent of immunotherapy holds great promise for improving GBM survival and has already made great strides toward changing management strategies. A diverse set of biomarkers have been implicated as immunotherapeutic targets and prognostic indicators in other cancers. Some of the more extensively studied examples include cytokines (IL-4, IL-13, and TGF-β), checkpoint molecules (PD-1, CTLA-4, TIM-3, LAG-3, CD137, GITR, OX40), and growth/angiogenesis proteins (endoglin and EGFR). Emerging theories involving the tumor mutational landscape and microbiome have also been explored in relation to cancer treatment. Although identification of novel biomarkers may improve and help direct treatment of patients with GBM, the next step is to explore the role of biomarkers in precision medicine and selection of specific immunotherapeutic drugs in an individualized manner.
Collapse
Affiliation(s)
- Alice L Hung
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
48
|
Patel MA, Kim JE, Theodros D, Tam A, Velarde E, Kochel CM, Francica B, Nirschl TR, Ghasemzadeh A, Mathios D, Harris-Bookman S, Jackson CC, Jackson C, Ye X, Tran PT, Tyler B, Coric V, Selby M, Brem H, Drake CG, Pardoll DM, Lim M. Erratum to: Agonist anti-GITR monoclonal antibody and stereotactic radiation induce immune-mediated survival advantage in murine intracranial glioma. J Immunother Cancer 2016; 4:74. [PMID: 27822377 PMCID: PMC5096004 DOI: 10.1186/s40425-016-0181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 10/25/2016] [Indexed: 11/10/2022] Open
Affiliation(s)
- Mira A Patel
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Jennifer E Kim
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Debebe Theodros
- The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ada Tam
- Department of Oncology, Baltimore, USA
| | | | | | | | | | | | - Dimitrios Mathios
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Sarah Harris-Bookman
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Christopher C Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Christina Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Xiaobu Ye
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | - Phuoc T Tran
- Department of Oncology, Baltimore, USA.,Department Radiation Oncology, Baltimore, USA.,The Brady Urological Institute, Baltimore, USA
| | - Betty Tyler
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | | | - Mark Selby
- Bristol-Myers Squibb Company, San Francisco, CA USA
| | - Henry Brem
- The Johns Hopkins University School of Medicine, Baltimore, USA.,Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| | | | | | - Michael Lim
- The Johns Hopkins University School of Medicine, Baltimore, USA.,Department of Neurosurgery, The Johns Hopkins University School of Medicine, 600 N. Wolfe St. Phipps Building Rm 123, Baltimore, 21287 MD USA
| |
Collapse
|
49
|
Miska J, Rashidi A, Chang AL, Muroski ME, Han Y, Zhang L, Lesniak MS. Anti-GITR therapy promotes immunity against malignant glioma in a murine model. Cancer Immunol Immunother 2016; 65:1555-1567. [PMID: 27734112 DOI: 10.1007/s00262-016-1912-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/30/2016] [Indexed: 01/06/2023]
Abstract
Regulatory T cells (Tregs) are potently immunosuppressive cells that accumulate within the glioma microenvironment. The reduction in their function and/or trafficking has been previously shown to enhance survival in preclinical models of glioma. Glucocorticoid-induced TNFR-related protein (GITR) is a tumor necrosis factor superfamily receptor enriched on Tregs that has shown promise as a target for immunotherapy. An agonistic antibody against GITR has been demonstrated to inhibit Tregs in a number of models and has only been recently addressed in glioma. In this study, we examined the modality of the antibody function at the tumor site as opposed to the periphery as the blood-brain barrier prevents efficient antibody delivery to brain tumors. Mice harboring established GL261 tumors were treated with anti-GITR monotherapy and were shown to have a significant increase in overall survival (p < 0.01) when antibodies were injected directly into the glioma core, whereas peripheral antibody treatment only had a modest effect. Peripheral treatment resulted in a significant decrease in granzyme B (GrB) expression by Tregs, whereas intratumoral treatment resulted in both a decrease in GrB expression by Tregs and their selective depletion, which was largely mediated by FcγR-mediated destruction. We also discovered that anti-GITR treatment results in the enhanced survival and functionality of dendritic cells (DCs)-a previously unreported effect of this immunotherapy. In effect, this study demonstrates that the targeting of GITR is a feasible and noteworthy treatment option for glioma, but is largely dependent on the anatomical location in which the antibodies are delivered.
Collapse
Affiliation(s)
- Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Alan L Chang
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Megan E Muroski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Lingjiao Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| |
Collapse
|