651
|
Neagu MR, Huang RY, Reardon DA, Wen PY. How treatment monitoring is influencing treatment decisions in glioblastomas. Curr Treat Options Neurol 2015; 17:343. [PMID: 25749847 DOI: 10.1007/s11940-015-0343-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Glioblastoma (GBM), the most common malignant primary tumor in adults, carries a dismal prognosis with an average median survival of 14-16 months. The current standard of care for newly diagnosed GBM consists of maximal safe resection followed by fractionated radiotherapy combined with concurrent temozolomide and 6 to 12 cycles of adjuvant temozolomide. The determination of treatment response and clinical decision-making in the treatment of GBM depends on accurate radiographic assessment. Differentiating treatment response from tumor progression is challenging and combines long-term follow-up using standard MRI, with assessing clinical status and corticosteroid dependency. At progression, bevacizumab is the mainstay of treatment. Incorporation of antiangiogenic therapies leads to rapid blood-brain barrier normalization with remarkable radiographic response often not accompanied by the expected survival benefit, further complicating imaging assessment. Improved radiographic interpretation criteria, such as the Response Assessment in Neuro-Oncology (RANO) criteria, incorporate non-enhancing disease but still fall short of definitely distinguishing tumor progression, pseudoresponse, and pseudoprogression. With new evolving treatment modalities for this devastating disease, advanced imaging modalities are increasingly becoming part of routine clinical care in a field where neuroimaging has such essential role in guiding treatment decisions and defining clinical trial eligibility and efficacy.
Collapse
Affiliation(s)
- Martha R Neagu
- Dana Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA
| | | | | | | |
Collapse
|
652
|
Kim JE, Lim M. The role of checkpoints in the treatment of GBM. J Neurooncol 2015; 123:413-23. [PMID: 25749875 DOI: 10.1007/s11060-015-1747-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/16/2015] [Indexed: 12/18/2022]
Abstract
Targeted immunotherapy is founded on the principle that augmentation of effector T cell activity in the tumor microenvironment can translate to tumor regression. Targeted checkpoint inhibitors in the form of agonist or antagonist monoclonal antibodies have come to the fore as a promising strategy to activate systemic immunity and enhance T cell activity by blocking negative signals, enhancing positive signals, or altering the cytokine milieu. This review will examine several immune checkpoints and checkpoint modulators that play a role in cancer pathogenesis, with an emphasis on malignant gliomas.
Collapse
Affiliation(s)
- Jennifer E Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
653
|
Wang JY, Bettegowda C. Genetics and immunotherapy: using the genetic landscape of gliomas to inform management strategies. J Neurooncol 2015; 123:373-83. [PMID: 25697584 DOI: 10.1007/s11060-015-1730-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/01/2015] [Indexed: 02/07/2023]
Abstract
Recent work in genetics has identified essential driver mutations in gliomas and has profoundly changed our understanding of tumorigenesis. New insights into the molecular basis of glioma has informed the development of therapies demonstrating considerable potential, including immunotherapeutic approaches such as peptide and dendritic cell vaccines against EGFRvIII. However, the selective targeting of one component of a dysregulated pathway may be inadequate for a durable clinical response, given the intratumoral heterogeneity of glioblastoma (GBM) and hypermutated profiles displayed by tumor recurrences. Immune checkpoint blockade with anti-cytotoxic T lymphocyte antigen-4 (CTLA) and anti-programmed cell death 1 (PD-1) have demonstrated encouraging results in clinical trials with other solid tumors, and recent data suggest that this type of therapy may be particularly useful for tumors with high mutational burdens. Although the survival for patients with GBM has remains grim, the use of immunotherapy may finally change patient outcomes.
Collapse
Affiliation(s)
- Joanna Y Wang
- Department of Neurosurgery, The Johns Hopkins Hospital, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Phipps 118, Baltimore, MD, 21287, USA
| | | |
Collapse
|
654
|
Park SS, Dong H, Liu X, Harrington SM, Krco CJ, Grams MP, Mansfield AS, Furutani KM, Olivier KR, Kwon ED. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol Res 2015; 3:610-9. [PMID: 25701325 DOI: 10.1158/2326-6066.cir-14-0138] [Citation(s) in RCA: 309] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/10/2015] [Indexed: 12/28/2022]
Abstract
We investigated the influence of PD-1 expression on the systemic antitumor response (abscopal effect) induced by stereotactic ablative radiotherapy (SABR) in preclinical melanoma and renal cell carcinoma models. We compared the SABR-induced antitumor response in PD-1-expressing wild-type (WT) and PD-1-deficient knockout (KO) mice and found that PD-1 expression compromises the survival of tumor-bearing mice treated with SABR. None of the PD-1 WT mice survived beyond 25 days, whereas 20% of the PD-1 KO mice survived beyond 40 days. Similarly, PD-1-blocking antibody in WT mice was able to recapitulate SABR-induced antitumor responses observed in PD-1 KO mice and led to increased survival. The combination of SABR plus PD-1 blockade induced near complete regression of the irradiated primary tumor (synergistic effect), as opposed to SABR alone or SABR plus control antibody. The combination of SABR plus PD-1 blockade therapy elicited a 66% reduction in size of nonirradiated, secondary tumors outside the SABR radiation field (abscopal effect). The observed abscopal effect was tumor specific and was not dependent on tumor histology or host genetic background. The CD11a(high) CD8(+) T-cell phenotype identifies a tumor-reactive population, which was associated in frequency and function with a SABR-induced antitumor immune response in PD-1 KO mice. We conclude that SABR induces an abscopal tumor-specific immune response in both the irradiated and nonirradiated tumors, which is potentiated by PD-1 blockade. The combination of SABR and PD-1 blockade has the potential to translate into a potent immunotherapy strategy in the management of patients with metastatic cancer.
Collapse
Affiliation(s)
- Sean S Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, Minnesota. Department of Urology, Mayo Clinic, Rochester, Minnesota
| | - Xin Liu
- Department of Urology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Michael P Grams
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Keith M Furutani
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Eugene D Kwon
- Department of Immunology, Mayo Clinic, Rochester, Minnesota. Department of Urology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
655
|
Abstract
Immune-regulatory mechanisms are used by cancer to hide from the immune system. Advances and in-depth understanding of the biology of melanoma and its interaction with the immune system have led to the development of some of antagonistic antibodies to the programmed death 1 pathway (PD-1) and one of its ligands, programmed death ligand 1 (PD-L1), which are demonstrating high clinical benefit rates and tolerability. Blocking the immune-regulatory checkpoints that limit T-cell responses to melanoma upon PD-1/PD-L1 modulation has provided clinically validated targets for cancer immunotherapy. Combinations with other anti-melanoma agents may result in additional benefits. Nivolumab, pembrolizumab (formerly known as MK-3475 and lambrolizumab), and pidilizumab are anti-PD-1 antibodies in clinical development for melanoma, non-small cell lung cancer, renal cell carcinoma, head and neck cancers, lymphoma, and several other cancers. Long-term survivors already have been reported with these therapies. In this review, we discuss the current state of anti-PD-1 agents, the evidence in the literature to support the combination of anti-PD-1 antibodies with other anti-cancer agents and discuss the future directions for rational design of clinical trials that keep on increasing the number of long-term survivors.
Collapse
Affiliation(s)
- Blanca Homet Moreno
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), CA
| | - Giulia Parisi
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), CA; Department of Oncology, Division of Medical Oncology and Immunotherapy, University Hospital of Siena, Italy
| | - Lidia Robert
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), CA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California Los Angeles (UCLA), CA; Department of Surgery, University of California Los Angeles (UCLA), Los Angeles, CA; Department of Medical and Molecular Pharmacology, University of California Los Angeles (UCLA), Los Angeles, CA; Jonsson Comprehensive Cancer Center (JCCC) at UCLA, Los Angeles, CA.
| |
Collapse
|
656
|
Higgins GS, O'Cathail SM, Muschel RJ, McKenna WG. Drug radiotherapy combinations: review of previous failures and reasons for future optimism. Cancer Treat Rev 2015; 41:105-13. [PMID: 25579753 DOI: 10.1016/j.ctrv.2014.12.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 12/22/2014] [Accepted: 12/29/2014] [Indexed: 12/14/2022]
Abstract
Combining chemotherapy with radiotherapy has resulted in significant clinical improvements in many different tumour types. However, the non-specific mechanisms by which these drugs exert their effects mean that this is often at the expense of increased side effects. Previous attempts at using targeted drugs to induce more tumour specific radiosensitisation have been generally disappointing. Although cetuximab, an EGFR monoclonal antibody, resulted in improved overall survival in HNSCC when combined with radiotherapy, it has failed to show benefit when added to chemo-radiotherapy. In addition, our inability to successfully use drug treatments to reverse tumour hypoxia is underlined by the fact that no such treatment is currently in widespread clinical use. The reasons for these failures include the lack of robust biomarkers, and the previous use of drugs with unacceptable side-effect profiles. Despite these disappointments, there is reason for optimism. Our improved understanding of key signal transduction pathways and of tumour specific DNA repair deficiencies has produced new opportunities to specifically radiosensitise tumours. Novel strategies to reduce tumour hypoxia include the use of drugs that cause vascular normalisation and drugs that reduce tumour oxygen consumption. These new strategies, combined with better compounds at our disposal, and an ability to learn from our previous mistakes, mean that there is great promise for future drug-radiotherapy combinations to result in significant clinical benefits.
Collapse
Affiliation(s)
- Geoff S Higgins
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Department of Oncology, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Trust, Department of Oncology, Churchill Hospital, Oxford, UK.
| | - Sean M O'Cathail
- Oxford University Hospitals NHS Trust, Department of Oncology, Churchill Hospital, Oxford, UK
| | - Ruth J Muschel
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Department of Oncology, University of Oxford, Oxford, UK
| | - W Gillies McKenna
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, Department of Oncology, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Trust, Department of Oncology, Churchill Hospital, Oxford, UK
| |
Collapse
|
657
|
Haikerwal SJ, Hagekyriakou J, MacManus M, Martin OA, Haynes NM. Building immunity to cancer with radiation therapy. Cancer Lett 2015; 368:198-208. [PMID: 25592036 DOI: 10.1016/j.canlet.2015.01.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/14/2022]
Abstract
Over the last decade there has been a dramatic shift in the focus of cancer research toward understanding how the body's immune defenses can be harnessed to promote the effectiveness of cytotoxic anti-cancer therapies. The ability of ionizing radiation to elicit anti-cancer immune responses capable of controlling tumor growth has led to the emergence of promising combination-based radio-immunotherapeutic strategies for the treatment of cancer. Herein we review the immunoadjuvant properties of localized radiation therapy and discuss how technological advances in radio-oncology and developments in the field of tumor-immunotherapy have started to revolutionize the therapeutic application of radiotherapy.
Collapse
Affiliation(s)
- Suresh J Haikerwal
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Jim Hagekyriakou
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Michael MacManus
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia
| | - Olga A Martin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia; Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Nicole M Haynes
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia.
| |
Collapse
|
658
|
Calinescu AA, Kamran N, Baker G, Mineharu Y, Lowenstein PR, Castro MG. Overview of current immunotherapeutic strategies for glioma. Immunotherapy 2015; 7:1073-104. [PMID: 26598957 PMCID: PMC4681396 DOI: 10.2217/imt.15.75] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last decade, numerous studies of immunotherapy for malignant glioma (glioblastoma multiforme) have brought new knowledge and new hope for improving the prognosis of this incurable disease. Some clinical trials have reached Phase III, following positive outcomes in Phase I and II, with respect to safety and immunological end points. Results are encouraging especially when considering the promise of sustained efficacy by inducing antitumor immunological memory. Progress in understanding the mechanisms of tumor-induced immune suppression led to the development of drugs targeting immunosuppressive checkpoints, which are used in active clinical trials for glioblastoma multiforme. Insights related to the heterogeneity of the disease bring new challenges for the management of glioma and underscore a likely cause of therapeutic failure. An emerging therapeutic strategy is represented by a combinatorial, personalized approach, including the standard of care: surgery, radiation, chemotherapy with added active immunotherapy and multiagent targeting of immunosuppressive checkpoints.
Collapse
Affiliation(s)
| | - Neha Kamran
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Gregory Baker
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University, Kyoto, Japan
| | - Pedro Ricardo Lowenstein
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria Graciela Castro
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
659
|
Sharabi AB, Nirschl CJ, Kochel CM, Nirschl TR, Francica BJ, Velarde E, Deweese TL, Drake CG. Stereotactic Radiation Therapy Augments Antigen-Specific PD-1-Mediated Antitumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol Res 2014; 3:345-55. [PMID: 25527358 DOI: 10.1158/2326-6066.cir-14-0196] [Citation(s) in RCA: 526] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/03/2014] [Indexed: 02/06/2023]
Abstract
The immune-modulating effects of radiotherapy (XRT) have gained considerable interest recently, and there have been multiple reports of synergy between XRT and immunotherapy. However, additional preclinical studies are needed to demonstrate the antigen-specific nature of radiation-induced immune responses and elucidate potential mechanisms of synergy with immunotherapy. Here, we demonstrate the ability of stereotactic XRT to induce endogenous antigen-specific immune responses when it is combined with anti-PD-1 checkpoint blockade immunotherapy. Using the small animal radiation research platform (SARRP), image-guided stereotactic XRT delivered to B16-OVA melanoma or 4T1-HA breast carcinoma tumors resulted in the development of antigen-specific T cell- and B cell-mediated immune responses. These immune-stimulating effects of XRT were significantly increased when XRT was combined with either anti-PD-1 therapy or regulatory T cell (Treg) depletion, resulting in improved local tumor control. Phenotypic analyses of antigen-specific CD8 T cells revealed that XRT increased the percentage of antigen-experienced T cells and effector memory T cells. Mechanistically, we found that XRT upregulates tumor-associated antigen-MHC complexes, enhances antigen cross-presentation in the draining lymph node, and increases T-cell infiltration into tumors. These findings demonstrate the ability of XRT to prime an endogenous antigen-specific immune response and provide an additional mechanistic rationale for combining radiation with PD-1 blockade in the clinic.
Collapse
Affiliation(s)
- Andrew B Sharabi
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher J Nirschl
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christina M Kochel
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas R Nirschl
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian J Francica
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Theodore L Deweese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Drake
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland. The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
660
|
O'Brien MA, Power DG, Clover AJP, Bird B, Soden DM, Forde PF. Local tumour ablative therapies: Opportunities for maximising immune engagement and activation. Biochim Biophys Acta Rev Cancer 2014; 1846:510-23. [DOI: 10.1016/j.bbcan.2014.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/05/2014] [Accepted: 09/20/2014] [Indexed: 12/12/2022]
|
661
|
Tillner F, Thute P, Bütof R, Krause M, Enghardt W. Pre-clinical research in small animals using radiotherapy technology – a bidirectional translational approach. Z Med Phys 2014; 24:335-51. [DOI: 10.1016/j.zemedi.2014.07.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 01/17/2023]
|
662
|
Agassi AM, Myslicki FA, Shulman JM, Rotterman Y, Dosoretz DE, Fernandez E, Mantz CA, Finkelstein SE. The promise of combining radiation therapy and immunotherapy: morbidity and toxicity. Future Oncol 2014; 10:2319-28. [DOI: 10.2217/fon.14.188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Radiation therapy and immunotherapy in partnership may have the capability of delivering a therapeutic effect exceeding the sum of its parts. The possible relationship has been demonstrated in murine models and has been extended to a variety of clinical trials. Though the standard notion of whole body radiation therapy is immunosuppressive, there is growing evidence toward the contrary for focal radiation therapy. Furthermore, if immunotherapeutic techniques can retune the immune system against cancerous cells, they should have obvious benefits for advanced treatments moving forward. Herein, we explore the promise in combining radiation therapy and immunotherapy with distinct focus on potential morbidities and toxicities through analysis of completed clinical trials.
Collapse
Affiliation(s)
- Andre M Agassi
- 21st Century Oncology Translational Research Consortium, 7340 E Thomas Road, Scottsdale, AZ, USA
| | - Francisco A Myslicki
- 21st Century Oncology Translational Research Consortium, 7340 E Thomas Road, Scottsdale, AZ, USA
| | - Jesse M Shulman
- 21st Century Oncology Translational Research Consortium, 7340 E Thomas Road, Scottsdale, AZ, USA
| | - Yosef Rotterman
- 21st Century Oncology Translational Research Consortium, 7340 E Thomas Road, Scottsdale, AZ, USA
| | - Daniel E Dosoretz
- 21st Century Oncology Translational Research Consortium, Fort Myers, FL, USA
| | - Eduardo Fernandez
- 21st Century Oncology Translational Research Consortium, Plantation, FL, USA
| | - Constantine A Mantz
- 21st Century Oncology Translational Research Consortium, Fort Myers, FL, USA
| | - Steven E Finkelstein
- 21st Century Oncology Translational Research Consortium, 7340 E Thomas Road, Scottsdale, AZ, USA
| |
Collapse
|
663
|
Platten M, Ochs K, Lemke D, Opitz C, Wick W. Microenvironmental clues for glioma immunotherapy. Curr Neurol Neurosci Rep 2014; 14:440. [PMID: 24604058 DOI: 10.1007/s11910-014-0440-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gliomas have been viewed for decades as inaccessible for a meaningful antitumor immune response as they grow in a sanctuary site protected from infiltrating immune cells. Moreover, the glioma microenvironment constitutes a hostile environment for an efficient antitumor immune response as glioma-derived factors such as transforming growth factor β and catabolites of the essential amino acid tryptophan paralyze T-cell function. There is growing evidence from preclinical and clinical studies that a meaningful antitumor immunity exists in glioma patients and that it can be activated by vaccination strategies. As a consequence, the concept of glioma immunotherapy appears to be experiencing a renaissance with the first phase 3 randomized immunotherapy trials entering the clinical arena. On the basis of encouraging results from other tumor entities using immunostimulatory approaches by blocking endogenous T-cell suppressive pathways mediated by cytotoxic T-lymphocyte antigen 4 or programmed cell death protein 1/programmed cell death protein 1 ligand 1 with humanized antibodies, there is now a realistic and promising option to combine active immunotherapy with agents blocking the immunosuppressive microenvironment in patients with gliomas to allow a peripheral antitumor immune response induced by vaccination to become effective. Here we review the current clinical and preclinical evidence of antimicroenvironment immunotherapeutic strategies in gliomas.
Collapse
Affiliation(s)
- Michael Platten
- Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, German Cancer Consortium (DKTK) Clinical Cooperation Units, Im Neuenheimer Feld, Heidelberg, Germany,
| | | | | | | | | |
Collapse
|
664
|
Berghoff AS, Ricken G, Widhalm G, Rajky O, Dieckmann K, Birner P, Bartsch R, Höller C, Preusser M. Tumour-infiltrating lymphocytes and expression of programmed death ligand 1 (PD-L1) in melanoma brain metastases. Histopathology 2014; 66:289-99. [DOI: 10.1111/his.12537] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 08/22/2014] [Indexed: 11/29/2022]
Affiliation(s)
- Anna Sophie Berghoff
- Department of Medicine I; Medical University of Vienna; Vienna Austria
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
| | - Gerda Ricken
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
- Institute of Neurology; Medical University of Vienna; Vienna Austria
| | - Georg Widhalm
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
- Department of Neurosurgery; Medical University of Vienna; Vienna Austria
| | - Orsolya Rajky
- Department of Medicine I; Medical University of Vienna; Vienna Austria
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
| | - Karin Dieckmann
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
- Department of Radiotherapy; Medical University of Vienna; Vienna Austria
| | - Peter Birner
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
- Institute of Clinical Pathology; Medical University of Vienna; Vienna Austria
| | - Rupert Bartsch
- Department of Medicine I; Medical University of Vienna; Vienna Austria
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
| | - Christoph Höller
- Department of Dermatology; Medical University of Vienna; Vienna Austria
| | - Matthias Preusser
- Department of Medicine I; Medical University of Vienna; Vienna Austria
- Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
| |
Collapse
|
665
|
|
666
|
Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wöhrer A, Dieckmann K, Filipits M, Brandstetter A, Weller M, Kurscheid S, Hegi ME, Zielinski CC, Marosi C, Hainfellner JA, Preusser M, Wick W. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol 2014; 17:1064-75. [PMID: 25355681 DOI: 10.1093/neuonc/nou307] [Citation(s) in RCA: 448] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/04/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors targeting programmed cell death 1 (PD1) or its ligand (PD-L1) showed activity in several cancer types. METHODS We performed immunohistochemistry for CD3, CD8, CD20, HLA-DR, phosphatase and tensin homolog (PTEN), PD-1, and PD-L1 and pyrosequencing for assessment of the O6-methylguanine-methyltransferase (MGMT) promoter methylation status in 135 glioblastoma specimens (117 initial resection, 18 first local recurrence). PD-L1 gene expression was analyzed in 446 cases from The Cancer Genome Atlas. RESULTS Diffuse/fibrillary PD-L1 expression of variable extent, with or without interspersed epithelioid tumor cells with membranous PD-L1 expression, was observed in 103 of 117 (88.0%) newly diagnosed and 13 of 18 (72.2%) recurrent glioblastoma specimens. Sparse-to-moderate density of tumor-infiltrating lymphocytes (TILs) was found in 85 of 117 (72.6%) specimens (CD3+ 78/117, 66.7%; CD8+ 52/117, 44.4%; CD20+ 27/117, 23.1%; PD1+ 34/117, 29.1%). PD1+ TIL density correlated positively with CD3+ (P < .001), CD8+ (P < .001), CD20+ TIL density (P < .001), and PTEN expression (P = .035). Enrichment of specimens with low PD-L1 gene expression levels was observed in the proneural and G-CIMP glioblastoma subtypes and in specimens with high PD-L1 gene expression in the mesenchymal subtype (P = 5.966e-10). No significant differences in PD-L1 expression or TIL density between initial and recurrent glioblastoma specimens or correlation of PD-L1 expression or TIL density with patient age or outcome were evident. CONCLUSION TILs and PD-L1 expression are detectable in the majority of glioblastoma samples but are not related to outcome. Because the target is present, a clinical study with specific immune checkpoint inhibitors seems to be warranted in glioblastoma.
Collapse
Affiliation(s)
- Anna Sophie Berghoff
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Barbara Kiesel
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Georg Widhalm
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Orsolya Rajky
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Gerda Ricken
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Adelheid Wöhrer
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Karin Dieckmann
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Martin Filipits
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Anita Brandstetter
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Michael Weller
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Sebastian Kurscheid
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Monika E Hegi
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Christoph C Zielinski
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Christine Marosi
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Johannes A Hainfellner
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Matthias Preusser
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| | - Wolfgang Wick
- Institute of Neurology, Medical University of Vienna, Vienna, Austria (A.S.B., G.R., A.W., J.A.H.); Department of Medicine I, Medical University of Vienna, Vienna, Austria (A.S.B., O.R., M.F., A.B., C.C.Z., C.M., M.P.); Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria (A.S.B., B.K., G.W., O.R., G.R., A.W., K.D., M.F., A.B., C.C.Z., C.M., J.A.H., M.P.); Department of Neurosurgery, Medical University of Vienna, Vienna, Austria (B.K., G.W.); Department of Radiotherapy, Medical University of Vienna, Vienna, Austria (K.D.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.); Laboratory of Brain Tumor Biology and Genetics, Service of Neurosurgery, Department of Clinical Neurosciences, University Hospital Lausanne (CHUV), Lausanne, Switzerland (S.K., M.E.H.); Neurology Clinic and National Center for Tumor Disease, University of Heidelberg, Heidelberg, Germany (W.W.); Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany (W.W.)
| |
Collapse
|
667
|
Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, Cheng Y, Kim JW, Qiao J, Zhang L, Han Y, Lesniak MS. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res 2014; 20:5290-301. [PMID: 24691018 PMCID: PMC4182350 DOI: 10.1158/1078-0432.ccr-14-0514] [Citation(s) in RCA: 460] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common form of malignant glioma in adults. Although protected by both the blood-brain and blood-tumor barriers, GBMs are actively infiltrated by T cells. Previous work has shown that IDO, CTLA-4, and PD-L1 are dominant molecular participants in the suppression of GBM immunity. This includes IDO-mediated regulatory T-cell (Treg; CD4(+)CD25(+)FoxP3(+)) accumulation, the interaction of T-cell-expressed, CTLA-4, with dendritic cell-expressed, CD80, as well as the interaction of tumor- and/or macrophage-expressed, PD-L1, with T-cell-expressed, PD-1. The individual inhibition of each pathway has been shown to increase survival in the context of experimental GBM. However, the impact of simultaneously targeting all three pathways in brain tumors has been left unanswered. EXPERIMENTAL DESIGN AND RESULTS In this report, we demonstrate that, when dually challenged, IDO-deficient tumors provide a selectively competitive survival advantage against IDO-competent tumors. Next, we provide novel observations regarding tryptophan catabolic enzyme expression, before showing that the therapeutic inhibition of IDO, CTLA-4, and PD-L1 in a mouse model of well-established glioma maximally decreases tumor-infiltrating Tregs, coincident with a significant increase in T-cell-mediated long-term survival. In fact, 100% of mice bearing intracranial tumors were long-term survivors following triple combination therapy. The expression and/or frequency of T cell expressed CD44, CTLA-4, PD-1, and IFN-γ depended on timing after immunotherapeutic administration. CONCLUSIONS Collectively, these data provide strong preclinical evidence that combinatorially targeting immunosuppression in malignant glioma is a strategy that has high potential value for future clinical trials in patients with GBM.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- Brain Neoplasms/drug therapy
- Brain Neoplasms/genetics
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- CTLA-4 Antigen/antagonists & inhibitors
- Cell Line, Tumor
- Dacarbazine/administration & dosage
- Dacarbazine/analogs & derivatives
- Dacarbazine/pharmacology
- Disease Models, Animal
- Drug Therapy, Combination
- Glioma/drug therapy
- Glioma/genetics
- Glioma/immunology
- Glioma/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Mice
- Mice, Knockout
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Temozolomide
Collapse
Affiliation(s)
- Derek A Wainwright
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Alan L Chang
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Mahua Dey
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Irina V Balyasnikova
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Chung Kwon Kim
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Alex Tobias
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Yu Cheng
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Julius W Kim
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jian Qiao
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Yu Han
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
668
|
Parikh F, Duluc D, Imai N, Clark A, Misiukiewicz K, Bonomi M, Gupta V, Patsias A, Parides M, Demicco EG, Zhang DY, Kim-Schulze S, Kao J, Gnjatic S, Oh S, Posner MR, Sikora AG. Chemoradiotherapy-induced upregulation of PD-1 antagonizes immunity to HPV-related oropharyngeal cancer. Cancer Res 2014; 74:7205-16. [PMID: 25320012 DOI: 10.1158/0008-5472.can-14-1913] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
While viral antigens in human papillomavirus (HPV)-related oropharyngeal cancer (HPVOPC) are attractive targets for immunotherapy, the effects of existing standard-of-care therapies on immune responses to HPV are poorly understood. We serially sampled blood from patients with stage III-IV oropharyngeal cancer undergoing concomitant chemoradiotherapy with or without induction chemotherapy. Circulating immunocytes including CD4(+) and CD8(+) T cells, regulatory T cells (Treg), and myeloid-derived suppressor cells (MDSC) were profiled by flow cytometry. Antigen-specific T-cell responses were measured in response to HPV16 E6 and E7 peptide pools. The role of PD-1 signaling in treatment-related immunosuppression was functionally defined by performing HPV-specific T-cell assays in the presence of blocking antibody. While HPV-specific T-cell responses were present in 13 of 18 patients before treatment, 10 of 13 patients lost these responses within 3 months after chemoradiotherapy. Chemoradiotherapy decreased circulating T cells and markedly elevated MDSCs. PD-1 expression on CD4(+) T cells increased by nearly 2.5-fold after chemoradiotherapy, and ex vivo culture with PD-1-blocking antibody enhanced HPV-specific T-cell responses in 8 of 18 samples tested. Chemoradiotherapy suppresses circulating immune responses in patients with HPVOPC by unfavorably altering effector:suppressor immunocyte ratios and upregulating PD-1 expression on CD4(+) T cells. These data strongly support testing of PD-1-blocking agents in combination with standard-of-care chemoradiotherapy for HPVOPC.
Collapse
Affiliation(s)
- Falguni Parikh
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Naoko Imai
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Amelia Clark
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Krzys Misiukiewicz
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marcello Bonomi
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Vishal Gupta
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexis Patsias
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Parides
- Health Evidence and Policy, Program, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elizabeth G Demicco
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - David Y Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Seunghee Kim-Schulze
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Johnny Kao
- Department of Radiation Oncology, Good Samaritan Hospital Medical Center, West Islip, New York
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sangkon Oh
- Baylor Institute of Immunology, Dallas, Texas
| | - Marshall R Posner
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrew G Sikora
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, New York. Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
669
|
Abstract
Radiation therapy and immunotherapy are both well-established treatments for malignant disease. Radiotherapy has long been utilized for purposes of providing local tumor control, and the recent success with novel immunomodulatory agents has brought immunotherapy into the forefront of clinical practice for the treatment of many tumor types. Although radiotherapy has traditionally been thought to mediate tumor regression through direct cytotoxic effects, it is now known that radiation also alters the local tumor microenvironment with effects on both the local and systemic anti-tumor immune response. There is growing evidence that the rational integration of the immunomodulatory effects of radiotherapy with the expanding armamentarium of clinically approved immunotherapeutics can yield potent anti-tumor responses exceeding the benefit of either therapy alone. Here we summarize current approaches to the combination of immunotherapy with radiation therapy.
Collapse
Affiliation(s)
- Susan M Hiniker
- Department of Radiation Oncology, Stanford University, Stanford, CA.
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University, Stanford, CA
| |
Collapse
|
670
|
The future of glioblastoma therapy: synergism of standard of care and immunotherapy. Cancers (Basel) 2014; 6:1953-85. [PMID: 25268164 PMCID: PMC4276952 DOI: 10.3390/cancers6041953] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/05/2014] [Accepted: 09/03/2014] [Indexed: 12/18/2022] Open
Abstract
The current standard of care for glioblastoma (GBM) is maximal surgical resection with adjuvant radiotherapy and temozolomide (TMZ). As the 5-year survival with GBM remains at a dismal <10%, novel therapies are needed. Immunotherapies such as the dendritic cell (DC) vaccine, heat shock protein vaccines, and epidermal growth factor receptor (EGFRvIII) vaccines have shown encouraging results in clinical trials, and have demonstrated synergistic effects with conventional therapeutics resulting in ongoing phase III trials. Chemoradiation has been shown to have synergistic effects when used in combination with immunotherapy. Cytotoxic ionizing radiation is known to trigger pro-inflammatory signaling cascades and immune activation secondary to cell death, which can then be exploited by immunotherapies. The future of GBM therapeutics will involve finding the place for immunotherapy in the current treatment regimen with a focus on developing strategies. Here, we review current GBM therapy and the evidence for combination of immune checkpoint inhibitors, DC and peptide vaccines with the current standard of care.
Collapse
|
671
|
Amer MH. Gene therapy for cancer: present status and future perspective. MOLECULAR AND CELLULAR THERAPIES 2014; 2:27. [PMID: 26056594 PMCID: PMC4452068 DOI: 10.1186/2052-8426-2-27] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/22/2014] [Indexed: 12/21/2022]
Abstract
Advancements in human genomics over the last two decades have shown that cancer is mediated by somatic aberration in the host genome. This discovery has incited enthusiasm among cancer researchers; many now use therapeutic approaches in genetic manipulation to improve cancer regression and find a potential cure for the disease. Such gene therapy includes transferring genetic material into a host cell through viral (or bacterial) and non-viral vectors, immunomodulation of tumor cells or the host immune system, and manipulation of the tumor microenvironment, to reduce tumor vasculature or to increase tumor antigenicity for better recognition by the host immune system. Overall, modest success has been achieved with relatively minimal side effects. Previous approaches to cancer treatment, such as retrovirus integration into the host genome with the risk of mutagenesis and second malignancies, immunogenicity against the virus and/or tumor, and resistance to treatment with disease relapse, have markedly decreased with the new generation of viral and non-viral vectors. Several tumor-specific antibodies and genetically modified immune cells and vaccines have been developed, yet few are presently commercially available, while many others are still ongoing in clinical trials. It is anticipated that gene therapy will play an important role in future cancer therapy as part of a multimodality treatment, in combination with, or following other forms of cancer therapy, such as surgery, radiation and chemotherapy. The type and mode of gene therapy will be determined based on an individual's genomic constituents, as well as his or her tumor specifics, genetics, and host immune status, to design a multimodality treatment that is unique to each individual's specific needs.
Collapse
Affiliation(s)
- Magid H Amer
- Department of Medicine, St Rita’s Medical Center, 825 West Market Street, Suite #203, Lima, OH 45805 USA
| |
Collapse
|
672
|
Amer MH. Gene therapy for cancer: present status and future perspective. MOLECULAR AND CELLULAR THERAPIES 2014; 2:27. [PMID: 26056594 PMCID: PMC4452068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 08/22/2014] [Indexed: 11/21/2023]
Abstract
Advancements in human genomics over the last two decades have shown that cancer is mediated by somatic aberration in the host genome. This discovery has incited enthusiasm among cancer researchers; many now use therapeutic approaches in genetic manipulation to improve cancer regression and find a potential cure for the disease. Such gene therapy includes transferring genetic material into a host cell through viral (or bacterial) and non-viral vectors, immunomodulation of tumor cells or the host immune system, and manipulation of the tumor microenvironment, to reduce tumor vasculature or to increase tumor antigenicity for better recognition by the host immune system. Overall, modest success has been achieved with relatively minimal side effects. Previous approaches to cancer treatment, such as retrovirus integration into the host genome with the risk of mutagenesis and second malignancies, immunogenicity against the virus and/or tumor, and resistance to treatment with disease relapse, have markedly decreased with the new generation of viral and non-viral vectors. Several tumor-specific antibodies and genetically modified immune cells and vaccines have been developed, yet few are presently commercially available, while many others are still ongoing in clinical trials. It is anticipated that gene therapy will play an important role in future cancer therapy as part of a multimodality treatment, in combination with, or following other forms of cancer therapy, such as surgery, radiation and chemotherapy. The type and mode of gene therapy will be determined based on an individual's genomic constituents, as well as his or her tumor specifics, genetics, and host immune status, to design a multimodality treatment that is unique to each individual's specific needs.
Collapse
Affiliation(s)
- Magid H Amer
- Department of Medicine, St Rita’s Medical Center, 825 West Market Street, Suite #203, Lima, OH 45805 USA
| |
Collapse
|
673
|
Reardon DA, Freeman G, Wu C, Chiocca EA, Wucherpfennig KW, Wen PY, Fritsch EF, Curry WT, Sampson JH, Dranoff G. Immunotherapy advances for glioblastoma. Neuro Oncol 2014; 16:1441-58. [PMID: 25190673 DOI: 10.1093/neuonc/nou212] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Survival for patients with glioblastoma, the most common high-grade primary CNS tumor, remains poor despite multiple therapeutic interventions including intensifying cytotoxic therapy, targeting dysregulated cell signaling pathways, and blocking angiogenesis. Exciting, durable clinical benefits have recently been demonstrated for a number of other challenging cancers using a variety of immunotherapeutic approaches. Much modern research confirms that the CNS is immunoactive rather than immunoprivileged. Preliminary results of clinical studies demonstrate that varied vaccine strategies have achieved encouraging evidence of clinical benefit for glioblastoma patients, although multiple variables will likely require systematic investigation before optimal outcomes are realized. Initial preclinical studies have also revealed promising results with other immunotherapies including cell-based approaches and immune checkpoint blockade. Clinical studies to evaluate a wide array of immune therapies for malignant glioma patients are being rapidly developed. Important considerations going forward include optimizing response assessment and identifiying correlative biomarkers for predict therapeutic benefit. Finally, the potential of complementary combinatorial immunotherapeutic regimens is highly exciting and warrants expedited investigation.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Gordon Freeman
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Catherine Wu
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - E Antonio Chiocca
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Kai W Wucherpfennig
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Edward F Fritsch
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - William T Curry
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - John H Sampson
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Glenn Dranoff
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| |
Collapse
|
674
|
Zietman A. The Red Journal's Top Downloads of 2013. Int J Radiat Oncol Biol Phys 2014; 89:937-939. [DOI: 10.1016/j.ijrobp.2014.04.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 04/30/2014] [Accepted: 04/30/2014] [Indexed: 12/25/2022]
|
675
|
Lin NU, Amiri-Kordestani L, Palmieri D, Liewehr DJ, Steeg PS. CNS metastases in breast cancer: old challenge, new frontiers. Clin Cancer Res 2014; 19:6404-18. [PMID: 24298071 DOI: 10.1158/1078-0432.ccr-13-0790] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Despite major therapeutic advances in the management of patients with breast cancer, central nervous system (CNS) metastases remain an intractable problem, particularly in patients with metastatic HER2-positive and triple-negative breast cancer. As systemic therapies to treat extracranial disease improve, some patients are surviving longer, and the frequency of CNS involvement seems to be increasing. Furthermore, in the early-stage setting, the CNS remains a potential sanctuary site for relapse. This review highlights advances in the development of biologically relevant preclinical models, including the development of brain-tropic cell lines for testing of agents to prevent and treat brain metastases, and summarizes our current understanding of the biology of CNS relapse. From a clinical perspective, a variety of therapeutic approaches are discussed, including methods to improve drug delivery, novel cytotoxic agents, and targeted therapies. Challenges in current trial design and endpoints are reviewed. Finally, we discuss promising new directions, including novel trial designs, correlative imaging techniques, and enhanced translational opportunities.
Collapse
Affiliation(s)
- Nancy U Lin
- Authors' Affiliations: Dana-Farber Cancer Institute, Boston, Massachusetts; Medical Oncology Branch, Center for Cancer Research, National Cancer Institute; Women's Cancers Section, Laboratory of Molecular Pharmacology; and Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | | | | |
Collapse
|
676
|
Belcaid Z, Phallen JA, Zeng J, See AP, Mathios D, Gottschalk C, Nicholas S, Kellett M, Ruzevick J, Jackson C, Albesiano E, Durham NM, Ye X, Tran PT, Tyler B, Wong JW, Brem H, Pardoll DM, Drake CG, Lim M. Focal radiation therapy combined with 4-1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS One 2014; 9:e101764. [PMID: 25013914 PMCID: PMC4094423 DOI: 10.1371/journal.pone.0101764] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/11/2014] [Indexed: 01/19/2023] Open
Abstract
Background Glioblastoma (GBM) is the most common malignant brain tumor in adults and is associated with a poor prognosis. Cytotoxic T lymphocyte antigen -4 (CTLA-4) blocking antibodies have demonstrated an ability to generate robust antitumor immune responses against a variety of solid tumors. 4-1BB (CD137) is expressed by activated T lymphocytes and served as a co-stimulatory signal, which promotes cytotoxic function. Here, we evaluate a combination immunotherapy regimen involving 4-1BB activation, CTLA-4 blockade, and focal radiation therapy in an immune-competent intracranial GBM model. Methods GL261-luciferace cells were stereotactically implanted in the striatum of C57BL/6 mice. Mice were treated with a triple therapy regimen consisted of 4-1BB agonist antibodies, CTLA-4 blocking antibodies, and focal radiation therapy using a small animal radiation research platform and mice were followed for survival. Numbers of brain-infiltrating lymphocytes were analyzed by FACS analysis. CD4 or CD8 depleting antibodies were administered to determine the relative contribution of T helper and cytotoxic T cells in this regimen. To evaluate the ability of this immunotherapy to generate an antigen-specific memory response, long-term survivors were re-challenged with GL261 glioma en B16 melanoma flank tumors. Results Mice treated with triple therapy had increased survival compared to mice treated with focal radiation therapy and immunotherapy with 4-1BB activation and CTLA-4 blockade. Animals treated with triple therapy exhibited at least 50% long-term tumor free survival. Treatment with triple therapy resulted in a higher density of CD4+ and CD8+ tumor infiltrating lymphocytes. Mechanistically, depletion of CD4+ T cells abrogated the antitumor efficacy of triple therapy, while depletion of CD8+ T cells had no effect on the treatment response. Conclusion Combination therapy with 4-1BB activation and CTLA-4 blockade in the setting of focal radiation therapy improves survival in an orthotopic mouse model of glioma by a CD4+ T cell dependent mechanism and generates antigen-specific memory.
Collapse
Affiliation(s)
- Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jillian A. Phallen
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jing Zeng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Alfred P. See
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Chelsea Gottschalk
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Sarah Nicholas
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Meghan Kellett
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jacob Ruzevick
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Christopher Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Emilia Albesiano
- Department of Oncology and Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Nicholas M. Durham
- Department of Oncology and Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Phuoc T. Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John W. Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Departments of Oncology, Ophthalmology, and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Drew M. Pardoll
- Department of Oncology and Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Charles G. Drake
- Department of Oncology and Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
677
|
Almo SC, Guha C. Considerations for combined immune checkpoint modulation and radiation treatment. Radiat Res 2014; 182:230-8. [PMID: 25003312 DOI: 10.1667/rr13667.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent advances indicate that new therapeutic strategies for the treatment of malignancies will be realized from combined radiation treatment and immune checkpoint modulation. Numerous biophysical properties must be considered for effective biologic development, including affinity, selectivity, oligomeric state and valency. High-resolution structural characterization contributes to our understanding of these properties and can lead to the realization of proteins with unique in vitro activities and novel in vivo therapeutic functions. In this article we focus on the importance of these factors for new potential biologics and consider these in the context of combination therapies with physical modalities, including radiation therapy. In particular, we examine the consequences of altered avidities and subset-specific ligand density on the rational modification of biological function in the immunoglobulin and tumor necrosis factor superfamilies and for new optimized combination therapies.
Collapse
Affiliation(s)
- Steven C Almo
- a Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
678
|
Zandberg DP, Strome SE. The role of the PD-L1:PD-1 pathway in squamous cell carcinoma of the head and neck. Oral Oncol 2014; 50:627-32. [DOI: 10.1016/j.oraloncology.2014.04.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/11/2014] [Accepted: 04/13/2014] [Indexed: 01/02/2023]
|
679
|
Abstract
Conventional therapy for malignant glioma (MG) fails to specifically eliminate tumor cells, resulting in toxicity that limits therapeutic efficacy. In contrast, antibody-based immunotherapy uses the immune system to eliminate tumor cells with exquisite specificity. Increased understanding of the pathobiology of MG and the profound immunosuppression present among patients with MG has revealed several biologic targets amenable to antibody-based immunotherapy. Novel antibody engineering techniques allow for the production of fully human antibodies or antibody fragments with vastly reduced antigen-binding dissociation constants, increasing safety when used clinically as therapeutics. In this report, we summarize the use of antibody-based immunotherapy for MG. Approaches currently under investigation include the use of antibodies or antibody fragments to: (1) redirect immune effector cells to target tumor mutations, (2) inhibit immunosuppressive signals and thereby stimulate an immunological response against tumor cells, and (3) provide costimulatory signals to evoke immunologic targeting of tumor cells. These approaches demonstrate highly compelling safety and efficacy for the treatment of MG, providing a viable adjunct to current standard-of-care therapy for MG.
Collapse
Affiliation(s)
- Patrick C Gedeon
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC.
| | - Katherine A Riccione
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC
| | - Peter E Fecci
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - John H Sampson
- Duke Brain Tumor Immunotherapy Program, Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC
| |
Collapse
|
680
|
Abstract
Glioblastoma is the most common and most aggressive primary brain tumor in adults. Optimized standard treatment only confers a modest improvement in progression and overall survival, underscoring the pressing need for the development of novel therapies. Our understanding of glioblastoma (a molecularly heterogeneous disorder) has been accelerated in the setting of large scale genomic analyses, lending insight into potential actionable targets. Antiangiogenic therapies have been used in the treatment of glioblastoma, and our understanding of the means to optimize the role of these agents is continuing to evolve. Recently, immunotherapy has garnered increasing attention as a therapeutic approach in the treatment of gliomas. Promising novel approaches are under active development in the treatment of glioblastoma.
Collapse
|
681
|
Bambury RM, Morris PG. The search for novel therapeutic strategies in the treatment of recurrent glioblastoma multiforme. Expert Rev Anticancer Ther 2014; 14:955-64. [PMID: 24814143 DOI: 10.1586/14737140.2014.916214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with ≤10% patients surviving 5 years from the time of diagnosis. After tumor progression on frontline therapy with concomitant chemoradiotherapy followed by consolidation temozolomide there are few effective treatment options. Bevacizumab and nitrosureas are the most commonly used systemic options in this instance but no overall survival benefit has been demonstrated. In this review we outline the major avenues of research for treatment of recurrent GBM including anti-angiogenic, signaling pathway blockade and immunotherapy approaches. Results of recent trials as well as pertinent ongoing studies are discussed. Enrollment of patients to clinical trials as well as incorporation of correlative translational science studies to identify predictive biomarkers of treatment response will be key to improving outcomes in this devastating disease.
Collapse
Affiliation(s)
- Richard M Bambury
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue New York, NY 10065, USA
| | | |
Collapse
|
682
|
Jackson CM, Lim M, Drake CG. Immunotherapy for brain cancer: recent progress and future promise. Clin Cancer Res 2014; 20:3651-9. [PMID: 24771646 DOI: 10.1158/1078-0432.ccr-13-2057] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immunotherapy is emerging as the newest pillar of cancer treatment, with the potential to assume a place alongside surgical debulking, radiotherapy, and chemotherapy. Early experiences with antitumor vaccines demonstrated the feasibility and potential efficacy of this approach, and newer agents, such as immune checkpoint blocking antibodies and modern vaccine platforms, have ushered in a new era. These efforts are headlined by work in melanoma, prostate cancer, and renal cell carcinoma; however, substantial progress has been achieved in a variety of other cancers, including high-grade gliomas. A recurrent theme of this work is that immunotherapy is not a one-size-fits-all solution. Rather, dynamic, tumor-specific interactions within the tumor microenvironment continually shape the immunologic balance between tumor elimination and escape. High-grade gliomas are a particularly fascinating example. These aggressive, universally fatal tumors are highly resistant to radiotherapy and chemotherapy and inevitably recur after surgical resection. Located in the immune-privileged central nervous system, high-grade gliomas also use an array of defenses that serve as direct impediments to immune attack. Despite these challenges, vaccines have shown activity against high-grade gliomas, and anecdotal, preclinical, and early clinical data bolster the notion that durable remission is possible with immunotherapy. Realizing this potential, however, will require an approach tailored to the unique aspects of glioma biology.
Collapse
Affiliation(s)
| | - Michael Lim
- Authors' Affiliations: Departments of Neurosurgery and
| | - Charles G Drake
- Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
683
|
Barker CA, Postow MA. Combinations of radiation therapy and immunotherapy for melanoma: a review of clinical outcomes. Int J Radiat Oncol Biol Phys 2014; 88:986-97. [PMID: 24661650 PMCID: PMC4667362 DOI: 10.1016/j.ijrobp.2013.08.035] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/19/2013] [Accepted: 08/26/2013] [Indexed: 01/12/2023]
Abstract
Radiation therapy has long played a role in the management of melanoma. Recent advances have also demonstrated the efficacy of immunotherapy in the treatment of melanoma. Preclinical data suggest a biologic interaction between radiation therapy and immunotherapy. Several clinical studies corroborate these findings. This review will summarize the outcomes of studies reporting on patients with melanoma treated with a combination of radiation therapy and immunotherapy. Vaccine therapies often use irradiated melanoma cells, and may be enhanced by radiation therapy. The cytokines interferon-α and interleukin-2 have been combined with radiation therapy in several small studies, with some evidence suggesting increased toxicity and/or efficacy. Ipilimumab, a monoclonal antibody which blocks cytotoxic T-lymphocyte antigen-4, has been combined with radiation therapy in several notable case studies and series. Finally, pilot studies of adoptive cell transfer have suggested that radiation therapy may improve the efficacy of treatment. The review will demonstrate that the combination of radiation therapy and immunotherapy has been reported in several notable case studies, series and clinical trials. These clinical results suggest interaction and the need for further study.
Collapse
Affiliation(s)
- Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York.
| | - Michael A Postow
- Department of Medicine, Melanoma and Sarcoma Oncology Service, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
684
|
Spary LK, Al-Taei S, Salimu J, Cook AD, Ager A, Watson HA, Clayton A, Staffurth J, Mason MD, Tabi Z. Enhancement of T cell responses as a result of synergy between lower doses of radiation and T cell stimulation. THE JOURNAL OF IMMUNOLOGY 2014; 192:3101-10. [PMID: 24600032 DOI: 10.4049/jimmunol.1302736] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
As a side effect of cancer radiotherapy, immune cells receive varying doses of radiation. Whereas high doses of radiation (>10 Gy) can lead to lymphopenia, lower radiation doses (2-4 Gy) represent a valid treatment option in some hematological cancers, triggering clinically relevant immunological changes. Based on our earlier observations, we hypothesized that lower radiation doses have a direct positive effect on T cells. In this study, we show that 0.6-2.4 Gy radiation enhances proliferation and IFN-γ production of PBMC or purified T cells induced by stimulation via the TCR. Radiation with 1.2 Gy also lowered T cell activation threshold and broadened the Th1 cytokine profile. Although radiation alone did not activate T cells, when followed by TCR stimulation, ERK1/2 and Akt phosphorylation increased above that induced by stimulation alone. These changes were followed by an early increase in glucose uptake. Naive (CD45RA(+)) or memory (CD45RA(-)) T cell responses to stimulation were boosted at similar rates by radiation. Whereas increased Ag-specific cytotoxic activity of a CD8(+) T cell line manifested in a 4-h assay (10-20% increase), highly significant (5- to 10-fold) differences in cytokine production were detected in 6-d Ag-stimulation assays of PBMC, probably as a net outcome of death of nonstimulated and enhanced response of Ag-stimulated T cells. T cells from patients receiving pelvic radiation (2.2-2.75 Gy) also displayed increased cytokine production when stimulated in vitro. We report in this study enhanced T cell function induced by synergistic radiation treatment, with potential physiological significance in a wide range of T cell responses.
Collapse
Affiliation(s)
- Lisa K Spary
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Whitchurch, Cardiff CF14 2TL, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
685
|
|
686
|
Reiss KA, Forde PM, Brahmer JR. Harnessing the power of the immune system via blockade of PD-1 and PD-L1: a promising new anticancer strategy. Immunotherapy 2014; 6:459-75. [PMID: 24815784 PMCID: PMC4732706 DOI: 10.2217/imt.14.9] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cancer cells employ several mechanisms to evade the immune system of their host, thus escaping immune recognition and elimination. Of particular interest is a cancer cell's ability to co-opt the immune system's innate ligands and inhibitory receptors (also known as checkpoints), thus creating an immunosuppressive microenvironment that downregulates T-cell activation and cell signaling. The recent development of the checkpoint inhibitors anti-programmed death-1 and anti-programmed death ligand-1 has generated an enormous amount of interest as a potential new anticancer strategy in solid tumors, particularly in non-small-cell lung cancer, renal cell carcinoma and melanoma. Data suggest significant disease response rates using anti-programmed death-1 and anti-programmed death ligand-1 antibodies, even in heavily pretreated patients. Future directions include optimization of drug delivery sequence and combination of immunotherapy with other therapies including cytotoxic chemotherapy, radiation, antiangiogenic agents and small-molecule tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Kim A Reiss
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room 186, 401 North Broadway Street, Baltimore, 21287, USA
| | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room 186, 401 North Broadway Street, Baltimore, 21287, USA
| | - Julie R Brahmer
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room G94, 401 North Broadway Street, Baltimore, 21287, USA
| |
Collapse
|
687
|
Wei B, Wang L, Zhao X, Du C, Guo Y, Sun Z. The upregulation of programmed death 1 on peripheral blood T cells of glioma is correlated with disease progression. Tumour Biol 2013; 35:2923-9. [PMID: 24375192 DOI: 10.1007/s13277-013-1376-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/29/2013] [Indexed: 12/31/2022] Open
Abstract
Glioma is the most common primary brain tumor. Programmed death 1 (PD-1) is a surface receptor expressed on activated and exhausted T cells, which mediate T cell inhibition upon binding with its ligand. In the current study, we investigated the expression of PD-1 on peripheral CD4+ and CD8+ T cells in glioma patients. Percentage of PD-1+ cells was measured by flow cytometry in 86 glioma cases and 62 healthy controls. Results showed that PD-1 expression was significantly increased in both peripheral CD4+ and CD8+ T cells in glioma (p < 0.001 and p < 0.001, respectively). When comparing PD-1 level in glioma patients with different histological types, patients with astrocytomas revealed clearly higher proportion of PD-1 on CD4+ T cells than those with oligodendrogliomas (p < 0.001), ependymomas (p < 0.001), or pilocytic astrocytomas (p < 0.001). Also, patients with the highest tumor grade (IV) demonstrated the most elevated expression of PD-1 on both CD4+ and CD8+ T cells. Interestingly, cases with tumor grade III and IV had downregulated PD-1 level on peripheral CD4+ T cells after surgery, whereas only grade IV patients showed decreased proportion of PD-1 on CD8+ T cells after treatment. In addition, no correlation between PD-1 expression and progression to secondary glioblastoma was observed. These data suggested PD-1 may act as a positive regulator in the pathogenesis and progression of glioma.
Collapse
Affiliation(s)
- Bo Wei
- The Second Division of Neurosurgery, The China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, Jilin, 130033, China
| | | | | | | | | | | |
Collapse
|
688
|
Wada S, Harris TJ, Tryggestad E, Yoshimura K, Zeng J, Yen HR, Getnet D, Grosso JF, Bruno TC, De Marzo AM, Netto GJ, Pardoll DM, DeWeese TL, Wong J, Drake CG. Combined treatment effects of radiation and immunotherapy: studies in an autochthonous prostate cancer model. Int J Radiat Oncol Biol Phys 2013; 87:769-76. [PMID: 24064321 PMCID: PMC4417352 DOI: 10.1016/j.ijrobp.2013.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 06/19/2013] [Accepted: 07/12/2013] [Indexed: 11/17/2022]
Abstract
PURPOSE To optimize the combination of ionizing radiation and cellular immunotherapy using a preclinical autochthonous model of prostate cancer. METHODS AND MATERIALS Transgenic mice expressing a model antigen under a prostate-specific promoter were treated using a platform that integrates cone-beam CT imaging with 3-dimensional conformal therapy. Using this technology we investigated the immunologic and therapeutic effects of combining ionizing radiation with granulocyte/macrophage colony-stimulating factor-secreting cellular immunotherapy for prostate cancer in mice bearing autochthonous prostate tumors. RESULTS The combination of ionizing radiation and immunotherapy resulted in a significant decrease in pathologic tumor grade and gross tumor bulk that was not evident with either single-modality therapy. Furthermore, combinatorial therapy resulted in improved overall survival in a preventive metastasis model and in the setting of established micrometastases. Mechanistically, combined therapy resulted in an increase of the ratio of effector-to-regulatory T cells for both CD4 and CD8 tumor-infiltrating lymphocytes. CONCLUSIONS Our preclinical model establishes a potential role for the use of combined radiation-immunotherapy in locally advanced prostate cancer, which warrants further exploration in a clinical setting.
Collapse
MESH Headings
- Adenocarcinoma/immunology
- Adenocarcinoma/mortality
- Adenocarcinoma/pathology
- Adenocarcinoma/therapy
- Adoptive Transfer/methods
- Animals
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Combined Modality Therapy/methods
- Combined Modality Therapy/mortality
- Cone-Beam Computed Tomography/methods
- Hemagglutinins/immunology
- Hemagglutinins/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/mortality
- Lymphocytes, Tumor-Infiltrating/cytology
- Male
- Mice
- Mice, Transgenic
- Neoplasm Grading
- Neoplasm Micrometastasis/prevention & control
- Organs at Risk/diagnostic imaging
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/mortality
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Radionuclide Imaging
- Radiotherapy Dosage
- Radiotherapy, Conformal/methods
- Radiotherapy, Conformal/mortality
- T-Lymphocytes, Helper-Inducer/cytology
- T-Lymphocytes, Regulatory/cytology
- Tumor Burden
- Urinary Bladder/diagnostic imaging
Collapse
Affiliation(s)
- Satoshi Wada
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Timothy J. Harris
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Erik Tryggestad
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Kiyoshi Yoshimura
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Jing Zeng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Hung-Rong Yen
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Derese Getnet
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Joseph F. Grosso
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Tullia C. Bruno
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - George J. Netto
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Pathology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Drew M. Pardoll
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Theodore L. DeWeese
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - John Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| | - Charles G. Drake
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287-2101
| |
Collapse
|
689
|
Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013; 14:1014-22. [PMID: 24048123 DOI: 10.1038/ni.2703] [Citation(s) in RCA: 3029] [Impact Index Per Article: 252.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023]
Abstract
Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.
Collapse
|
690
|
Zeng J, Harris TJ, Lim M, Drake CG, Tran PT. Immune modulation and stereotactic radiation: improving local and abscopal responses. BIOMED RESEARCH INTERNATIONAL 2013; 2013:658126. [PMID: 24324970 PMCID: PMC3845488 DOI: 10.1155/2013/658126] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 10/05/2013] [Accepted: 10/05/2013] [Indexed: 01/21/2023]
Abstract
New and innovative treatment strategies for cancer patients in the fields of immunotherapy and radiotherapy are rapidly developing in parallel. Among the most promising preclinical treatment approaches is combining immunotherapy with radiotherapy where early data suggest synergistic effects in several tumor model systems. These studies demonstrate that radiation combined with immunotherapy can result in superior efficacy for local tumor control. More alluring is the emergence of data suggesting an equally profound systemic response also known as "abscopal" effects with the combination of radiation and certain immunotherapies. Studies addressing optimal radiation dose, fractionation, and modality to be used in combination with immunotherapy still require further exploration. However, recent anecdotal clinical reports combining stereotactic or hypofractionated radiation regimens with immunotherapy have resulted in dramatic sustained clinical responses, both local and abscopal. Technologic advances in clinical radiation therapy has made it possible to deliver hypofractionated regimens anywhere in the body using stereotactic radiation techniques, facilitating further clinical investigations. Thus, stereotactic radiation in combination with immunotherapy agents represents an exciting and potentially fruitful new space for improving cancer therapeutic responses.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Radiation Oncology, University of Washington, 1959 NE Pacific Street, P.O. Box 356043, Seattle, WA 98195, USA
| | - Timothy J. Harris
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD 21218, USA
| | - Michael Lim
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD 21218, USA
- Department of Neurosurgery, Johns Hopkins Medicine, Baltimore, MD 21218, USA
| | - Charles G. Drake
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD 21218, USA
- Department of Urology, Johns Hopkins Medicine, Baltimore, MD 21218, USA
| | - Phuoc T. Tran
- Department of Radiation Oncology & Molecular Radiation Sciences, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD 21218, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD 21218, USA
- Department of Urology, Johns Hopkins Medicine, Baltimore, MD 21218, USA
| |
Collapse
|
691
|
Ahn BJ, Pollack IF, Okada H. Immune-checkpoint blockade and active immunotherapy for glioma. Cancers (Basel) 2013; 5:1379-412. [PMID: 24202450 PMCID: PMC3875944 DOI: 10.3390/cancers5041379] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/24/2013] [Accepted: 10/24/2013] [Indexed: 02/01/2023] Open
Abstract
Cancer immunotherapy has made tremendous progress, including promising results in patients with malignant gliomas. Nonetheless, the immunological microenvironment of the brain and tumors arising therein is still believed to be suboptimal for sufficient antitumor immune responses for a variety of reasons, including the operation of “immune-checkpoint” mechanisms. While these mechanisms prevent autoimmunity in physiological conditions, malignant tumors, including brain tumors, actively employ these mechanisms to evade from immunological attacks. Development of agents designed to unblock these checkpoint steps is currently one of the most active areas of cancer research. In this review, we summarize recent progresses in the field of brain tumor immunology with particular foci in the area of immune-checkpoint mechanisms and development of active immunotherapy strategies. In the last decade, a number of specific monoclonal antibodies designed to block immune-checkpoint mechanisms have been developed and show efficacy in other cancers, such as melanoma. On the other hand, active immunotherapy approaches, such as vaccines, have shown encouraging outcomes. We believe that development of effective immunotherapy approaches should ultimately integrate those checkpoint-blockade agents to enhance the efficacy of therapeutic approaches. With these agents available, it is going to be quite an exciting time in the field. The eventual success of immunotherapies for brain tumors will be dependent upon not only an in-depth understanding of immunology behind the brain and brain tumors, but also collaboration and teamwork for the development of novel trials that address multiple layers of immunological challenges in gliomas.
Collapse
Affiliation(s)
- Brian J. Ahn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; E-Mail:
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
| | - Ian F. Pollack
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hideho Okada
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; E-Mail:
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA; E-Mail:
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-412-623-3111; Fax: +1-412-623-1415
| |
Collapse
|
692
|
Kyi C, Postow MA. Checkpoint blocking antibodies in cancer immunotherapy. FEBS Lett 2013; 588:368-76. [PMID: 24161671 DOI: 10.1016/j.febslet.2013.10.015] [Citation(s) in RCA: 179] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 01/05/2023]
Abstract
Cancers can be recognized by the immune system, and the immune system may regulate and even eliminate tumors. The development of checkpoint blocking antibodies, such as those directed against cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death 1 receptor (PD-1), have demonstrated significant recent promise in the treatment of an expanding list of malignancies. While both CTLA-4 and PD-1 function as negative regulators, each plays a non-redundant role in modulating immune responses. CTLA-4 attenuates the early activation of naïve and memory T cells. In contrast, PD-1 is primarily involved in modulating T cell activity in peripheral tissues via interaction with its ligands, PD-L1 and PD-L2. Unfortunately, not all patients respond to these therapies, and evaluation of biomarkers associated with clinical outcomes is ongoing. This review will examine the efficacy, toxicities, and clinical development of checkpoint blocking antibodies, including agents already approved by the US Food and Drug Administration (anti-CTLA-4, ipilimumab) or in development (anti-PD-1, PD-L1). Future studies will likely uncover new promising immunologic checkpoints to target alone or in combination with other immunotherapeutic approaches, chemotherapy, radiotherapy, and small molecules.
Collapse
Affiliation(s)
- Chrisann Kyi
- Department of Medicine, New York Presbyterian Hospital Cornell, 525 E 68th St., New York, NY 10065, United States
| | - Michael A Postow
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 300 E 66th St., New York, NY 10065, United States.
| |
Collapse
|
693
|
Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br J Cancer 2013; 110:501-9. [PMID: 24129245 PMCID: PMC3899751 DOI: 10.1038/bjc.2013.640] [Citation(s) in RCA: 243] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/12/2013] [Accepted: 09/11/2013] [Indexed: 12/22/2022] Open
Abstract
Background: We aimed to investigate the prognostic value of tumour-infiltrating lymphocytes' (TILs) expression in pretreatment specimens from patients with head and neck squamous cell carcinoma (HNSCC) treated with definitive chemoradiotherapy (CRT). Methods: The prevalence of CD3+, CD8+, CD4+ and FOXP3+ TILs was assessed using immunohistochemistry in tumour tissue obtained from 101 patients before CRT and was correlated with clinicopathological characteristics as well as local failure-free- (LFFS), distant metastases free- (DMFS), progression-free (PFS) and overall survival (OS). Survival curves were measured using the Kaplan–Meier method, and differences in survival between the groups were estimated using the log-rank test. Prognostic effects of TIL subset density were determined using the Cox regression analysis. Results: With a mean follow-up of 25 months (range, 2.3–63 months), OS at 2 years was 57.4% for the entire cohort. Patients with high immunohistochemical CD3 and CD8 expression had significantly increased OS (P=0.024 and P=0.028), PFS (P=0.044 and P=0.047) and DMFS (P=0.021 and P=0.026) but not LFFS (P=0.90 and P=0.104) in multivariate analysis that included predictive clinicopathologic factors, such as age, sex, T-stage, N-stage, tumour grading and localisation. Neither CD4 nor FOXP3 expression showed significance for the clinical outcome. The lower N-stage was associated with improved OS in the multivariate analysis (P=0.049). Conclusion: The positive correlation between a high number of infiltrating CD3+ and CD8+ cells and clinical outcome indicates that TILs may have a beneficial role in HNSCC patients and may serve as a biomarker to identify patients likely to benefit from definitive CRT.
Collapse
|
694
|
Griesinger AM, Birks DK, Donson AM, Amani V, Hoffman LM, Waziri A, Wang M, Handler MH, Foreman NK. Characterization of distinct immunophenotypes across pediatric brain tumor types. THE JOURNAL OF IMMUNOLOGY 2013; 191:4880-8. [PMID: 24078694 DOI: 10.4049/jimmunol.1301966] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Despite increasing evidence that antitumor immune control exists in the pediatric brain, these findings have yet to be exploited successfully in the clinic. A barrier to development of immunotherapeutic strategies in pediatric brain tumors is that the immunophenotype of these tumors' microenvironment has not been defined. To address this, the current study used multicolor FACS of disaggregated tumor to systematically characterize the frequency and phenotype of infiltrating immune cells in the most common pediatric brain tumor types. The initial study cohort consisted of 7 pilocytic astrocytoma (PA), 19 ependymoma (EPN), 5 glioblastoma (GBM), 6 medulloblastoma (MED), and 5 nontumor brain (NT) control samples obtained from epilepsy surgery. Immune cell types analyzed included both myeloid and T cell lineages and respective markers of activated or suppressed functional phenotypes. Immune parameters that distinguished each of the tumor types were identified. PA and EPN demonstrated significantly higher infiltrating myeloid and lymphoid cells compared with GBM, MED, or NT. Additionally, PA and EPN conveyed a comparatively activated/classically activated myeloid cell-skewed functional phenotype denoted in particular by HLA-DR and CD64 expression. In contrast, GBM and MED contained progressively fewer infiltrating leukocytes and more muted functional phenotypes similar to that of NT. These findings were recapitulated using whole tumor expression of corresponding immune marker genes in a large gene expression microarray cohort of pediatric brain tumors. The results of this cross-tumor comparative analysis demonstrate that different pediatric brain tumor types exhibit distinct immunophenotypes, implying that specific immunotherapeutic approaches may be most effective for each tumor type.
Collapse
|
695
|
Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013. [PMID: 24048123 DOI: 10.1038/ni.2703.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most tumor cells express antigens that can mediate recognition by host CD8(+) T cells. Cancers that are detected clinically must have evaded antitumor immune responses to grow progressively. Recent work has suggested two broad categories of tumor escape based on cellular and molecular characteristics of the tumor microenvironment. One major subset shows a T cell-inflamed phenotype consisting of infiltrating T cells, a broad chemokine profile and a type I interferon signature indicative of innate immune activation. These tumors appear to resist immune attack through the dominant inhibitory effects of immune system-suppressive pathways. The other major phenotype lacks this T cell-inflamed phenotype and appears to resist immune attack through immune system exclusion or ignorance. These two major phenotypes of tumor microenvironment may require distinct immunotherapeutic interventions for maximal therapeutic effect.
Collapse
|
696
|
Tartour E, Zitvogel L. Lung cancer: potential targets for immunotherapy. THE LANCET RESPIRATORY MEDICINE 2013; 1:551-63. [PMID: 24461616 DOI: 10.1016/s2213-2600(13)70159-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Lung cancer is the most common cause of cancer-related mortality worldwide and a therapeutic challenge. Recent success with antibodies blocking immune checkpoints in non-small-cell lung cancers (NSCLC) highlights the potential of immunotherapy for lung cancer treatment, and the need for trials of combination regimens of immunotherapy plus chemotherapy that lead to immunogenic cell death. Here, we review the development of immunogenic cytotoxic compounds, vaccines, and antibodies in NSCLC, in view of their integration into personalised oncology.
Collapse
Affiliation(s)
- Eric Tartour
- Hôpital Européen Georges Pompidou, Service d'Immunologie Biologique, Paris, France; INSERM, U970 PARCC, Université Paris Descartes, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Institut Gustave Roussy, Villejuif, France; Université Paris Sud, Kremlin Bicêtre, France.
| |
Collapse
|
697
|
Kalbasi A, June CH, Haas N, Vapiwala N. Radiation and immunotherapy: a synergistic combination. J Clin Invest 2013; 123:2756-63. [PMID: 23863633 DOI: 10.1172/jci69219] [Citation(s) in RCA: 221] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy can be an effective treatment for metastatic cancer, but a significant subpopulation will not respond, likely due to the lack of antigenic mutations or the immune-evasive properties of cancer. Likewise, radiation therapy (RT) is an established cancer treatment, but local failures still occur. Clinical observations suggest that RT may expand the therapeutic reach of immunotherapy. We examine the immunobiologic and clinical rationale for combining RT and immunotherapy, two modalities yet to be used in combination in routine practice. Preclinical data indicate that RT can potentiate the systemic efficacy of immunotherapy, while activation of the innate and adaptive immune system can enhance the local efficacy of RT.
Collapse
Affiliation(s)
- Anusha Kalbasi
- Department of Radiation Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
698
|
Nicholas S, Mathios D, Ruzevick J, Jackson C, Yang I, Lim M. Current trends in glioblastoma multiforme treatment: radiation therapy and immune checkpoint inhibitors. Brain Tumor Res Treat 2013; 1:2-8. [PMID: 24904882 PMCID: PMC4027120 DOI: 10.14791/btrt.2013.1.1.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/20/2013] [Accepted: 04/05/2013] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer. Even with aggressive combination therapy, the median life expectancy for patients with GBM remains approximately 14 months. In order to improve the outcomes of patients with GBM, the development of newer treatments is critical. The concept of using the immune system as a therapeutic option has been suggested for several decades; by harnessing the body's adaptive immune mechanisms, immunotherapy could provide a durable and targeted treatment against cancer. However, many cancers, including GBM, have developed mechanisms that protect tumor cells from being recognized and eliminated by the immune system. For new immunotherapeutic regimens to be successful, overcoming immunosuppression via immune checkpoint signaling should be taken into consideration.
Collapse
Affiliation(s)
- Sarah Nicholas
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dimitris Mathios
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob Ruzevick
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isaac Yang
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael Lim
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. ; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
699
|
Evans T, Ciunci C, Hertan L, Gomez D. Special topics in immunotherapy and radiation therapy: reirradiation and palliation. Transl Lung Cancer Res 2007; 6:119-130. [PMID: 28529895 DOI: 10.21037/tlcr.2017.04.03] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has revolutionized the treatment of non-small cell lung cancer (NSCLC). However, thus far, its use has only been established in patients with advanced disease either as first-line therapy in selected patients or following chemotherapy. What is not yet known is how best to incorporate radiation with immunotherapy agents. Many patients with advanced disease can benefit from palliative radiation, but the combination of radiation with immunotherapy has the potential to increase the toxicity of both modalities. Intriguingly, the combination also has the potential to enhance the efficacy of both modalities. For this reason, combining immunotherapy and radiation may help salvage patients with recurrent localized disease who are candidates for re-irradiation. We review the current data evaluating immunotherapy with both palliative radiation as well as definitive re-irradiation in NSCLC.
Collapse
Affiliation(s)
- Tracey Evans
- University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Christine Ciunci
- University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Lauren Hertan
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Daniel Gomez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|