151
|
Uveal Melanoma: A European Network to Face the Many Challenges of a Rare Cancer. Cancers (Basel) 2019; 11:cancers11060817. [PMID: 31200439 PMCID: PMC6628377 DOI: 10.3390/cancers11060817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Uveal melanoma (UM) is the most frequent primary ocular cancer in adults, accounting for 5% of all melanomas. Despite effective treatments for the primary tumour, up to 50% of UM patients will develop metastasis, leading to a very poor prognosis and a median overall survival of 6 to 12 months, with no major improvements in the last 30 years. There is no standard oncological treatment available for metastatic UM patients, and BRAF/MEK and immune checkpoint inhibitors show disappointing results when compared to cutaneous melanoma (CM). Recent advances in biology, however, identified specific gene and chromosome alterations, potentially permitting an actively tailored surveillance strategy, and dedicated clinical studies. Being a rare cancer, UM patients have to overcome issues such as identifying referral centres, having access to information, and partnering with oncologists for specific management strategies and research priorities. Here, we describe how the European Rare Adult solid Cancer Network (EURACAN) will help in addressing these challenges and accelerating international collaborations to enhance the development of innovative treatments in UM.
Collapse
|
152
|
Results from phase II trial of HSP90 inhibitor, STA-9090 (ganetespib), in metastatic uveal melanoma. Melanoma Res 2019; 28:605-610. [PMID: 30211813 DOI: 10.1097/cmr.0000000000000509] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Uveal melanoma (UM) is a rare form of melanoma without effective therapy. The biology of UM relies on several heat-shock protein 90 (Hsp90)-dependent molecules such as MET, MEK and AKT, making Hsp90 inhibition a rational approach. Patients with stage IV UM, measurable disease, and no previous chemotherapy were eligible. Patients received either ganetespib 200 mg weekly (cohort A) or 150 mg twice a week (cohort B). Primary endpoint response rate (RR) was assessed by RECIST. A total of 17 patients were accrued for this study, with seven in cohort A and 10 in cohort B. Liver metastases were present in 59%. Response outcomes included one partial response, four stable disease, 11 progressive disease, and one withdrawal for ORR: 5.9% and disease control rate of 29.4%. Progression-free survival was 1.6 months (cohort A) and 1.8 months (cohort B). Overall survival was 8.5 months (cohort A) and 4.9 months (cohort B). An overall 31% of adverse events were grade 3-4 and were mostly related to gastrointestinal toxicities. Early on-treatment (1 months) positron emission tomography showed reduction in metabolic activity in 24% of patients, suggesting a pharmacodynamic effect of Hsp90 inhibition. These early metabolic changes did not seem to be durable and/or clinically significant in relation to the 2-month response assessment. Hsp90 inhibition with ganetespib resulted in modest clinical benefit on two dosing schedules and was associated with significant, although manageable, gastrointestinal toxicity. Evidence of pharmacodynamic activity for Hsp90 inhibition was observed via positron emission tomography, which did not translate into clinical benefit, suggesting rapid development of resistance.
Collapse
|
153
|
Violanti SS, Bononi I, Gallenga CE, Martini F, Tognon M, Perri P. New Insights into Molecular Oncogenesis and Therapy of Uveal Melanoma. Cancers (Basel) 2019; 11:E694. [PMID: 31109147 PMCID: PMC6562554 DOI: 10.3390/cancers11050694] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma (UM), which is the most common cancer of the eye, was investigated in recent years by many teams in the field of biomedical sciences and eye clinicians. New knowledge was acquired on molecular pathways found to be dysregulated during the multistep process of oncogenesis, whereas novel therapeutic approaches gave significant results in the clinical applications. Uveal melanoma-affected patients greatly benefited from recent advances of the research in this eye cancer. Tumour biology, genetics, epigenetics and immunology contributed significantly in elucidating the role of different genes and related pathways during uveal melanoma onset/progression and UM treatments. Indeed, these investigations allowed identification of new target genes and to develop new therapeutic strategies/compounds to cure this aggressive melanoma of the eye. Unfortunately, the advances reported in the treatment of cutaneous melanoma have not produced analogous benefits in metastatic uveal melanoma. Nowadays, no systemic adjuvant therapy has been shown to improve overall survival or reduce the risk of metastasis. However, the increasing knowledge of this disease, and the encouraging results seen in clinical trials, offer promise for future effective therapies. Herein, different pathways/genes involved in uveal melanoma onset/progression were taken into consideration, together with novel therapeutic approaches.
Collapse
Affiliation(s)
- Sara Silvia Violanti
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| | - Ilaria Bononi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Carla Enrica Gallenga
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| | - Fernanda Martini
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Mauro Tognon
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, School of Medicine, University of Ferrara, 44121 Ferrara, Italy.
| | - Paolo Perri
- Department of Biomedical Sciences and Specialized Surgeries, School of Medicine, University of Ferrara and Eye Unit of University Hospital of Ferrara, 44124 Ferrara, Italy.
| |
Collapse
|
154
|
Uveal melanoma: physiopathology and new in situ-specific therapies. Cancer Chemother Pharmacol 2019; 84:15-32. [DOI: 10.1007/s00280-019-03860-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/02/2019] [Indexed: 12/12/2022]
|
155
|
Jespersen H, Olofsson Bagge R, Ullenhag G, Carneiro A, Helgadottir H, Ljuslinder I, Levin M, All-Eriksson C, Andersson B, Stierner U, Nilsson LM, Nilsson JA, Ny L. Concomitant use of pembrolizumab and entinostat in adult patients with metastatic uveal melanoma (PEMDAC study): protocol for a multicenter phase II open label study. BMC Cancer 2019; 19:415. [PMID: 31046743 PMCID: PMC6498539 DOI: 10.1186/s12885-019-5623-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 04/18/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND While recent years have seen a revolution in the treatment of metastatic cutaneous melanoma, no treatment has yet been able to demonstrate any prolonged survival in metastatic uveal melanoma. Thus, metastatic uveal melanoma remains a disease with an urgent unmet medical need. Reports of treatment with immune checkpoint inhibitors have thus far been disappointing. Based on animal experiments, it is reasonable to hypothesize that the effect of immunotherapy may be augmented by epigenetic therapy. Proposed mechanisms include enhanced expression of HLA class I and cancer antigens on cancer cells, as well as suppression of myeloid suppressor cells. METHODS The PEMDAC study is a multicenter, open label phase II study assessing the efficacy of concomitant use of the PD1 inhibitor pembrolizumab and the class I HDAC inhibitor entinostat in adult patients with metastatic uveal melanoma. Primary endpoint is objective response rate. Eligible patients have histologically confirmed metastatic uveal melanoma, ECOG performance status 0-1, measurable disease as per RECIST 1.1 and may have received any number of prior therapies, with the exception of anticancer immunotherapy. Twenty nine patients will be enrolled. Patients receive pembrolizumab 200 mg intravenously every third week in combination with entinostat 5 mg orally once weekly. Treatment will continue until progression of disease or intolerable toxicity or for a maximum of 24 months. DISCUSSION The PEMDAC study is the first trial to assess whether the addition of an HDAC inhibitor to anti-PD1 therapy can yield objective anti-tumoral responses in metastatic UM. TRIAL REGISTRATION ClinicalTrials.gov registration number: NCT02697630 . (Registered 3 March 2016). EudraCT registration number: 2016-002114-50.
Collapse
Affiliation(s)
- Henrik Jespersen
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 2, 413 45, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 5, 413 45, Gothenburg, Sweden
| | - Gustav Ullenhag
- Department of Radiology, Oncology and Radiation Science, Section of Oncology, Uppsala University, 751 05, Uppsala, Sweden
| | - Ana Carneiro
- Department of Oncology, Skåne University Hospital, Getingevägen 4, 221 85, Lund, Sweden
| | - Hildur Helgadottir
- Department of Oncology, Karolinska University Hospital, Karolinska vägen, 171 76, Stockholm, Sweden
| | - Ingrid Ljuslinder
- Department of Oncology, Norrlands University Hospital, 901 85, Umeå, Sweden
| | - Max Levin
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 2, 413 45, Gothenburg, Sweden
| | | | - Bengt Andersson
- Department of Clinical immunology and transfusion medicine, Sahlgrenska University Hospital, Guldhedsgatan 10, 413 45, Gothenburg, Sweden
| | - Ulrika Stierner
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 2, 413 45, Gothenburg, Sweden
| | - Lisa M Nilsson
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Jonas A Nilsson
- Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Sahlgrenska University Hospital, Blå stråket 2, 413 45, Gothenburg, Sweden.
| |
Collapse
|
156
|
Beck RJ, Slagter M, Beltman JB. Contact-Dependent Killing by Cytotoxic T Lymphocytes Is Insufficient for EL4 Tumor Regression In Vivo. Cancer Res 2019; 79:3406-3416. [PMID: 31040155 DOI: 10.1158/0008-5472.can-18-3147] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/08/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
Immunotherapies are an emerging strategy for treatment of solid tumors. Improved understanding of the mechanisms employed by cytotoxic T lymphocytes (CTL) to control tumors will aid in the development of immunotherapies. CTLs can directly kill tumor cells in a contact-dependent manner or may exert indirect effects on tumor cells via secretion of cytokines. Here, we aim to quantify the importance of these mechanisms in murine thymoma EL4/EG7 cells. We developed an agent-based model (ABM) and an ordinary differential equation model of tumor regression after adoptive transfer of a population of CTLs. Models were parameterized based on in vivo measurements of CTL infiltration and killing rates applied to EL4/EG7 tumors and OTI T cells. We quantified whether infiltrating CTLs are capable of controlling tumors through only direct, contact-dependent killing. Both models agreed that the low measured killing rate of CTLs in vivo was insufficient to cause tumor regression. In our ABM, we also simulated CTL production of the cytokine IFNγ in order to explore how an antiproliferative effect of IFNγ might aid CTLs in tumor control. In this model, IFNγ substantially reduced tumor growth compared with direct killing alone. Collectively, these data demonstrate that contact-dependent killing is insufficient for EL4 regression in vivo and highlight the potential importance of cytokine-induced antiproliferative effects in T-cell-mediated tumor control. SIGNIFICANCE: Computational modeling highlights the importance of cytokine-induced antiproliferative effects in T-cell-mediated control of tumor progression.
Collapse
Affiliation(s)
- Richard J Beck
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Maarten Slagter
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
157
|
Kittler JM, Sommer J, Fischer A, Britting S, Karg MM, Bock B, Atreya I, Heindl LM, Mackensen A, Bosch JJ. Characterization of CD4+ T cells primed and boosted by MHCII primary uveal melanoma cell-based vaccines. Oncotarget 2019; 10:1812-1828. [PMID: 30956760 PMCID: PMC6442993 DOI: 10.18632/oncotarget.26737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/09/2019] [Indexed: 02/03/2023] Open
Abstract
Uveal melanoma is the most common primary malignancy of the eye in adults. Despite significant improvements in treatment of the primary tumor, to date none of these therapies prevent metastatic disease or improve overall survival. We are exploring immunotherapeutic options for metastatic uveal melanoma using MHC II uveal melanoma cell-based vaccines that target the activation of tumor-reactive CD4+ T cells. Previously, we showed that these uveal melanoma cell-based vaccines activate CD4+ T cells within total peripheral blood lymphocytes (PBMC). Since PBMC include professional antigen presenting cells, we now demonstrate that Mel202/DR1/CD80 vaccine cells directly activate a diverse repertoire of purified, naïve CD4+ T cells. The activated CD4+ T cells proliferated, secreted high amounts of interferon gamma (IFNγ) and produced a heterogeneous profile of Th1, Th2 and Th17 cytokines. Analysis of the TCR-Vβ-repertoire showed that a polyclonal T cell response was induced, suggesting the capacity of vaccine-activated CD4+ T cells to target multiple tumor (neo)antigens. In addition, a subset of the responding CD4+ T cells expressed forkhead box protein P3 (FoxP3), indicating that although a regulatory component of the vaccine-activated CD4+ T cell response was induced, the anti-tumor vaccine response was not limited by these regulatory CD4+ T cells. Finally, Mel202/DR1/CD80 uveal melanoma vaccine cells expressed the intercellular adhesion molecule 1 (ICAM-1) that was pivotal for CD4+ T cell activation via lymphocyte function-associated antigen 1(LFA-1). In conclusion, MHC II uveal melanoma vaccines activate purified CD4+ T cells and may serve as a novel immunotherapy for uveal melanoma patients.
Collapse
Affiliation(s)
- Julia M Kittler
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Jonas Sommer
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Anika Fischer
- Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Sabine Britting
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Margarete M Karg
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Barbara Bock
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Imke Atreya
- Department of Internal Medicine 1 - Gastroenterology, Pneumology and Endocrinology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Ludwig M Heindl
- Department of Ophthalmology and Center for Integrated Oncology (CIO) Cologne-Bonn, University of Cologne, Cologne, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Jacobus J Bosch
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
158
|
Yamamoto TN, Lee PH, Vodnala SK, Gurusamy D, Kishton RJ, Yu Z, Eidizadeh A, Eil R, Fioravanti J, Gattinoni L, Kochenderfer JN, Fry TJ, Aksoy BA, Hammerbacher JE, Cruz AC, Siegel RM, Restifo NP, Klebanoff CA. T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J Clin Invest 2019; 129:1551-1565. [PMID: 30694219 DOI: 10.1172/jci121491] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 01/15/2019] [Indexed: 12/29/2022] Open
Abstract
Across clinical trials, T cell expansion and persistence following adoptive cell transfer (ACT) have correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis to identify actionable ligand-receptor pairs capable of compromising T cell durability following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments (TMEs). Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T cells used for clinical ACT. We hypothesized that a cognate Fas-FasL interaction within the TME might limit both T cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired in the ability to bind FADD functioned as dominant negative receptors (DNRs), preventing FasL-induced apoptosis in Fas-competent T cells. T cells coengineered with a Fas DNR and either a T cell receptor or chimeric antigen receptor exhibited enhanced persistence following ACT, resulting in superior antitumor efficacy against established solid and hematologic cancers. Despite increased longevity, Fas DNR-engineered T cells did not undergo aberrant expansion or mediate autoimmunity. Thus, T cell-intrinsic disruption of Fas signaling through genetic engineering represents a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies.
Collapse
Affiliation(s)
- Tori N Yamamoto
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ping-Hsien Lee
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Suman K Vodnala
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Devikala Gurusamy
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Rigel J Kishton
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Zhiya Yu
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Arash Eidizadeh
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA
| | - Robert Eil
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
| | - Jessica Fioravanti
- Experimental Transplantation and Immunology Branch, NCI, NIH, Bethesda, Maryland, USA
| | - Luca Gattinoni
- Experimental Transplantation and Immunology Branch, NCI, NIH, Bethesda, Maryland, USA
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, NCI, NIH, Bethesda, Maryland, USA
| | - Terry J Fry
- Children's Hospital Colorado, University of Colorado Denver, Aurora, Colorado, USA
| | - Bulent Arman Aksoy
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeffrey E Hammerbacher
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anthony C Cruz
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Richard M Siegel
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Nicholas P Restifo
- Center for Cancer Research and.,Center for Cell-Based Therapy, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher A Klebanoff
- Parker Institute for Cancer Immunotherapy, New York, New York, USA.,Center for Cell Engineering and Department of Medicine, MSKCC, New York, New York, USA.,Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
159
|
Combination immunotherapies implementing adoptive T-cell transfer for advanced-stage melanoma. Melanoma Res 2019. [PMID: 29521881 DOI: 10.1097/cmr.0000000000000436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunotherapy is a promising method of treatment for a number of cancers. Many of the curative results have been seen specifically in advanced-stage melanoma. Despite this, single-agent therapies are only successful in a small percentage of patients, and relapse is very common. As chemotherapy is becoming a thing of the past for treatment of melanoma, the combination of cellular therapies with immunotherapies appears to be on the rise in in-vivo models and in clinical trials. These forms of therapies include tumor-infiltrating lymphocytes, T-cell receptor, or chimeric antigen receptor-modified T cells, cytokines [interleukin (IL-2), IL-15, IL-12, granulocyte-macrophage colony stimulating factor, tumor necrosis factor-α, interferon-α, interferon-γ], antibodies (αPD-1, αPD-L1, αTIM-3, αOX40, αCTLA-4, αLAG-3), dendritic cell-based vaccines, and chemokines (CXCR2). There are a substantial number of ongoing clinical trials using two or more of these combination therapies. Preliminary results indicate that these combination therapies are a promising area to focus on for cancer treatments, especially melanoma. The main challenges with the combination of cellular and immunotherapies are adverse events due to toxicities and autoimmunity. Identifying mechanisms for reducing or eliminating these adverse events remains a critical area of research. Many important questions still need to be elucidated in regard to combination cellular therapies and immunotherapies, but with the number of ongoing clinical trials, the future of curative melanoma therapies is promising.
Collapse
|
160
|
Wolf B, Zimmermann S, Arber C, Irving M, Trueb L, Coukos G. Safety and Tolerability of Adoptive Cell Therapy in Cancer. Drug Saf 2019; 42:315-334. [DOI: 10.1007/s40264-018-0779-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
161
|
Liang C, Peng L, Zeng S, Zhao Q, Tang L, Jiang X, Zhang J, Yan N, Chen Y. Investigation of indoleamine 2,3-dioxygenase 1 expression in uveal melanoma. Exp Eye Res 2019; 181:112-119. [PMID: 30639792 DOI: 10.1016/j.exer.2019.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 01/03/2019] [Accepted: 01/03/2019] [Indexed: 02/05/2023]
Abstract
The purpose of this study was to investigate indoleamine 2,3-dioxygenase 1 (IDO1) expression and its implications in uveal melanoma (UM). Bioinformatics analysis was performed on microarray data (GSE22138 and GSE27831) from the Gene Expression Omnibus (GEO) database to evaluate IDO1 expression in mRNA level. Ninety-two cases in the database were divided into the IDO1-high group (46 cases) and IDO1-low group (46 cases). Paraffin embedded tumor sections from 27 patients with UM were studied by immunofluorescence. The mRNA results showed that IDO1 expression was inversely correlated with tumor thickness (9.93 ± 3.33 mm in IDO1-high group vs. 11.56 ± 2.38 mm in IDO1-low group) (p = 0.016) and metastatic rate (30.4% in IDO1-high group vs. 69.6% in IDO1-low group, p < 0.001). The IDO1-high group showed higher immune cell gene expression: CD3D (6.56 ± 1.0 vs. 5.46 ± 0.53, p < 0.0001), CD4 (4.72 ± 0.4 vs. 4.2 ± 0.42, p < 0.0001), and CD68 (6.17 ± 1.23 vs. 5.53 ± 0.77, p = 0.015). Further analysis showed that immune-suppressive T regulatory cell genes (CD3D, CD4, IL2RA and FOXP3) were expressed in 67.4% (31/46) cases in the IDO1-high group and 23.91% (11/46) cases in the IDO1-low group. In addition, IDO1 and interferon gamma (IFNG) mRNA expression were strongly correlated (r = 0.70, p < 0.0001). The correlation analysis of different immune checkpoints showed that IDO1 was positively correlated with CD274(PDL1), but not CTLA4 or PDCD1.The disease-free survival (DFS) in the IDO1-high/IFNG-high group was better than that of the IDO1-low/IFNG-low group. The IDO1 immunostaining result showed that 2 cases in 18 UMs with Bruch's membrane (BM) rupture and 7 out of 9 cases without BM rupture were scored high (Grade 2-3) (p = 0.001). Comparing the immune cells staining results between IDO1-high group and IDO1-low group, higher percentage of patients in the former group had high levels of T cells and macrophages infiltration, but only the difference in macrophage was statistically significant (CD68, 77.78% vs. 27.78%, p = 0.04). The analysis based on GEO data and the result from immunostaining study are consistent with each other. In conclusion, the expression of IDO1 is probably induced by IFNγ from infiltrated immune cells in UM. BM rupture is an important indicator of IDO1 expression level and distribution pattern. The complex role of IDO1 may limit its therapeutic effect in UM.
Collapse
Affiliation(s)
- Chen Liang
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China; Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| | - Lanya Peng
- West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| | - Shaoxue Zeng
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China; Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| | - Qing Zhao
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China; Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| | - Linqiao Tang
- Research Core Facility of West China Hospital, Sichuan University, China.
| | - Xiaoshuang Jiang
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China.
| | - JunJun Zhang
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China.
| | - Naihong Yan
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China; Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| | - YingYing Chen
- Research Laboratory of Ophthalmology and Vision Sciences, Torsten-Wiesel Research Institute of World Eye Organization, State Key Laboratory of Biotherapy, West China Hospital, SiChuan University, Chengdu, China; Department of Ophthalmology, West China Hospital, Sichuan University, Cheng Du, Sichuan, China.
| |
Collapse
|
162
|
Man F, Lim L, Volpe A, Gabizon A, Shmeeda H, Draper B, Parente-Pereira AC, Maher J, Blower PJ, Fruhwirth GO, T M de Rosales R. In Vivo PET Tracking of 89Zr-Labeled Vγ9Vδ2 T Cells to Mouse Xenograft Breast Tumors Activated with Liposomal Alendronate. Mol Ther 2019; 27:219-229. [PMID: 30429045 PMCID: PMC6318719 DOI: 10.1016/j.ymthe.2018.10.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
Gammadelta T (γδ-T) cells are strong candidates for adoptive immunotherapy in oncology due to their cytotoxicity, ease of expansion, and favorable safety profile. The development of γδ-T cell therapies would benefit from non-invasive cell-tracking methods and increased targeting to tumor sites. Here we report the use of [89Zr]Zr(oxinate)4 to track Vγ9Vδ2 T cells in vivo by positron emission tomography (PET). In vitro, we showed that 89Zr-labeled Vγ9Vδ2 T cells retained their viability, proliferative capacity, and anti-cancer cytotoxicity with minimal DNA damage for amounts of 89Zr ≤20 mBq/cell. Using a mouse xenograft model of human breast cancer, 89Zr-labeled γδ-T cells were tracked by PET imaging over 1 week. To increase tumor antigen expression, the mice were pre-treated with PEGylated liposomal alendronate. Liposomal alendronate, but not placebo liposomes or non-liposomal alendronate, significantly increased the 89Zr signal in the tumors, suggesting increased homing of γδ-T cells to the tumors. γδ-T cell trafficking to tumors occurred within 48 hr of administration. The presence of γδ-T cells in tumors, liver, and spleen was confirmed by histology. Our results demonstrate the suitability of [89Zr]Zr(oxinate)4 as a cell-labeling agent for therapeutic T cells and the potential benefits of liposomal bisphosphonate treatment before γδ-T cell administration.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Lindsay Lim
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Alessia Volpe
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Alberto Gabizon
- Oncology Institute, Shaare Zedek Medical Center and Hebrew University-School of Medicine, Jerusalem 9103102, Israel
| | - Hilary Shmeeda
- Oncology Institute, Shaare Zedek Medical Center and Hebrew University-School of Medicine, Jerusalem 9103102, Israel
| | - Benjamin Draper
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Ana C Parente-Pereira
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - John Maher
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London SE1 9RT, UK
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Gilbert O Fruhwirth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
163
|
Wang Y, Li P, Wang B, Wang S, Liu P. Identification of myeloid-derived suppressor cells that have an immunosuppressive function in NF2 patients. J Cancer Res Clin Oncol 2019; 145:523-533. [PMID: 30603902 DOI: 10.1007/s00432-018-02825-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE There is no targeted drug therapy for NF2 patients, and surgery or radiosurgery is not always effective. Therefore, the exploration of new therapeutic pathways is urgently needed. METHODS We analyzed the expression of cytokines in the serum of NF2 patients and determined the percentage of HLA-DR-CD33+CD11b+ cells in blood and NF2-associated schwannomas. Furthermore, we analyzed the role of HLA-DR-CD33+CD11b+ cells in inhibiting T-cell proliferation, cytokine production, and transforming growth factor expression. RESULTS NF2 patients are in an immunosuppressed state with elevated IL-10 and TGF-β expression in plasma and the lymphocytes from NF2 patients secrete less IFN-γ and CD3+ T cells proliferate slower than normal healthy donors. HLA-DR-CD33+CD11b+ cells frequency significantly increased in the PBMCs and infiltrated in the tumor, these cells express higher iNOS, NOX2 and TGF-β, and induce TGF-β secretion to inhibit CD8+ T-cell proliferation, and induce T-cell transformation to a CD4+CD25+Foxp3+ regulatory T cells phenotype. NF2-associated schwannoma cells induced monocytes transformation into an HLA-DR-CD33+CD11b+ phenotype, and surgical removal of the tumor reduced the percentage of these cells. CONCLUSIONS HLA-DR-CD33+CD11b+ cells may represent a population of MDSCs in NF2 patients. Dissecting the mechanisms behind these suppressive mechanisms will be helpful for the design of effective immunotherapeutic protocols and likely provide a new effective treatment for NF2 patients.
Collapse
Affiliation(s)
- Ying Wang
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China
| | - Peng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Wang
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China
| | - Pinan Liu
- Beijing Neurosurgical Institute, Capital Medical University, No. 6, Tiantan Xili, Chongwen District, Beijing, 100050, China. .,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
164
|
Harbour JW, Paez-Escamilla M, Cai L, Walter SD, Augsburger JJ, Correa ZM. Are Risk Factors for Growth of Choroidal Nevi Associated With Malignant Transformation? Assessment With a Validated Genomic Biomarker. Am J Ophthalmol 2019; 197:168-179. [PMID: 30195895 PMCID: PMC6291343 DOI: 10.1016/j.ajo.2018.08.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/21/2022]
Abstract
PURPOSE To test the hypothesis that widely used clinical risk factors for growth of choroidal nevi are associated with malignant transformation. METHODS Fine needle biopsy for assignment of gene expression profile (class 1 or class 2) was performed in 207 choroidal melanocytic tumors < 3.5 mm in thickness. The class 2 profile was employed as a validated biomarker for malignant transformation. The following data were collected: patient age and sex, tumor diameter and thickness, distance of posterior tumor margin from the optic disc, and the presence or absence of serous retinal detachment, orange lipofuscin pigment, drusen, retinal pigment epithelial fibrosis, retinal pigment epithelial atrophy, visual symptoms, and documented tumor growth. RESULTS Clinical features associated with the class 2 profile included patient age > 60 years and tumor thickness > 2.25 mm (Fisher exact test, P = .002 for both). Documented growth was not associated with the class 2 profile (P = .5). The odds ratio of a tumor having the class 2 profile was 2.8 (95% confidence interval 1.3-5.9) for patient age > 60 years and 3.5 (95% confidence interval 1.4-8.8) for tumor thickness > 2.25 mm. For patients with both risk factors, the "number needed to treat" to identify 1 patient with a class 2 tumor was 4.3 (P = .0002). No other clinical feature or combination of features was associated with the class 2 profile. CONCLUSIONS None of the widely used choroidal nevus risk factors for tumor growth, nor documented growth itself, is pathognomonic of malignant transformation as defined by class 2 gene expression profile. Patient age and tumor thickness may be helpful for identifying small choroidal melanocytic tumors that are more likely to have the class 2 profile. Observation for growth prior to treatment continues to be reasonable for most patients with suspicious choroidal nevi. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- J William Harbour
- Ocular Oncology Service, Bascom Palmer Eye Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Manuel Paez-Escamilla
- Ocular Oncology Service, Bascom Palmer Eye Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Louis Cai
- Ocular Oncology Service, Bascom Palmer Eye Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Scott D Walter
- Ocular Oncology Service, Bascom Palmer Eye Institute and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - James J Augsburger
- Department of Ophthalmology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Zelia M Correa
- Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
165
|
Smith JR, Pe'er J, Belfort RN, Cardoso F, Carvajal RD, Carvalho C, Coupland SE, Desjardins L, Francis JH, Gallie BL, Gombos DS, Grossniklaus HE, Heegaard S, Jager MJ, Kaliki S, Ksander BR, Maeurer M, Moreno E, Pulido JS, Ryll B, Singh AD, Zhao J, Parreira A, Wilson DJ, O'Brien JM. Proceedings of the Association for Research in Vision and Ophthalmology and Champalimaud Foundation Ocular Oncogenesis and Oncology Conference. Transl Vis Sci Technol 2019; 8:9. [PMID: 30652059 PMCID: PMC6333107 DOI: 10.1167/tvst.8.1.9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
The 2018 Ocular Oncogenesis and Oncology Conference was held through a partnership of the Association for Research in Vision and Ophthalmology (ARVO) and the Champalimaud Foundation. Twenty-one experts from international ocular oncology centers, from the Champalimaud Clinical Centre and the Champalimaud Foundation Cancer Research Program, and from patient advocacy organizations, delivered lectures on subjects that ranged from global ocular oncology, to basic research in mechanisms of ocular malignancy, to clinical research in ocular cancers, and to anticipated future developments in the area. The scientific program of the conference covered a broad range of ocular tumors-including uveal melanoma, retinoblastoma, ocular surface tumors, and adnexal and intraocular lymphomas-and pathogenesis and management were deliberated in the context of the broader systemic cancer discipline. In considering the latest basic and clinical research developments in ocular oncogenesis and oncology, and providing the opportunity for cross-talk between ocular cancer biologists, systemic cancer biologists, ocular oncologists, systemic oncologists, patients, and patient advocates, the forum generated new knowledge and novel insights for the field. This report summarizes the content of the invited talks at the 2018 ARVO-Champalimaud Foundation Ocular Oncogenesis and Oncology Conference.
Collapse
Affiliation(s)
- Justine R. Smith
- Eye & Vision Health, Flinders University College of Medicine & Public Health, Adelaide, Australia
| | - Jacob Pe'er
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rubens N. Belfort
- Ophthalmology Department, Federal University of São Paulo, São Paulo, Brazil
| | - Fatima Cardoso
- Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Richard D. Carvajal
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Carlos Carvalho
- Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Sarah E. Coupland
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool and Royal Liverpool University Hospital, Liverpool, UK
| | | | - Jasmine H. Francis
- Ophthalmic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brenda L. Gallie
- Department of Ophthalmology and Vision Science, SickKids Hospital, Toronto, Canada
| | - Dan S. Gombos
- Section of Ophthalmology, M.D. Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Hans E. Grossniklaus
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Steffen Heegaard
- Departments of Ophthalmology and Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Swathi Kaliki
- Operation Eyesight Universal Institute for Eye Cancer, L.V. Prasad Eye Institute, Hyderabad, India
| | - Bruce R. Ksander
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Markus Maeurer
- Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Eduardo Moreno
- Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Jose S. Pulido
- Departments of Ophthalmology and Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Bettina Ryll
- Melanoma Patient Network Europe, Knivsta, Sweden
| | - Arun D. Singh
- Department of Ophthalmic Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Junyang Zhao
- Department of Ophthalmology, Beijing Children's Hospital, Beijing, China
| | - António Parreira
- Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - David J. Wilson
- Casey Eye Institute and Department of Ophthalmology, Oregon Health & Science University, Portland, OR, USA
| | - Joan M. O'Brien
- Scheie Eye Institute and Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
166
|
Yi Q, Zou WJ. A novel four‑snoRNA signature for predicting the survival of patients with uveal melanoma. Mol Med Rep 2018; 19:1294-1301. [PMID: 30569172 DOI: 10.3892/mmr.2018.9766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/14/2018] [Indexed: 11/06/2022] Open
Abstract
Uveal melanoma (UM), the predominant histological subtype of intraocular malignant tumors in adults, often results in high rates of mortality; effective prognostic signatures used to predict the survival of patients with UM are limited. Small nucleolar RNAs (snoRNAs) are emerging as important regulators in the processes of carcinogenesis and tumor progression, but knowledge of their application as prognostic markers in UM is limited. In the present study, the expression profiles of snoRNAs in UM were determined; a total of 60 snoRNAs were notably associated with the overall survival of patients with UM via univariate Cox survival analysis. Subsequently, a prognostic signature based on four snoRNAs was proposed, which retained their prognostic significance determined by a multivariate Cox survival analysis. The formula is as follows: ACA17 * (‑1.602) + ACA45 * 0.803 + HBII‑276 * 0.603 + SNORD12 * 1.348. Furthermore, the results of in silico analysis indicated that perturbation of the phototransduction, GABAergic synapse and amphetamine addiction pathways may be the potential molecular mechanisms underlying the poor prognosis of patients with UM. Collectively, the present study proposed a potential prognostic signature for patients with UM and the prospective mechanisms at the genome‑wide level were determined.
Collapse
Affiliation(s)
- Qiong Yi
- Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wen-Jin Zou
- Department of Ophthalmology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
167
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
168
|
IL-12 Gene Electrotransfer Triggers a Change in Immune Response within Mouse Tumors. Cancers (Basel) 2018; 10:cancers10120498. [PMID: 30544810 PMCID: PMC6315808 DOI: 10.3390/cancers10120498] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 02/03/2023] Open
Abstract
Metastatic melanoma is an aggressive skin cancer with a relatively low survival rate. Immune-based therapies have shown promise in the treatment of melanoma, but overall complete response rates are still low. Previous studies have demonstrated the potential of plasmid IL-12 (pIL-12) delivered by gene electrotransfer (GET) to be an effective immunotherapy for melanoma. However, events occurring in the tumor microenvironment following delivery have not been delineated. Therefore, utilizing a B16F10 mouse melanoma model, we evaluated changes in the tumor microenvironment following delivery of pIL-12 using different GET parameters or injection of plasmid alone. The results revealed a unique immune cell composition after intratumoral injection of pIL-12 GET. The number of immune memory cells was markedly increased in pIL-12 GET melanoma groups compared to control group. This was validated using flow cytometry to analyze peripheral blood mononuclear cells as well as delineating immune cell content using immunohistochemistry. Significant differences in multiple cell types were observed, including CD8⁺ T cells, regulatory T cells and myeloid cells, which were induced to mount a CD8⁺PD1- T cells immune response. Taken together, these findings suggest a basic understanding of the sequence of immune activity following pIL-12 GET and also illuminates that adjuvant immunotherapy can have a positive influence on the host immune response to cancer.
Collapse
|
169
|
Adoptive cellular therapies: the current landscape. Virchows Arch 2018; 474:449-461. [PMID: 30470934 PMCID: PMC6447513 DOI: 10.1007/s00428-018-2484-0] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/11/2022]
Abstract
For many cancer types, the immune system plays an essential role in their development and growth. Based on these rather novel insights, immunotherapeutic strategies have been developed. In the past decade, immune checkpoint blockade has demonstrated a major breakthrough in cancer treatment and has currently been approved for the treatment of multiple tumor types. Adoptive cell therapy (ACT) with tumor-infiltrating lymphocytes (TIL) or gene-modified T cells expressing novel T cell receptors (TCR) or chimeric antigen receptors (CAR) is another strategy to modify the immune system to recognize tumor cells and thus carry out an anti-tumor effector function. These treatments have shown promising results in various tumor types, and multiple clinical trials are being conducted worldwide to further optimize this treatment modality. Most successful results were obtained in hematological malignancies with the use of CD19-directed CAR T cell therapy and already led to the commercial approval by the FDA. This review provides an overview of the developments in ACT, the associated toxicity, and the future potential of ACT in cancer treatment.
Collapse
|
170
|
Meng M, Li L, Li R, Wang W, Chen Y, Xie Y, Han R, Zhu K, Huang W, Yang L, Li S, Shi J, Tan W, Gao H, Zhao Y, Yang L, Tan J, Hou Z. A dynamic transcriptomic atlas of cytokine-induced killer cells. J Biol Chem 2018; 293:19600-19612. [PMID: 30333226 PMCID: PMC6314136 DOI: 10.1074/jbc.ra118.003280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Several clinical immunotherapy trials with cytokine-induced killer (CIK) cells have been reported. However, molecular evidence of cell expansion, acquisition of tumor cytotoxicity, and safety of CIK cells is required before putting them to clinical use. Here, we performed dynamic transcriptomic analyses of CIKs generated from primary peripheral blood mononuclear cells exposed to interferon-γ, OKT3, and interleukin-2. CIK mRNAs were extracted and sequenced at days 0, 1, 7, and 14 and subjected to bioinformatics analyses. Using weighted correlation network analysis (WGCNA), we identified two major gene modules that mediate immune cell activation and mitosis. We found that activation and cytotoxicity of CIK cells likely rely on cluster of differentiation 8 (CD8) and its protein partner LCK proto-oncogene, Src family tyrosine kinase (LCK). A time-course series analysis revealed that CIK cells have relatively low immunogenicity because of decreased expression of some self-antigens. Importantly, we identified several crucial activating receptors and auxiliary adhesion receptors expressed on CIK cells that may function as tumor sensors. Interestingly, cytotoxicity-associated genes, including those encoding PRF1, GZMB, FASL, and several cytokines, were up-regulated in mature CIK cells. Most immune-checkpoint molecules and inflammatory tumor-promoting factors were down-regulated in the CIK cells, suggesting efficacy and safety in future clinical trials. Notably, insulin-like growth factor 1 (IGF-1) was highly expressed in CIK cells and may promote cytotoxicity, although it also could facilitate tumorigenesis. The transcriptomic atlas of CIK cells presented here may inform efforts to improve CIK-associated tumor cytotoxicity and safety in clinical trials.
Collapse
Affiliation(s)
- Mingyao Meng
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lin Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Ruhong Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenju Wang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yang Chen
- the Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanhua Xie
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Rui Han
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Kai Zhu
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenwen Huang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lili Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Shuo Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jianlin Shi
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Weiwei Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Hui Gao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yiyi Zhao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Li Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jing Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Zongliu Hou
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| |
Collapse
|
171
|
Rohaan MW, van den Berg JH, Kvistborg P, Haanen JBAG. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: a viable treatment option. J Immunother Cancer 2018; 6:102. [PMID: 30285902 PMCID: PMC6171186 DOI: 10.1186/s40425-018-0391-1] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023] Open
Abstract
The treatment of metastatic melanoma patients with autologous tumor-infiltrating lymphocytes (TIL) shows robust, reproducible, clinical responses in clinical trials executed in several specialized centers over the world. Even in the era of targeted therapy and immune checkpoint inhibition, TIL therapy can be an additional and clinically relevant treatment line. This review provides an overview of the clinical experiences with TIL therapy thus far, including lymphodepleting regimens, the use of interleukin-2 (IL-2) and the associated toxicity. Characteristics of the TIL products and the antigen recognition pattern will be discussed, as well as the current and upcoming production strategies, including the selective expansion of specific fractions from the cell product. In addition, the future potential of TIL therapy in melanoma and other tumor types will be covered.
Collapse
Affiliation(s)
- Maartje W Rohaan
- Department of Medical Oncology, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Joost H van den Berg
- Biotherapeutics Unit, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute (NKI), Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands. .,Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
172
|
Cancer Immunotherapy: A Simple Guide for Interventional Radiologists of New Therapeutic Approaches. Cardiovasc Intervent Radiol 2018; 42:1221-1229. [PMID: 30209564 DOI: 10.1007/s00270-018-2074-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/04/2018] [Indexed: 12/27/2022]
Abstract
The therapeutic options in the treatment of cancer therapy have been recently significantly increased with systemic immune-targeted therapies. Novel immunotherapy approaches based on immune checkpoint blockade or engineered cytotoxic T lymphocytes have reached late-stage clinical development, with highly encouraging results. The success of cancer immunotherapy has generated a tremendous interest in further developing and exploring these strategies in combination with other approaches such as radiotherapy and local ablative therapies in oncology. The goal of this review is to discuss current approaches in immunotherapy and provide simple and constructive explanations on their mechanisms of action as well as certain more common and serious toxicities.
Collapse
|
173
|
Mathis T, Cassoux N, Tardy M, Piperno S, Gastaud L, Dendale R, Maschi C, Nguyen AM, Meyer L, Bonnin N, Baillif S, Tick S, Mouriaux F, Jaspart F, Dellis J, Rosier L, Desjardins L, Herault J, Caujolle JP, Thariat J. [Management of uveal melanomas, guidelines for oncologists]. Bull Cancer 2018; 105:967-980. [PMID: 30217336 DOI: 10.1016/j.bulcan.2018.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/08/2018] [Accepted: 07/12/2018] [Indexed: 10/28/2022]
Abstract
Uveal melanomas are the most frequent primary malignant eye tumor. Enucleation was historically the gold standard. Since then, several studies showed that conservative treatments did not increase the risk of metastasis or survival. Choroidal melanomas are both radioresistant and located close to visual structures (the optic nerve and macula) of the eye, which may be preserved in some settings without compromising tumor control, as this is the first priority. Different types of radiation therapy may be used for such tumors: brachytherapy and charged particles, including proton beam therapy. If visual prognosis is dependent to the local treatment, the vital prognosis is dependent on the metastatic risk, with a risk of liver involvement in 20 to 50% of patients, depending on tumor size and genomics. Median survival after the discovery of liver metastases is about 15 months. The management of these patients is often complex. Systemic therapies (chemotherapy, targeted therapies, immunotherapy, etc.) yield limited response rates and although local treatments of liver metastases are promising, they are only feasible in selected patients. The mission of the MELACHONAT national network is to improve the management of patients regardless of the stage of the disease. The patient association ANPACO is dedicated to help uveal melanoma patients in their health care path and to promote knowledge dissemination within the patient community. The aim of this review is to focus on the local treatments of uveal melanomas as well as the management of their metastatic evolution.
Collapse
Affiliation(s)
- Thibaud Mathis
- Hôpital de la Croix-Rousse, 103 grande rue de la Croix-Rousse, 69004 Lyon, France
| | | | - Magali Tardy
- Hôpital Pasteur, 30, voie romaine, 06000 Nice, France
| | - Sophie Piperno
- Institut Curie, 26, rue de l'ulm, 75248 Paris cedex 05, France
| | | | - Rémi Dendale
- Institut Curie, 26, rue de l'ulm, 75248 Paris cedex 05, France
| | - Celia Maschi
- Hôpital Pasteur, 30, voie romaine, 06000 Nice, France
| | - Anh-Minh Nguyen
- Hôpital de la Croix-Rousse, 103 grande rue de la Croix-Rousse, 69004 Lyon, France
| | - Laurent Meyer
- Hôpitaux civils de Colmar, 39, avenue de la liberté, 68024 Colmar, France
| | - Nicolas Bonnin
- Centre d'ophtalmologie du Zénith, 63800 Cournon D'auvergne, France
| | | | - Sarah Tick
- CHNO des XV-XX, 28, rue de Charenton, 75012 Paris, France
| | - Fréderic Mouriaux
- Centre hospitalier universitaire Pontchaillou, 2, rue Henri-Le-Guilloux, 35033 Rennes, France
| | - Franck Jaspart
- Polyclinique du Parc, route d'Assevent, 59600 Maubeuge, France
| | - Josette Dellis
- Association nationale des patients atteints du cancer de l'œil, ANPACO, 5, rue de Fontfrède, 15230 Pierrefort, France
| | - Laurence Rosier
- Centre Retine Gallien, 68, rue du palais-Gallien, 33000 Bordeaux, France
| | | | - Joel Herault
- Centre Lacassagne, 227, avenue Valombrose, 06200 Nice, France
| | | | - Juliette Thariat
- Centre François Baclesse, ARCHADE, service de radiothérapie, 3, avenue du général-Harris, 14000 Caen, France.
| |
Collapse
|
174
|
Pfeiffer H, Völkl S, Gary R, Mackensen A, Achenbach S, Strasser E, Aigner M. Impact of collection programs for the generation of monocyte apheresis products on product quality and composition as starting material for the generation of cellular therapeutics. Transfusion 2018; 58:2175-2183. [DOI: 10.1111/trf.14817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Hella Pfeiffer
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Simon Völkl
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Regina Gary
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Andreas Mackensen
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Susanne Achenbach
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Erwin Strasser
- Department of Transfusion Medicine and Haemostaseology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| | - Michael Aigner
- Department of Medicine 5, Haematology and Oncology; Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsklinikum; Erlangen Germany
| |
Collapse
|
175
|
Kurupati RK, Zhou X, Xiang Z, Keller LH, Ertl HCJ. Safety and immunogenicity of a potential checkpoint blockade vaccine for canine melanoma. Cancer Immunol Immunother 2018; 67:1533-1544. [PMID: 30051333 PMCID: PMC7080056 DOI: 10.1007/s00262-018-2201-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 07/06/2018] [Indexed: 12/13/2022]
Abstract
Human immunotherapy with checkpoint blockades has achieved significant breakthroughs in recent years. In this study, a checkpoint blockade vaccine for canine melanoma was tested for safety and immunogenicity. Five healthy adult dogs received a mixture of three replication-defective chimpanzee-derived adenoviral vectors, one expressing mouse fibroblast-associated protein (mFAP) and the others expressing canine melanoma-associated antigens Trp-1 or Trp-2 fused into Herpes Simplex-1 glycoprotein D, a checkpoint inhibitor of herpes virus entry mediator (HVEM) pathways. The vaccine mixture was shown to be well tolerated and increased frequencies of canineTrp-1-specific activated CD8+ and CD4+ T cells secreting interferon-(IFN)-γ, tumor necrosis factor (TNF)-α, or interleukin (IL)-2 alone or in combinations in four and five out of five dogs, respectively. To avoid excessive bleeds, responses to cTrp-2 were not analyzed. All dogs responded with increased frequencies of mFAP-specific activated CD8+ and CD4+ T cells. The results of this safety/immunogenicity trial invite further testing of this checkpoint blockade vaccine combination in dogs with melanoma.
Collapse
Affiliation(s)
- Raj K Kurupati
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Xiangyang Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Zhiquan Xiang
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | | | | |
Collapse
|
176
|
Zhu G, Nemoto S, Mailloux AW, Perez-Villarroel P, Nakagawa R, Falahat R, Berglund AE, Mulé JJ. Induction of Tertiary Lymphoid Structures With Antitumor Function by a Lymph Node-Derived Stromal Cell Line. Front Immunol 2018; 9:1609. [PMID: 30061886 PMCID: PMC6054958 DOI: 10.3389/fimmu.2018.01609] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/28/2018] [Indexed: 02/03/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) associate with better prognosis in certain cancer types, but their underlying formation and immunological benefit remain to be determined. We established a mouse model of TLSs to study their contribution to antitumor immunity. Because the stroma in lymph nodes (sLN) participates in architectural support, lymphogenesis, and lymphocyte recruitment, we hypothesized that TLSs can be created by sLN. We selected a sLN line with fibroblast morphology that expressed sLN surface markers and lymphoid chemokines. The subcutaneous injection of the sLN line successfully induced TLSs that attracted infiltration of host immune cell subsets. Injection of MC38 tumor lysate-pulsed dendritic cells activated TLS-residing lymphocytes to demonstrate specific cytotoxicity. The presence of TLSs suppressed MC38 tumor growth in vivo by improving antitumor activity of tumor-infiltrating lymphocytes with downregulated immune checkpoint proteins (PD-1 and Tim-3). Future engineering of sLN lines may allow for further enhancements of TLS functions and immune cell compositions.
Collapse
Affiliation(s)
- Genyuan Zhu
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Satoshi Nemoto
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Adam W Mailloux
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Ryosuke Nakagawa
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Rana Falahat
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Anders E Berglund
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - James J Mulé
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States.,Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
177
|
Javed A, Arguello D, Johnston C, Gatalica Z, Terai M, Weight RM, Orloff M, Mastrangelo MJ, Sato T. PD-L1 expression in tumor metastasis is different between uveal melanoma and cutaneous melanoma. Immunotherapy 2018; 9:1323-1330. [PMID: 29185395 DOI: 10.2217/imt-2017-0066] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIM To compare PD-L1 expression between metastatic uveal melanoma (MUM) and metastatic cutaneous melanoma (MCM). MATERIALS & METHODS A total of 295 MCM and 78 MUM specimens were analyzed for tumor cell PD-L1 expression. Additionally, 91 MCM and 45 MUM specimens were analyzed for PD-1 expression on tumor-infiltrating lymphocytes. RESULTS A total of 77/295 (26.1%) MCM specimens expressed PD-L1 as compared to 4/78 (5.1%) MUM specimens (p < 0.0001). PD-1 expression on tumor-infiltrating lymphocytes was greater in MCM (73.6%; 67/91) than in MUM (51.1%; 23/45), respectively (p = 0.009). CONCLUSION Significant differences exist in PD-L1 expression between MCM and MUM. The lower PD-L1 expression in MUM may provide a rationale for failure of PD-1 inhibitor therapy and suggests that immune evasion in this disease may occur via alternative mechanisms.
Collapse
Affiliation(s)
- Asad Javed
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | - Mizue Terai
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ryan M Weight
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Marlana Orloff
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Michael J Mastrangelo
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Takami Sato
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
178
|
Yi F, Frazzette N, Cruz AC, Klebanoff CA, Siegel RM. Beyond Cell Death: New Functions for TNF Family Cytokines in Autoimmunity and Tumor Immunotherapy. Trends Mol Med 2018; 24:642-653. [PMID: 29880309 DOI: 10.1016/j.molmed.2018.05.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/04/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
Abstract
Originally discovered as an inducer of apoptosis, the TNF-family receptor Fas (CD95, APO-1, TNFRSF6) has more recently been found to have functions beyond cell death, including T cell co-stimulation and promoting terminal differentiation of CD4+ and CD8+ T cells. Other TNF family members also discovered as apoptosis inducers, such as TRAIL (APO-2L, TNFSF10), can promote inflammation through caspase-8. Surprisingly, non-apoptotic signaling through Fas can protect from the autoimmunity seen in Fas deficiency independently from the cell death inducing functions of the receptor. Non-apoptotic Fas signaling can induce tumor cell growth and migration, and impair the efficacy of T cell adoptive immunotherapy. Blocking of non-apoptotic functions of these receptors may be a novel strategy to regulate autoimmunity and inflammation, and enhance antitumor immunity.
Collapse
Affiliation(s)
- Fei Yi
- Immunoregulation Section, Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicholas Frazzette
- Immunoregulation Section, Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anthony C Cruz
- Immunoregulation Section, Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christopher A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, 10065 USA; Parker Institute for Cancer Immunotherapy, MSKCC, New York, NY, 10065 USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
179
|
Advanced Melanoma: Current Treatment Options, Biomarkers, and Future Perspectives. Am J Clin Dermatol 2018; 19:303-317. [PMID: 29164492 DOI: 10.1007/s40257-017-0325-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant melanoma accounts for the highest number of deaths from skin cancer, and the prognosis of patients with stage IV disease has historically been poor. Novel insights into both mutations driving tumorigenesis and immune escape mechanisms of these tumors have led to effective treatment options that have revolutionized the treatment of this disease. Targeting the MAPK kinase pathway (with BRAF and MEK inhibitors), as well as targeting checkpoints, such as cytotoxic T-lymphocyte associated protein 4 (CTLA-4) or programmed death 1 (PD-1), have improved overall survival in patients with late-stage melanoma, and biomarker research for personalized therapy is ongoing for each of these treatment modalities. In this review, we will discuss current first-line treatment options, discuss biomarkers supporting treatment decisions, and give an outlook on (combination) therapies we expect to become relevant in the near future.
Collapse
|
180
|
Sacco JJ, Kalirai H, Kenyani J, Figueiredo CR, Coulson JM, Coupland SE. Recent breakthroughs in metastatic uveal melanoma: a cause for optimism? Future Oncol 2018; 14:1335-1338. [PMID: 29741103 DOI: 10.2217/fon-2018-0116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Joseph J Sacco
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK.,Department of Medical Oncology, Clatterbridge Cancer Centre, Bebington, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Jenna Kenyani
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Carlos R Figueiredo
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK
| | - Judy M Coulson
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK.,Department of Cellular & Molecular Physiology, Institute of Translational Medicine, University of Liverpool, UK
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular & Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, UK.,Department of Cellular Pathology, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
181
|
Kunert A, Debets R. Engineering T cells for adoptive therapy: outsmarting the tumor. Curr Opin Immunol 2018; 51:133-139. [PMID: 29579622 DOI: 10.1016/j.coi.2018.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 02/01/2018] [Accepted: 03/13/2018] [Indexed: 12/20/2022]
Abstract
Adoptive transfer of T cells gene-engineered with antigen-specific receptors, whether it be chimeric antigen receptors (CARs) or T cell receptors (TCRs), has proven its feasibility and therapeutic potential in the treatment of tumors. Despite clinical successes, the majority of patients experiences no or non-sustainable clearance of solid tumors, which is attributed to local T cell evasive mechanisms. A rapidly expanding understanding of molecular and cellular events that contribute to a reduction in numbers and/or activation of intra-tumor T cells has facilitated the development of gene-engineering strategies, enabling T cells to counter immune tolerance. Here, we present an overview of gene-engineering approaches and considerations to improve tumor-selectivity and effectiveness of adoptively transferred T cells.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands.
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC-Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
182
|
Yang J, Manson DK, Marr BP, Carvajal RD. Treatment of uveal melanoma: where are we now? Ther Adv Med Oncol 2018; 10:1758834018757175. [PMID: 29497459 PMCID: PMC5824910 DOI: 10.1177/1758834018757175] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/12/2018] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma, a rare subset of melanoma, is the most common primary intraocular malignancy in adults. Despite effective primary therapy, nearly 50% of patients will develop metastatic disease. Outcomes for those with metastatic disease remain dismal due to a lack of effective therapies. The unique biology and immunology of uveal melanoma necessitates the development of dedicated management and treatment approaches. Ongoing efforts seek to optimize the efficacy of targeted therapy and immunotherapy in both the adjuvant and metastatic setting. This review provides a comprehensive, updated overview of disease biology and risk stratification, the management of primary disease, options for adjuvant therapy, and the current status of treatment strategies for metastatic disease.
Collapse
Affiliation(s)
- Jessica Yang
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Daniel K. Manson
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Brian P. Marr
- Department of Ophthalmology, Columbia University Medical Center, New York, NY, USA
| | - Richard D. Carvajal
- Assistant Professor of Medicine, Director of Experimental Therapeutics and Melanoma Services, Division of Hematology/Oncology, Columbia University Medical Center, 177 Fort Washington Avenue, MHB 6GN-435, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
183
|
Dong J, Chen H. Cardiotoxicity of Anticancer Therapeutics. Front Cardiovasc Med 2018; 5:9. [PMID: 29473044 PMCID: PMC5810267 DOI: 10.3389/fcvm.2018.00009] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/22/2018] [Indexed: 12/31/2022] Open
Abstract
As cancer therapeutics continues to improve and progress, the adverse side effects associated with anticancer treatments have also attracted more attention and have become extensively explored. Consequently, the importance of posttreatment follow-ups is becoming increasingly relevant to the discussion. Contemporary treatment methods, such as tyrosine kinase inhibitors, anthracycline chemotherapy, and immunotherapy regimens are effective in treating different modalities of cancers; however, these reagents act through interference with DNA replication or prevent DNA repair, causing endothelial dysfunction, generating reactive oxygen species, or eliciting non-specific immune responses. Therefore, cardiotoxic effects, such as hypertension, heart failure, and left ventricular dysfunction, arise posttreatment. Rising awareness of cardiovascular complications has led to meticulous attention for the evolution of treatment strategies and carefully monitoring between enhanced treatment effectiveness and minimization of adverse toxicity to the cardiovasculature, in which psychological assessments, early detection methods such as biomarkers, magnetic resonance imaging, and various drugs to reverse the damage from cardiotoxic events are more prevalent and their emphasis has increased tremendously. Fully understanding the mechanisms by which the risk factors action for various patients undergoing cancer treatment is also becoming more prevalent in preventing cardiotoxicity down the line.
Collapse
Affiliation(s)
- Jerry Dong
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Case Western Reserve University, Cleveland, OH, United States
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Hong Chen
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Surgery, Vascular Biology Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
184
|
Crompton JG, Klemen N, Kammula US. Metastasectomy for Tumor-Infiltrating Lymphocytes: An Emerging Operative Indication in Surgical Oncology. Ann Surg Oncol 2018; 25:565-572. [PMID: 29188500 PMCID: PMC5758677 DOI: 10.1245/s10434-017-6266-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Indexed: 12/22/2022]
Abstract
Adoptive cell transfer (ACT) of tumor-infiltrating lymphocytes (TILs) is an emerging immunotherapy for metastatic cancer. Surgeons play a central role in ACT treatments by performing resection of tumors from which TILs are isolated. It is important that surgeons have familiarity with this emerging treatment method because it is increasingly performed for an expanding variety of solid tumors at institutions around the world. This report offers a brief introduction to ACT for cancer, highlights historical milestones in its development, and provides patient selection and operative considerations for surgeons called upon to perform metastasectomy for the purpose of isolating TILs.
Collapse
Affiliation(s)
- Joseph G Crompton
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Nicholas Klemen
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Udai S Kammula
- Division of Surgical Oncology, Department of Surgery, University of Pittsburgh School of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
185
|
Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent Advances in Targeting CD8 T-Cell Immunity for More Effective Cancer Immunotherapy. Front Immunol 2018; 9:14. [PMID: 29403496 PMCID: PMC5786548 DOI: 10.3389/fimmu.2018.00014] [Citation(s) in RCA: 322] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer treatment have emerged from new immunotherapies targeting T-cell inhibitory receptors, including cytotoxic T-lymphocyte associated antigen (CTLA)-4 and programmed cell death (PD)-1. In this context, anti-CTLA-4 and anti-PD-1 monoclonal antibodies have demonstrated survival benefits in numerous cancers, including melanoma and non-small-cell lung carcinoma. PD-1-expressing CD8+ T lymphocytes appear to play a major role in the response to these immune checkpoint inhibitors (ICI). Cytotoxic T lymphocytes (CTL) eliminate malignant cells through recognition by the T-cell receptor (TCR) of specific antigenic peptides presented on the surface of cancer cells by major histocompatibility complex class I/beta-2-microglobulin complexes, and through killing of target cells, mainly by releasing the content of secretory lysosomes containing perforin and granzyme B. T-cell adhesion molecules and, in particular, lymphocyte-function-associated antigen-1 and CD103 integrins, and their cognate ligands, respectively, intercellular adhesion molecule 1 and E-cadherin, on target cells, are involved in strengthening the interaction between CTL and tumor cells. Tumor-specific CTL have been isolated from tumor-infiltrating lymphocytes and peripheral blood lymphocytes (PBL) of patients with varied cancers. TCRβ-chain gene usage indicated that CTL identified in vitro selectively expanded in vivo at the tumor site compared to autologous PBL. Moreover, functional studies indicated that these CTL mediate human leukocyte antigen class I-restricted cytotoxic activity toward autologous tumor cells. Several of them recognize truly tumor-specific antigens encoded by mutated genes, also known as neoantigens, which likely play a key role in antitumor CD8 T-cell immunity. Accordingly, it has been shown that the presence of T lymphocytes directed toward tumor neoantigens is associated with patient response to immunotherapies, including ICI, adoptive cell transfer, and dendritic cell-based vaccines. These tumor-specific mutation-derived antigens open up new perspectives for development of effective second-generation therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Aurélie Durgeau
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,ElyssaMed, Paris Biotech Santé, Paris, France
| | - Yasemin Virk
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
186
|
Ren L, Matsuda T, Deng B, Kiyotani K, Kato T, Park JH, Seiwert TY, Vokes EE, Agrawal N, Nakamura Y. Similarity and difference in tumor-infiltrating lymphocytes in original tumor tissues and those of in vitro expanded populations in head and neck cancer. Oncotarget 2017; 9:3805-3814. [PMID: 29423084 PMCID: PMC5790501 DOI: 10.18632/oncotarget.23454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/10/2017] [Indexed: 01/04/2023] Open
Abstract
Though adoptive tumor-infiltrating lymphocyte (TIL) therapy has been explored in clinical trials for many years, there is little information for the clonotype composition between TILs in original tumor tissues and TILs that were in vitro expanded and infused to cancer patients. To investigate the similarity/difference in TILs in original tumor tissues and those of in vitro expanded populations in squamous cell carcinoma of head and neck (SCCHN) as well as their correlation with somatic mutations in cancer cells, we performed whole exome analysis, expression profile analysis of immune-related genes, and T cell receptor (TCR) analysis of original TILs and in vitro expanded TILs in 8 surgically-resected HPV-negative fresh tumors with SCCHN. We found an unusually high number of non-synonymous somatic mutations (4290, 1779 and 901 mutations) in three SCCHN tumors, in which we identified mutations in mismatch repair genes, MSH2 or MSH4, or a DNA polymerase gene, POLE. Interestingly, dominant TCR clonotypes of expanded CD8+ TILs derived from these three tumors revealed high similarity to those in original tumors while for remaining tumors with the lower mutational load, we found that T cell clonotypes between TILs in original tumor tissues and those expanded in vitro were almost entirely different. Our findings might provide clinically useful information for identification of tumor-antigen-specific T cell clones that may lead to further improvement of adoptive TIL therapy for SCCHN patients.
Collapse
Affiliation(s)
- Lili Ren
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Cytotherapy Laboratory, Shenzhen People's Hospital (The second Clinical Medical College of Jinan University), Shenzhen, China
| | - Tatsuo Matsuda
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Boya Deng
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Taigo Kato
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tanguy Y Seiwert
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Everett E Vokes
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Nishant Agrawal
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA.,Department of Surgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
187
|
McWilliams RR, Allred JB, Slostad JA, Katipamula R, Dronca RS, Rumilla KM, Erickson LA, Bryce AH, Joseph RW, Kottschade LA, King DM, Leitch JM, Markovic SN. NCCTG N0879 (Alliance): A randomized phase 2 cooperative group trial of carboplatin, paclitaxel, and bevacizumab ± everolimus for metastatic melanoma. Cancer 2017; 124:537-545. [PMID: 29044496 DOI: 10.1002/cncr.31072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Despite the success of immune checkpoint and targeted therapy, many patients with melanoma ultimately require further treatment. The combination of carboplatin, paclitaxel, and bevacizumab (CPB) has demonstrated promising activity in a single-arm study. In the current study, the authors performed a randomized phase 2 study to confirm efficacy and to determine whether adding everolimus would increase the activity of the combination. METHODS Through the North Central Cancer Treatment Group, a total of 149 patients with unresectable AJCC 6th edition stage IV melanoma were randomized from May 2010 to May 2014 to either CPB or CPB with everolimus (CPBE). The primary endpoint was progression-free survival (PFS), with secondary endpoints of overall survival (OS), response rate, and tolerability. RESULTS The CPB and CPBE treatment arms were balanced with regard to age (median age: 59 years vs 58 years) and high lactate dehydrogenase (48% vs 51%), but were unbalanced with regard to sex (male sex: 72% vs 55%; P = .03). Overall, there was no difference noted with regard to PFS, with a median PFS of 5.6 months for CPB versus 5.1 months for CPBE (hazard ratio [HR], 1.14; 95% confidence interval [95% CI], 0.81-1.62 [P = .44]), or for OS, with a median OS of 14.5 months for CPB versus 10.8 months for CPBE (HR, 1.16; 95% CI, 0.84-1.84). The confirmed response rate was 13% for CPB and 23% for CPBE (P = .13). Toxicity was higher for CPBE compared with CPB (83% for grade 3 + and 14% for grade 4 + vs 63% for grade 3 + and 11% for grade 4+, respectively) (toxicities were graded using the Cancer Therapy Evaluation Program of the National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). Common grade 3 + toxicities were neutropenia, leukopenia, and fatigue, which occurred in both treatment arms with comparable frequency. CONCLUSIONS Both experimental arms demonstrated activity, with a PFS of >5 months. However, the addition of everolimus to CPB failed to improve outcomes, with increased toxicity noted. These findings replicate the moderate antitumor activity of CPB, with future development possibly in combination with targeted or immunotherapy. Cancer 2018;124:537-45. © 2017 American Cancer Society.
Collapse
Affiliation(s)
| | - Jacob B Allred
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota
| | | | | | - Roxana S Dronca
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | | | - Lori A Erickson
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota
| | - Alan H Bryce
- Division of Hematology/Oncology, Mayo Clinic in Arizona, Scottsdale, Arizona
| | - Richard W Joseph
- Division of Hematology/Oncology, Mayo Clinic in Florida, Jacksonville, Florida
| | | | - David M King
- Unity Hospital, Metro Minnesota Community Oncology Research Consortium, Saint Louis Park, Minnesota
| | | | | |
Collapse
|
188
|
Fournier C, Martin F, Zitvogel L, Kroemer G, Galluzzi L, Apetoh L. Trial Watch: Adoptively transferred cells for anticancer immunotherapy. Oncoimmunology 2017; 6:e1363139. [PMID: 29147628 DOI: 10.1080/2162402x.2017.1363139] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 07/29/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022] Open
Abstract
Immunotherapies aimed at strengthening immune effector responses against malignant cells are growing at exponential rates. Alongside, the impressive benefits obtained by patients with advanced melanoma who received adoptively transferred tumor-infiltrating lymphocytes (TILs) have encouraged the scientific community to pursue adoptive cell transfer (ACT)-based immunotherapy. ACT involves autologous or allogenic effector lymphocytes that are generally obtained from the peripheral blood or resected tumors, expanded and activated ex vivo, and administered to lymphodepleted patients. ACT may be optionally associated with chemo- and/or immunotherapeutics, with the overall aim of enhancing the proliferation, persistence and functionality of infused cells, as well as to ensure their evolution in an immunological permissive local and systemic microenvironment. In addition, isolated lymphocytes can be genetically engineered to endow them with the ability to target a specific tumor-associated antigen (TAA), to increase their lifespan, and/or to reduce their potential toxicity. The infusion of chimeric antigen receptor (CAR)-expressing cytotoxic T lymphocytes redirected against CD19 has shown promising clinical efficacy in patients with B-cell malignancies. Accordingly, the US Food and Drug Administration (FDA) has recently granted 'breakthrough therapy' designation to a CAR-based T-cell therapy (CTL019) for patients with B-cell malignancies. Considerable efforts are now being devoted to the development of efficient ACT-based immunotherapies for non-hematological neoplasms. In this Trial Watch, we summarize recent clinical advances on the use of ACT for oncological indications.
Collapse
Affiliation(s)
- Carole Fournier
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - François Martin
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France.,Université Pierre et Marie Curie/Paris VI, Paris.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France.,Faculté de Médecine, Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
189
|
Chan PY, Hall P, Hay G, Cohen VML, Szlosarek PW. A major responder to ipilimumab and nivolumab in metastatic uveal melanoma with concomitant autoimmunity. Pigment Cell Melanoma Res 2017. [PMID: 28640512 DOI: 10.1111/pcmr.12607] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of immune checkpoint inhibition has led to major improvements in outcome for patients with metastatic cutaneous melanoma. The combination of ipilimumab and nivolumab has demonstrated greater activity over single-agent immunotherapy in phase III trials. Clinical trials of combination CTLA-4 and PD-1 inhibition are underway in uveal melanoma, for which there are currently no data. Here, we present the case of a 74-year-old male patient with metastatic uveal melanoma, who was treated with a combination of ipilimumab and nivolumab. He developed sequential autoimmune transaminitis, diabetes and uveitis, which necessitated discontinuation of maintenance nivolumab 3 months after commencement of treatment. The patient continues to demonstrate an ongoing partial response 10 months from the initial combination immunotherapy, with the evidence of depigmentation of the primary ocular tumour.
Collapse
Affiliation(s)
- Pui Ying Chan
- Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - Peter Hall
- Department of Medical Oncology, St Bartholomew's Hospital, London, UK
| | - Gordon Hay
- Department of Ocular Oncology, Moorfields Eye Hospital, London, UK.,UCL Institute of Ophthalmology, London, UK
| | - Victoria M L Cohen
- Department of Ocular Oncology, Moorfields Eye Hospital, London, UK.,Department of Ocular Oncology, St Bartholomew's Hospital, London, UK
| | - Peter W Szlosarek
- Department of Medical Oncology, St Bartholomew's Hospital, London, UK.,Centre for Molecular Oncology, Barts Cancer Institute, London, UK
| |
Collapse
|
190
|
Adopting a new stance on immunotherapy for uveal melanoma. Lancet Oncol 2017; 18:702-704. [DOI: 10.1016/s1470-2045(17)30254-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 03/31/2017] [Indexed: 12/12/2022]
|
191
|
|