1
|
Pham JP, On L, Ardolino L, Hurwitz J, Salaun H, Sim HW, Joshua AM. Efficacy of immune checkpoint inhibition in metastatic uveal melanoma: a systematic review and meta-analysis. Melanoma Res 2023; 33:316-325. [PMID: 37199712 DOI: 10.1097/cmr.0000000000000900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Metastatic uveal melanoma (mUM) has historically been associated with short survival and limited effective treatments. Immune checkpoint inhibitors (ICIs) have been trialed in mUM; however, robust conclusions regarding their efficacy are difficult to draw given small study sizes and heterogeneous patient populations. Five databases were searched using a combination of 'ICI' and 'mUM' headings, and data on patient demographics, objective response rate (ORR), overall survival (OS) and progression-free survival (PFS) were extracted. Pooled ORR was calculated using a random effects model and the inverse variance method. Available Kaplan-Meier OS and PFS curves were used to construct summary OS and PFS plots, from which median values were derived. Pooled ORR was 9.2% overall (95% CI 7.2-11.8) [4.1% for anti-CTLA4 (95% CI 2.1-7.7), 7.1% for anti-PD(L)1 (95% CI 4.5-10.9) and 13.5% for anti-CTLA4 plus anti-PD1 (95% CI 10.0-18.0)]. Median OS was 11.5 months overall (95% CI 9.5-13.8) [8.0 months for anti-CTLA4 (95% CI 5.5-9.9), 11.7 months for anti-PD(L)1 (95% CI 9.0-14.0) and 16.0 months for ipilimumab plus anti-PD1 (95% CI 11.5-17.7) ( P < 0.001)]. Median PFS was 3.0 months overall (95% CI 2.9-3.1). ICIs have limited efficacy in mUM and a recommendation for their use must consider the balance of benefit and risk for individual patients if no other options are available. Further biomarker profiling studies may be helpful in assessing which patients will benefit from ICIs, in particular the addition of ipilimumab to anti-PD1 therapy.
Collapse
Affiliation(s)
- James P Pham
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
| | - Lawrence On
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
- Department of Ophthalmology, Sydney Eye Hospital, Sydney, NSW, Australia
| | - Luke Ardolino
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
| | - Joshua Hurwitz
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
| | - Helene Salaun
- Department of Medical Oncology, PSL Research University, Institut Curie, Paris, France
| | - Hao-Wen Sim
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
- Department of Medical Oncology, NHMRC Clinical Trials Centre, University of Sydney, Camperdown
| | - Anthony M Joshua
- Department of Medical Oncology, The Kinghorn Cancer Centre, St. Vincent's Hospital Sydney
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Hospital, Darlinghurst
- Melanoma Institute Australia, University of Sydney, North Sydney, NSW, Australia
| |
Collapse
|
2
|
Shen L, Wu Y, Qi H, Jiang Y, Jin J, Cao F, Chen S, Yang Y, Huang T, Song Z, Chen Q, Zhang Y, Mo J, Li D, Zhang X, Fan W. Inducible Regulatory T Cell Predicts Efficacy of PD‐1 Blockade Therapy in Melanoma. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Lujun Shen
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Ying Wu
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Han Qi
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Yiquan Jiang
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Jietian Jin
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
- Department of Pathology Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
| | - Fei Cao
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Shuanggang Chen
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Yuanzhong Yang
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
- Department of Pathology Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
| | - Tao Huang
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Ze Song
- Department of Medical Oncology Seventh Affiliated Hospital of Sun Yat‐sen University Shenzhen 518107 P. R. China
| | - Qifeng Chen
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| | - Yinqi Zhang
- Zhong Shan School of Medicine Sun Yat‐sen University Guangzhou 510080 P. R. China
| | - Jinqing Mo
- Zhong Shan School of Medicine Sun Yat‐sen University Guangzhou 510080 P. R. China
| | - Dandan Li
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
- Department of Biological Therapy Center Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
| | - Xiaoshi Zhang
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
- Department of Biological Therapy Center Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
| | - Weijun Fan
- Department of Minimally Invasive Interventional Therapy Sun Yat‐sen University Cancer Center Guangzhou 510060 P. R. China
- State Key Laboratory of Oncology in South China Collaborative Innovation Center of Cancer Medicine Sun Yat‐sen University Guangzhou 510060 P. R. China
| |
Collapse
|
3
|
Rossi E, Croce M, Reggiani F, Schinzari G, Ambrosio M, Gangemi R, Tortora G, Pfeffer U, Amaro A. Uveal Melanoma Metastasis. Cancers (Basel) 2021; 13:5684. [PMID: 34830841 PMCID: PMC8616038 DOI: 10.3390/cancers13225684] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023] Open
Abstract
Uveal melanoma (UM) is characterized by relatively few, highly incident molecular alterations and their association with metastatic risk is deeply understood. Nevertheless, this knowledge has so far not led to innovative therapies for the successful treatment of UM metastases or for adjuvant therapy, leaving survival after diagnosis of metastatic UM almost unaltered in decades. The driver mutations of UM, mainly in the G-protein genes GNAQ and GNA11, activate the MAP-kinase pathway as well as the YAP/TAZ pathway. At present, there are no drugs that target the latter and this likely explains the failure of mitogen activated kinase kinase inhibitors. Immune checkpoint blockers, despite the game changing effect in cutaneous melanoma (CM), show only limited effects in UM probably because of the low mutational burden of 0.5 per megabase and the unavailability of antibodies targeting the main immune checkpoint active in UM. The highly pro-tumorigenic microenvironment of UM also contributes to therapy resistance. However, T-cell redirection by a soluble T-cell receptor that is fused to an anti-CD3 single-chain variable fragment, local, liver specific therapy, new immune checkpoint blockers, and YAP/TAZ specific drugs give new hope to repeating the success of innovative therapy obtained for CM.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
| | - Michela Croce
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Francesco Reggiani
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Marianna Ambrosio
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Rosaria Gangemi
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Ulrich Pfeffer
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Adriana Amaro
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| |
Collapse
|
4
|
How to Make Immunotherapy an Effective Therapeutic Choice for Uveal Melanoma. Cancers (Basel) 2021; 13:cancers13092043. [PMID: 33922591 PMCID: PMC8122936 DOI: 10.3390/cancers13092043] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/26/2021] [Accepted: 04/21/2021] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Despite improvements in the early identification and successful control of primary uveal melanoma, 50% of patients will develop metastatic disease with only marginal improvements in survival. This review focuses on the tumor microenvironment and the cross-talk between tumor and immune cells in a tumor characterized by low mutational load, the induction of immune-suppressive cells, and the expression of alternative immune checkpoint molecules. The choice of combining different strategies of immunotherapy remains a feasible and promising option on selected patients. Abstract Uveal melanoma (UM), though a rare form of melanoma, is the most common intraocular tumor in adults. Conventional therapies of primary tumors lead to an excellent local control, but 50% of patients develop metastases, in most cases with lethal outcome. Somatic driver mutations that act on the MAP-kinase pathway have been identified, yet targeted therapies show little efficacy in the clinics. No drugs are currently available for the G protein alpha subunitsGNAQ and GNA11, which are the most frequent driver mutations in UM. Drugs targeting the YAP–TAZ pathway that is also activated in UM, the tumor-suppressor gene BRCA1 Associated Protein 1 (BAP1) and the Splicing Factor 3b Subunit 1 gene (SF3B1) whose mutations are associated with metastatic risk, have not been developed yet. Immunotherapy is highly effective in cutaneous melanoma but yields only poor results in the treatment of UM: anti-PD-1 and anti-CTLA-4 blocking antibodies did not meet the expectations except for isolated cases. Here, we discuss how the improved knowledge of the tumor microenvironment and of the cross-talk between tumor and immune cells could help to reshape anti-tumor immune responses to overcome the intrinsic resistance to immune checkpoint blockers of UM. We critically review the dogma of low mutational load, the induction of immune-suppressive cells, and the expression of alternative immune checkpoint molecules. We argue that immunotherapy might still be an option for the treatment of UM.
Collapse
|
5
|
Masaoutis C, Kokkali S, Theocharis S. Immunotherapy in uveal melanoma: novel strategies and opportunities for personalized treatment. Expert Opin Investig Drugs 2021; 30:555-569. [PMID: 33650931 DOI: 10.1080/13543784.2021.1898587] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Uveal melanoma (UM) is the most common intraocular cancer and represents a discrete subtype of melanoma. Metastatic disease, which occurs in half of patients, has a dismal prognosis. Immunotherapy with immune checkpoint inhibitors has produced promising results in cutaneous melanoma but has failed to show analogous efficacy in metastatic UM. This is attributable to UM's distinct genetics and its complex interaction with the immune system. Hence, more efficacious immunotherapeutic approaches are under investigation. AREAS COVERED We discuss those novel immunotherapeutic strategies in clinical and preclinical studies for advanced disease and which are thought to overcome the hurdles set by UM in terms of immune recognition. We also highlight the need to determine predictive markers in relation to these strategies to improve clinical outcomes. We used a simple narrative analysis to summarize the data. The search methodology is located in the Introduction. EXPERT OPINION Novel immunotherapeutic strategies focus on transforming immune excluded tumor microenvironment in metastatic UM to T cell inflamed. Preliminary results of approaches such as vaccines, adoptive cell transfer and other novel molecules are encouraging. Factors such as HLA compatibility and expression level of targeted antigens should be considered to optimize personalized management.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefania Kokkali
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,First Medical Oncology Clinic, Saint-Savvas Anticancer Hospital, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Abstract
Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. UMs are usually initiated by a mutation in GNAQ or GNA11, unlike cutaneous melanomas, which usually harbour a BRAF or NRAS mutation. The annual incidence in Europe and the USA is ~6 per million population per year. Risk factors include fair skin, light-coloured eyes, congenital ocular melanocytosis, ocular melanocytoma and the BAP1-tumour predisposition syndrome. Ocular treatment aims at preserving the eye and useful vision and, if possible, preventing metastases. Enucleation has largely been superseded by various forms of radiotherapy, phototherapy and local tumour resection, often administered in combination. Ocular outcomes are best with small tumours not extending close to the optic disc and/or fovea. Almost 50% of patients develop metastatic disease, which usually involves the liver, and is usually fatal within 1 year. Although UM metastases are less responsive than cutaneous melanoma to chemotherapy or immune checkpoint inhibitors, encouraging results have been reported with partial hepatectomy for solitary metastases, with percutaneous hepatic perfusion with melphalan or with tebentafusp. Better insight into tumour immunology and metabolism may lead to new treatments.
Collapse
|
7
|
Wessely A, Steeb T, Erdmann M, Heinzerling L, Vera J, Schlaak M, Berking C, Heppt MV. The Role of Immune Checkpoint Blockade in Uveal Melanoma. Int J Mol Sci 2020; 21:ijms21030879. [PMID: 32013269 PMCID: PMC7037664 DOI: 10.3390/ijms21030879] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/25/2022] Open
Abstract
Uveal melanoma (UM) represents the most common intraocular malignancy in adults and accounts for about 5% of all melanomas. Primary disease can be effectively controlled by several local therapy options, but UM has a high potential for metastatic spread, especially to the liver. Despite its clinical and genetic heterogeneity, therapy of metastatic UM has largely been adopted from cutaneous melanoma (CM) with discouraging results until now. The introduction of antibodies targeting CTLA-4 and PD-1 for immune checkpoint blockade (ICB) has revolutionized the field of cancer therapy and has achieved pioneering results in metastatic CM. Thus, expectations were high that patients with metastatic UM would also benefit from these new therapy options. This review provides a comprehensive and up-to-date overview on the role of ICB in UM. We give a summary of UM biology, its clinical features, and how it differs from CM. The results of several studies that have been investigating ICB in metastatic UM are presented. We discuss possible reasons for the lack of efficacy of ICB in UM compared to CM, highlight the pitfalls of ICB in this cancer entity, and explain why other immune-modulating therapies could still be an option for future UM therapies.
Collapse
Affiliation(s)
- Anja Wessely
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Theresa Steeb
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Michael Erdmann
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Julio Vera
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Max Schlaak
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Frauenlobstr. 9-11, 80337 Munich, Germany;
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
| | - Markus Vincent Heppt
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich Alexander University, Ulmenweg 18, 91054 Erlangen, Germany; (A.W.); (T.S.); (M.E.); (L.H.); (J.V.); (C.B.)
- Correspondence: ; Tel.: +49-9131-85-35747
| |
Collapse
|
8
|
Basile MS, Mazzon E, Fagone P, Longo A, Russo A, Fallico M, Bonfiglio V, Nicoletti F, Avitabile T, Reibaldi M. Immunobiology of Uveal Melanoma: State of the Art and Therapeutic Targets. Front Oncol 2019; 9:1145. [PMID: 31750244 PMCID: PMC6848265 DOI: 10.3389/fonc.2019.01145] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Uveal Melanoma (UM) represents the most common primary intraocular malignant tumor in adults. Although it originates from melanocytes as cutaneous melanoma, it shows significant clinical and biological differences with the latter, including high resistance to immune therapy. Indeed, UM can evade immune surveillance via multiple mechanisms, such as the expression of inhibitory checkpoints (e.g., PD-L1, CD47, CD200) and the production of IDO-1 and soluble FasL, among others. More in-depth understanding of these mechanisms will suggest potential targets for the design of novel and more effective management strategies for UM patients.
Collapse
Affiliation(s)
- Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, Messina, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Longo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Matteo Fallico
- Department of Ophthalmology, University of Catania, Catania, Italy
| | | | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Michele Reibaldi
- Department of Ophthalmology, University of Catania, Catania, Italy
| |
Collapse
|
9
|
Bol KF, Mensink HW, Aarntzen EHJG, Schreibelt G, Keunen JEE, Coulie PG, de Klein A, Punt CJA, Paridaens D, Figdor CG, de Vries IJM. Long overall survival after dendritic cell vaccination in metastatic uveal melanoma patients. Am J Ophthalmol 2014; 158:939-47. [PMID: 25038326 DOI: 10.1016/j.ajo.2014.07.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 12/27/2022]
Abstract
PURPOSE To assess the safety and efficacy of dendritic cell vaccination in metastatic uveal melanoma. DESIGN Interventional case series. METHODS We analyzed 14 patients with metastatic uveal melanoma treated with dendritic cell vaccination. Patients with metastatic uveal melanoma received at least 3 vaccinations with autologous dendritic cells, professional antigen-presenting cells loaded with melanoma antigens gp100 and tyrosinase. The main outcome measures were safety, immunologic response, and overall survival. RESULTS Tumor-specific immune responses were induced with dendritic cell vaccination in 4 (29%) of 14 patients. Dendritic cell-vaccinated patients showed a median overall survival with metastatic disease of 19.2 months, relatively long compared with that reported in the literature. No severe treatment-related toxicities (common toxicity criteria grade 3 or 4) were observed. CONCLUSIONS Dendritic cell vaccination is feasible and safe in metastatic uveal melanoma. Dendritic cell-based immunotherapy is potent to enhance the host's antitumor immunity against uveal melanoma in approximately one third of patients. Compared with other prospective studies with similar inclusion criteria, dendritic cell vaccination may be associated with longer than average overall survival in patients with metastatic uveal melanoma.
Collapse
Affiliation(s)
- Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Hanneke W Mensink
- Department of Ophthalmology, Rotterdam Eye Hospital, Rotterdam, the Netherlands; Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Erik H J G Aarntzen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Jan E E Keunen
- Department of Ophthalmology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Pierre G Coulie
- De Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, the Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, Amsterdam, the Netherlands
| | - Dion Paridaens
- Department of Ophthalmology, Rotterdam Eye Hospital, Rotterdam, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands.
| |
Collapse
|
10
|
Abstract
Mechanisms that maintain ocular immune privilege may contribute to ocular tumor progression by inhibiting tumoricidal immune responses. Consistent with that notion are observations from transplantable tumor models in mice demonstrating that the tumoricidal activity of CD8(+) cytolytic T lymphocytes (CTL) may be inhibited directly by interfering with CTL effector function in the eye or indirectly by abrogating the effector function of CD8+ T cell-activated intratumoral macrophages that are critical for ocular tumor rejection. In addition, epigenetic gene regulation by factors within the ocular tumor environment favors the generation of tumor variants that are resistant to CD8(+) CTL. Intratumoral macrophages may be essential for eliminating these variants because, unlike CTL, their tumoricidal activity is nonspecific. Hence, the inhibition of macrophage effector function within the eye, presumably to preserve immune privilege by minimizing ocular immunopathology, may hasten the outgrowth of tumor escape variants which contributes to ocular tumor progression.
Collapse
Affiliation(s)
- Kyle C McKenna
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
11
|
Abstract
The notion that the immune system might control the growth of tumors was suggested over 100 years ago by the eminent microbiologist Paul Ehrlich. This concept was refined and expanded by Burnet and Thomas 50 years later with their articulation of the "immune surveillance" hypothesis. In its simplest form, the immune surveillance hypothesis suggests that neoplasms arise spontaneously and express novel antigens that are recognized by the immune system, which either eliminates the tumors or restrains their growth. Within the eye, immune responses are controlled and sometimes profoundly inhibited - a condition known as immune privilege. Immune privilege in the eye is the result of a complex array of anatomical, physiological, and immunoregulatory mechanisms that prevent the induction and expression of many immune responses. Tumors arising in the eye would seem to have an advantage in evading immune surveillance due to ocular immune privilege. Uveal melanoma, the most common and malignant intraocular tumor in adults, not only benefits from the immune privilege of the eye but also has adopted many of the mechanisms that contribute to ocular immune privilege as a strategy for protecting uveal melanoma cells once they leave the sanctuary of the eye and are disseminated systemically in the form of metastases. Although the immune system possesses a battery of effector mechanisms designed to rid the body of neoplasms, tumors are capable of rapidly evolving and countering even the most sophisticated immunological effector mechanisms. To date, tumors seem to be winning this arms race, but an increased understanding of these mechanisms should provide insights for designing immunotherapy that was envisioned over half a century ago, but has failed to materialize to date.
Collapse
Affiliation(s)
- Jerry Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9057, USA.
| |
Collapse
|
12
|
Uveal melanoma cell-based vaccines express MHC II molecules that traffic via the endocytic and secretory pathways and activate CD8+ cytotoxic, tumor-specific T cells. Cancer Immunol Immunother 2009; 59:103-12. [PMID: 19557412 DOI: 10.1007/s00262-009-0729-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 06/02/2009] [Indexed: 01/22/2023]
Abstract
We are exploring cell-based vaccines as a treatment for the 50% of patients with large primary uveal melanomas who develop lethal metastatic disease. MHC II uveal melanoma vaccines are MHC class I(+) uveal melanoma cells transduced with CD80 genes and MHC II genes syngeneic to the recipient. Previous studies demonstrated that the vaccines activate tumor-specific CD4(+) T cells from patients with metastatic uveal melanoma. We have hypothesized that vaccine potency is due to the absence of the MHC II-associated invariant chain (Ii). In the absence of Ii, newly synthesized MHC II molecules traffic intracellularly via a non-traditional pathway where they encounter and bind novel tumor peptides. Using confocal microscopy, we now confirm this hypothesis and demonstrate that MHC II molecules are present in both the endosomal and secretory pathways in vaccine cells. We also demonstrate that uveal melanoma MHC II vaccines activate uveal melanoma-specific, cytolytic CD8(+) T cells that do not lyse normal fibroblasts or other tumor cells. Surprisingly, the CD8(+) T cells are cytolytic for HLA-A syngeneic and MHC I-mismatched uveal melanomas. Collectively, these studies demonstrate that MHC II uveal melanoma vaccines are potent activators of tumor-specific CD4(+) and CD8(+) T cells and suggest that the non-conventional intracellular trafficking pattern of MHC II may contribute to their enhanced immunogenicity. Since MHC I compatibility is unnecessary for the activation of cytolytic CD8(+) T cells, the vaccines could be used in uveal melanoma patients without regard to MHC I genotype.
Collapse
|
13
|
McKenna KC, Beatty KM, Bilonick RA, Schoenfield L, Lathrop KL, Singh AD. Activated CD11b+ CD15+ granulocytes increase in the blood of patients with uveal melanoma. Invest Ophthalmol Vis Sci 2009; 50:4295-303. [PMID: 19369244 DOI: 10.1167/iovs.08-3012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine whether activated CD11b(+) CD15(+) granulocytes increase in the blood of patients with uveal melanoma. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated by density gradient centrifugation from the blood of patients with primary choroidal/ciliochoroidal uveal melanomas (six women, four men; age range, 46-91 years) and healthy control donors (14 women, 10 men; age range, 50-81 years). The expression of CD15 and CD68 on CD11b(+) myeloid cells within PBMCs and primary uveal melanomas was evaluated by flow cytometry. CD3zeta chain expression by CD3epsilon(+) T cells in PBMCs and within primary uveal melanomas was measured as an indirect indication of T-cell function. RESULTS The percentage of CD11b(+) cells in PBMCs of patients with uveal melanoma increased 1.8-fold in comparison to healthy donors and comprised three subsets: CD68 negative CD15(+) granulocytes, which increased 4.1-fold; CD68(-) CD15(-) cells, which increased threefold; and CD68(+) CD15(low) cells, which were unchanged. A significant (2.7-fold) reduction in CD3zeta chain expression on CD3epsilon(+) T cells, a marker of T-cell dysfunction, was observed in PBMCs of patients with uveal melanoma in comparison with healthy control subjects and correlated significantly with the percentage of CD11b(+) cells in PBMCs. CD3zeta chain expression on T cells within primary tumors was equivalent to CD3zeta expression in PBMCs of the same patient in four of five patients analyzed. CONCLUSIONS Activated CD11b(+) CD15(+) granulocytes expand in the blood of patients with uveal melanoma and may contribute to immune evasion by ocular tumors by inhibiting T-cell function via decreasing CD3zeta chain expression.
Collapse
Affiliation(s)
- Kyle C McKenna
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Polak ME, Borthwick NJ, Johnson P, Hungerford JL, Higgins B, Di Palma S, Jager MJ, Cree IA. Presence and phenotype of dendritic cells in uveal melanoma. Br J Ophthalmol 2007; 91:971-6. [PMID: 17347328 PMCID: PMC1955657 DOI: 10.1136/bjo.2006.110908] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2007] [Indexed: 01/10/2023]
Abstract
BACKGROUND Uveal melanoma arises in an immune-privileged site and can itself add to the immunosuppressive environment. Previous studies on cutaneous melanoma have shown the presence of tolerogenic dendritic cells (DCs), which could play an important role in the progression of the tumour. AIM To examine the presence and functional status of DCs in a small series of uveal melanomas. METHODS 10 cases of uveal melanoma were examined for the expression of FXIIIa, CD68, human leucocyte antigen (HLA)-DR, CD40, CD83, transforming growth factor betaR1 and indolamine 2,3 dioxygenase by immunohistochemical analysis on sections embedded in paraffin wax. RESULTS CD68-positive macrophages were present in all of the tumours and were evenly distributed throughout. DCs expressing FXIIIa-positive were seen in 7 cases, and were often found concentrated in foci within the tumour mass. These cells were dendritic and expressed high levels of HLA-DR. The DCs did not express the maturation markers CD83 or CD40. In one case, concentration of DCs around the area of tumour necrosis was observed, and some of these cells expressed CD83. CONCLUSION Numerous tolerising antigen-presenting cells may play a role in melanoma-related immunosuppression in the eye, although activation of DCs may be associated with tumour necrosis.
Collapse
Affiliation(s)
- Marta E Polak
- Translational Oncology Research Centre, Queen Alexandra Hospital, Southwick Hill, Portsmouth PO6 3LY, UK.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Bosch JJ, Thompson JA, Srivastava MK, Iheagwara UK, Murray TG, Lotem M, Ksander BR, Ostrand-Rosenberg S. MHC class II-transduced tumor cells originating in the immune-privileged eye prime and boost CD4(+) T lymphocytes that cross-react with primary and metastatic uveal melanoma cells. Cancer Res 2007; 67:4499-506. [PMID: 17483366 DOI: 10.1158/0008-5472.can-06-3770] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Uveal melanoma, the most common malignancy of the eye, has a 50% rate of liver metastases among patients with large primary tumors. Several therapies prolong survival of metastatic patients; however, none are curative and no patients survive. Therefore, we are exploring immunotherapy as an alternative or adjunctive treatment. Uveal melanoma may be particularly appropriate for immunotherapy because primary tumors arise in an immune-privileged site and may express antigens to which the host is not tolerized. We are developing MHC class II (MHC II)-matched allogeneic, cell-based uveal melanoma vaccines that activate CD4(+) T lymphocytes, which are key cells for optimizing CD8(+) T-cell immunity, facilitating immune memory, and preventing tolerance. Our previous studies showed that tumor cells genetically modified to express costimulatory and MHC II molecules syngeneic to the recipient are potent inducers of antitumor immunity. Because the MHC II-matched allogeneic vaccines do not express the accessory molecule, Invariant chain, they present MHC II-restricted peptides derived from endogenously encoded tumor antigens. We now report that MHC II-matched allogeneic vaccines, prepared from primary uveal melanomas that arise in the immune-privileged eye, prime and boost IFNgamma-secreting CD4(+) T cells from the peripheral blood of either healthy donors or uveal melanoma patients that cross-react with primary uveal melanomas from other patients and metastatic tumors. In contrast, vaccines prepared from metastatic cells in the liver are less effective at activating CD4(+) T cells, suggesting that tumor cells originating in immune-privileged sites may have enhanced capacity for inducing antitumor immunity and for serving as immunotherapeutic agents.
Collapse
Affiliation(s)
- Jacobus J Bosch
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Mittica N, Vemuganti GK, Duffy M, Torczynski E, Edward DP. Late orbital recurrence of a choroidal melanoma following internal resection: report of a case and review of the literature. Surv Ophthalmol 2003; 48:181-90. [PMID: 12686303 DOI: 10.1016/s0039-6257(02)00458-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The management of uveal melanomas always has been a challenge to the clinicians and has evolved from the era of eye-removing surgeries to elimination of the tumor by the body's own immune system through vaccines. Evaluating the outcome of each strategy improves our understanding of the disease process and helps us to improvise on the existing modalities of treatment. Internal resection of choroidal melanomas has been described as one of the treatment modalities for this malignant tumor. Tumor recurrences reported following this surgical procedure have been uncommon. We report a rare case of a 61-year-old Caucasian woman who presented with a 2-month history of progressive, painful proptosis in her right eye. Thirteen years ago she underwent internal resection of an intraocular choroidal melanoma in the same eye and had no evidence of metastatic disease. Magnetic resonance imaging demonstrated a mass occupying the superotemporal portion of the right orbit adjacent to the globe and behind the area of prior internal resection. Biopsy of the lesion and the subsequent A right orbital exenteration confirmed the diagnosis of recurrent malignant melanoma. Twenty-four months following exenteration the patient continues to be free of metastatic disease. Since internal surgical resection was described in 1984, this is the latest known recurrence of a posterior choroidal melanoma. In this review, we highlight the clinical features of this rare case and discuss in brief the rationale of various treatment modalities for choroidal melanoma.
Collapse
Affiliation(s)
- Nicholas Mittica
- Department of Ophthalmology, University of Illinois College of Medicine at Chicago, 1855 West Taylor Street (M/C 648), Chicago, IL 60062, USA
| | | | | | | | | |
Collapse
|
17
|
Mulcahy KA, Alexander S, Platts KE, Wardle C, Sisley K, Rennie IG, Murray AK. CD80-mediated induction of immunostimulation in two ocular melanoma cell lines is augmented by interferon-gamma. Melanoma Res 2002; 12:129-38. [PMID: 11930109 DOI: 10.1097/00008390-200204000-00005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although the transfection of the T-cell costimulatory molecule CD80 cDNA into human tumours can augment their immunogenicity in vitro, its expression alone is ineffective in many tumour systems. We evaluated the influence of CD80 expression on the immunostimulatory activity of ocular melanoma cell lines and determined whether IFN-gamma could enhance the effect. Two ocular melanoma cell lines were transfected with CD80 cDNA. The immunostimulatory capacity of the CD80+ transfectants was determined by their ability to stimulate the proliferation of allogeneic peripheral blood mononuclear cells (PBMC). The influence of additional accessory molecules on PBMC proliferation was assessed by pre-treating the CD80 transfectants with IFN-gamma. The CD80+ transfectants induced proliferation of allogeneic PBMC. IFN-gamma treatment of the tumour cells induced upregulated expression of MHC class I, de novo expression of MHC class II and CD54, and enhanced the ability of the CD80+ transfectants to stimulate PBMC proliferation. CD4+ T cells were not required for the proliferative response against untreated CD80+ tumour cells but were necessary for the augmentation of proliferation observed following IFN-gamma treatment. CD80+ ocular melanoma cells possess immunostimulatory potential which is augmented by IFN-gamma induced upregulation of cell surface molecules. Further studies on the role of costimulatory molecules in inducing anti-tumour immunity in ocular melanoma may help to define new strategies for application of immunotherapeutic approaches to treat this aggressive disease.
Collapse
Affiliation(s)
- K A Mulcahy
- Section of Oncology and Pathology (Cancer Studies), University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | | | | | | | | | | | | |
Collapse
|
18
|
Hurks HM, Metzelaar-Blok JA, Mulder A, Claas FH, Jager MJ. High frequency of allele-specific down-regulation of HLA class I expression in uveal melanoma cell lines. Int J Cancer 2000; 85:697-702. [PMID: 10699951 DOI: 10.1002/(sici)1097-0215(20000301)85:5<697::aid-ijc16>3.0.co;2-h] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Uveal melanoma is the most common primary intra-ocular tumor in adults and has a high mortality rate due to liver metastases, for which no effective treatment is available. To investigate whether immunotherapy might be feasible in uveal melanoma, the HLA class I surface expression of 6 uveal melanoma cell lines was analyzed by flow cytometry using a broad panel of allele-specific monoclonal antibodies. To up-regulate HLA expression, cells were also cultured with IFN-alpha or -gamma. In general, expression of HLA-A alleles was high (except for cell line EOM-3) and could be further up-regulated by both IFN-alpha and -gamma. In cell line EOM-3, IFN-gamma treatment resulted in significant HLA-A expression while IFN-alpha treatment did not. Expression of HLA-B alleles was low or even negative. Variable effects were observed after IFN treatment. In 3 cell lines, expression of some HLA-B alleles could not be induced by IFN-alpha or -gamma: HLA-B44 in cell line 92-1, HLA-B15 in cell line OCM-1 and HLA-B5 in cell line MEL-202. The other B alleles of these cell lines showed enhanced expression levels upon IFN stimulation. In OMM-1 cells, IFN-alpha and -gamma increased the expression of HLA-A but did not induce expression of the 2 B alleles, indicating an HLA-B locus-specific loss. We thus found a high frequency of allele-specific and locus-specific down-regulation of HLA expression in uveal melanoma cell lines. Some of these defects were not restored by IFN-alpha or -gamma treatment. The lack of HLA expression may explain why uveal melanoma cells escape immune surveillance by cytotoxic T cells and complicate the development of immunotherapy in uveal melanoma.
Collapse
Affiliation(s)
- H M Hurks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
19
|
de Vries TJ, Trancikova D, Ruiter DJ, van Muijen GN. High expression of immunotherapy candidate proteins gp100, MART-1, tyrosinase and TRP-1 in uveal melanoma. Br J Cancer 1998; 78:1156-61. [PMID: 9820172 PMCID: PMC2063001 DOI: 10.1038/bjc.1998.646] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In the treatment of cutaneous melanoma, provisional therapeutic strategies have been designed to combat tumour load using T cells that are sensitized with peptides derived from melanoma autoantigens, such as glycoprotein 100 (gp100), melanoma antigen recognized by T cells 1 (MART-1 or MelanA), tyrosinase and tyrosinase-related protein 1 (TRP-1). We recently found that gp100, MART-1 and tyrosinase are heterogeneously expressed in human cutaneous melanoma (De Vries et al (1997) Cancer Res 57: 3223-3229). Here, we extended our investigations on expression of these immunotherapy candidate proteins to uveal melanoma lesions. Cryostat sections from 11 spindle-type, 21 mixed and epithelioid tumours and four metastasis lesions were stained with antibodies specifically recognizing gp100, MART-1, tyrosinase and TRP-1. In addition, we used the DOPA reaction to detect tyrosinase enzyme activity as a confirmation of the tyrosinase immunohistochemical results. High expression of gp100, MART-1 and tyrosinase was found in the uveal melanoma lesions: 80% of the lesions displayed 75-100% positive tumour cells. TRP-1 positivity was slightly less: approximately 65% of the lesions stained in the 75-100% positive tumour cell category. All uveal melanoma lesions were positive for the four markers studied, this being in contrast to cutaneous melanoma where 17% of the advanced primary lesions and metastases were negative. The presence of these antigens was a little lower in metastases. We conclude that uveal melanomas and their metastases express melanocyte-lineage immunotherapy candidate proteins very abundantly. Uveal melanomas differ in this respect from cutaneous melanoma, in which the expression of these immunotherapy antigens was much more heterogeneous. This makes uveal melanoma a suitable candidate tumour for immunotherapeutic approaches.
Collapse
Affiliation(s)
- T J de Vries
- Department of Pathology, University Hospital, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
20
|
Abstract
Intraocular tumours reside within an organ that provides sanctuary from many immunological defence mechanisms. Antigens displayed on many intraocular tumours can elicit an aberrant systemic immune response in which systemic antigen-specific delayed-type hypersensitivity (DTH) is actively downregulated, thus denying the host one potential effector mechanism for controlling its intraocular tumour. Constituents within the aqueous humour inhibit the expression of DTH and natural killer cell effector mechanisms within the eye and thus protect intraocular tumours from immune-mediated rejection. Some experimental intraocular tumours in mice express potent tumour-specific antigens that stimulate the expansion of tumour-specific robust cytotoxic T lymphocyte (CTL) populations which enter the eye and mediate tumour rejection by piecemeal tumour necrosis. However, the presence of tumour-infiltrating lymphocytes (TIL) within an intraocular tumour does not inevitably lead to tumour resolution. In some cases CTL precursors infiltrate the intraocular tumour but fail to differentiate into mature cytolytic effector cells. Although uveal melanomas express melanoma-specific and melanoma-associated antigens that are capable of eliciting both humoral and cellular immunity, formidable barriers prevent the expression of tumour immunity. These barriers include: (a) anterior chamber-associated immune deviation; (b) in situ suppression of DTH effector cells; (c) suppression of natural killer cell activity in oculi; and (d) inactivation of the complement cascade by regulatory proteins expressed on uveal melanoma cells.
Collapse
Affiliation(s)
- J Y Niederkorn
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, USA
| |
Collapse
|
21
|
Hishii M, Andrews D, Boyle LA, Wong JT, Pandolfi F, van den Elsen PJ, Kurnick JT. In vivo accumulation of the same anti-melanoma T cell clone in two different metastatic sites. Proc Natl Acad Sci U S A 1997; 94:1378-83. [PMID: 9037061 PMCID: PMC19799 DOI: 10.1073/pnas.94.4.1378] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In a patient with progressing metastatic melanoma, we showed that the same autologous tumor-cytolytic CD8+ tumor infiltrating lymphocyte (TIL) clone accumulated in two separate metastatic sites. This clone, which represented three of eight independently derived clones from a tumor deposit on the skin of the abdomen, also represented two of eight clones derived from a skin lesion on the shoulder. This clone could be identified by its use of a unique TCRBV2-nD1n-J1S6 sequence, and could also be detected by single-stranded conformational polymorphism (SSCP) as the dominant TCRBV2-expressing clone among CD8+ TILs propagated from both shoulder and abdominal lesions. Using SSCP analysis, we also demonstrated that this clone was dominant in the fresh tumor tissue and in all TILs in which CD8+ were strongly represented, including several separate but parallel cultures. The SSCP pattern for this clone was not apparent among CD4+ TILs or CD8+ peripheral blood mononuclear cells. The SSCP analysis of the tumor tissue prior to in vitro culture is an indication that the selection for this anti-tumor cytotoxic T cell clone was a reflection of its in vivo accumulation. Thus, we provide evidence that melanomas are immunogenic and able to select for cytotoxic antitumor-specific TIL clones that are expanded in vivo and can circulate to accumulate in different tumor sites. However, because these clones were isolated from progressing tumor metastases, the accumulation of these specific cytotoxic T cells was not sufficient to contain tumor growth.
Collapse
Affiliation(s)
- M Hishii
- Pathology Research Laboratory, Massachusetts General Hospital, Charlestown 02129, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Fisk B, Flytzanis CN, Pollack MS, Wharton JT, Ioannides CG, Flytzanes CN. Characterization of T-cell receptor V beta repertoire in ovarian tumour-reacting CD3+ CD8+ CD4- CTL lines. Scand J Immunol 1994; 40:591-600. [PMID: 7848491 DOI: 10.1111/j.1365-3083.1994.tb03510.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
T cells from tumour infiltrating lymphocytes (TIL) cultured in media containing IL-2 were shown to mediate in vitro and in vivo antitumor responses. To characterize the T-cell antigen receptor (TCR) V beta expression in autologous cytotoxic effectors we isolated CD3+ CD8+ CD4- cells from cultures of TIL and tumour-associated lymphocytes (TAL) and analysed the TCR V beta repertoire of CD3+ CD8+ CD4- lines of known HLA-A, -B and -C phenotype, using polymerase chain reaction (PCR). These lines showed preferential lysis of autologous tumours and lysed, to a much lesser extent, NK and LAK cell-sensitive targets. Tumour lysis was inhibited by antibodies to CD3 and MHC class I antigens indicating that they are cytotoxic T lymphocytes (CTL). These CD8+ CTL lines expressed a broad distribution of TCR V beta repertoire which was dominated by particular groups of V beta families in each CTL line. However, no predominant expression of one or the same V beta segment in all CTL lines was observed although statistical correlations between V beta family usage and magnitude of the antitumour cytolytic response were found. These results suggest that certain TCR V beta families may be selected by antigen in ovarian tumour-reactive T cells and this selection may be affected by Ag expression, and/or host factors. To our knowledge, this is the first documentation of TCR V beta repertoire of human ovarian tumour-reactive CD3+ CD8+ CD4- CTL from different individuals of known HLA types.
Collapse
MESH Headings
- Autoantigens/immunology
- Blotting, Southern
- Cytotoxicity Tests, Immunologic
- Female
- Flow Cytometry
- HLA Antigens/immunology
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Ovarian Neoplasms/immunology
- Polymerase Chain Reaction
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- B Fisk
- Department of Gynecologic Oncology, University of Texas M.D. Anderson Cancer Center, Houston
| | | | | | | | | | | |
Collapse
|
24
|
Huang XQ, Mitchell MS, Liggett PE, Murphree AL, Kan-Mitchell J. Non-fastidious, melanoma-specific CD8+ cytotoxic T lymphocytes from choroidal melanoma patients. Cancer Immunol Immunother 1994; 38:399-405. [PMID: 8205561 PMCID: PMC11038977 DOI: 10.1007/bf01517210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/1993] [Accepted: 02/15/1994] [Indexed: 01/29/2023]
Abstract
To characterize the anti-melanoma reactivity of CD8+ cytotoxic T lymphocytes (CTL) from choroidal melanoma patients, CTL clones were isolated from the peripheral blood of three patients after mixed lymphocyte/tumor cell culture (MLTC). Clones were derived from lymphocytes stimulated by allogeneic (OCM-1, A24, A28) or autologous (OCM-3, A1, A30) melanoma cells. Their reactivity against a panel of HLA-typed melanoma and nonmelanoma cells was assessed, to determine whether a single CTL clone could recognize and lyse a variety of allogeneic melanoma cell lines. While proportionately more clones derived from autologous MLTC were melanoma-specific than allogeneic MLTC (42% versus 14%), melanoma-specific CTL were recovered from both. Notably, a novel melanoma specificity was identified. These CTL clones were termed non-fastidious because they were capable of lysing melanoma cells with which they had no HLA class I alleles in common. Nonetheless, lysis was mediated by the HLA class I molecule. Since lysis was specific for melanoma cells, these CTL appeared to recognize a shared melanoma peptide(s). Because of their prevalence, we propose that non-fastidious CTL are integral to human anti-melanoma T cell immunity. This reinforces clinical findings that allogeneic melanomas can substitute for autologous tumors in active specific immunotherapy. By circumventing the need for autologous melanoma, it is possible to treat patients after removal of the primary choroidal melanoma in an attempt to prevent metastasis.
Collapse
Affiliation(s)
- X Q Huang
- Department of Pathology, University of Southern California School of Medicine, Los Angeles 90033
| | | | | | | | | |
Collapse
|
25
|
LeMay LG, Kan-Mitchell J, Goedegebuure P, Harel W, Mitchell MS. Detection of melanoma-reactive CD4+ HLA-class I-restricted cytotoxic T cell clones with long-term assay and pretreatment of targets with interferon-gamma. Cancer Immunol Immunother 1993; 37:187-94. [PMID: 8101473 PMCID: PMC11038543 DOI: 10.1007/bf01525434] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/1992] [Accepted: 02/05/1993] [Indexed: 01/28/2023]
Abstract
Twenty-five CD4+ cytotoxic T lymphocyte (CTL) clones were obtained from the peripheral blood or tumor tissues of melanoma patients undergoing active specific immunotherapy. Melanoma-reactive T cells were cloned by limiting dilution using either autologous or allogeneic melanoma cells to stimulate their proliferation. Sixteen of the clones reacted against autologous melanoma cells but not against the autologous lymphoblastoid cell line, which we defined as "melanoma-specific." Optimal demonstration of the lytic activity of CD4+ CTL required a 16-h incubation period and an effector:target cell ratio of 40:1. In addition, a 24-h pre-incubation of the target melanoma cells with 100 U interferon (IFN) gamma consistently augmented lysis by these CD4+ CTL, increasing it from a mean level of 20% to one of 52%. Lysis by 8 of the 11 melanoma-reactive CD4+ T cell clones was exclusively HLA-class-I-restricted, as judged by blocking with monoclonal antibodies (mAb). Five of these HLA class-I-restricted clones were reactive only with the autologous melanoma cells, while the other 3 clones were also reactive with allogeneic melanoma cells. In all cases, the T cells and melanoma targets shared at least one HLA class I allele, usually HLA-A2, HLA-C3 or HLA-B62. Interestingly, lysis by 2 of the 11 clones was inhibited by both anti-HLA-class-I or -HLA-class-II mAb, while lysis by 1 other clone was inhibited by neither. HLA class I molecules and several accessory molecules were maximally expressed by the melanoma target cells, both in terms of distribution and copy number before IFN gamma treatment. Thus, IFN gamma may have acted by increasing the expression of melanoma-associated epitopes as presented by HLA class I (or HLA class II) molecules. A proportion of human CD4+ CTL appeared to recognize melanoma-associated epitopes presented by the HLA class I molecule, although their lytic potency may be less than that of their CD8+ counterparts.
Collapse
Affiliation(s)
- L G LeMay
- Department of Medicine, University of Southern California, Los Angeles 90033
| | | | | | | | | |
Collapse
|
26
|
Kan-Mitchell J, Huang XQ, Steinman L, Oksenberg JR, Harel W, Parker JW, Goedegebuure PS, Darrow TL, Mitchell MS. Clonal analysis of in vivo activated CD8+ cytotoxic T lymphocytes from a melanoma patient responsive to active specific immunotherapy. Cancer Immunol Immunother 1993; 37:15-25. [PMID: 8513449 PMCID: PMC11038976 DOI: 10.1007/bf01516937] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/1992] [Accepted: 11/17/1992] [Indexed: 01/31/2023]
Abstract
To study in vivo activated cytolytic T cells, CD8+ T cells clones were isolated from a melanoma patient (HLA A2, A11) treated with active specific immunotherapy for 5 years. CD8+ T lymphocytes, purified by fluorescence-activated cell sorting, were cloned directly from the peripheral blood without antigen-presenting cells in the presence of irradiated autologous melanoma cells and recombinant interleukin-2 (IL-2) and IL-4. These conditions were inhibitory to de novo in vitro immunization. Of the 28 cytolytic CD8+ T cell clones, 21 lysed the autologous melanoma cell line (M7) but not the autologous lymphoblastoid cell line (LCL-7) nor the two melanoma cell line, M1 (HLA A28) and M2 (HLA A28, A31), used to immunize the patient. The remaining 7 clones were also melanoma-specific, although their reactivities were broader, lysing several melanoma cell lines but not HLA-matched lymphoblastoid cells. Eight clones from the first group, ostensibly self-MHC-restricted, were expanded for further analysis. All expressed cluster determinants characteristic of mature, activated T cells, but not those of thymocytes, naive T cells, B cells or natural killer (NK) cells. They also expressed CD13, a myeloid marker. Of the 8 clones, 3 expressed both CD4 and CD8, but dual expression was not correlated with specificity of lysis. Two CD8+ and 2 CD4+ CD8+ clones were specific for the autologous melanoma cells, the other 4 were also reactive against other HLA-A2-positive melanomas. Cytotoxicity for both singly and doubly positive clones was restricted by HLA class I but not class II antigens. Analysis of the RNA expression of the T cell receptor (TCR) V alpha and V beta gene segments revealed heterogeneous usage by the A2-restricted clones and, perhaps, also by the broadly melanoma-specific clones. Apparent TCR-restricted usage was noted for the self-MHC-restricted clones; 2 of the 4 expressed the V alpha 17/V beta 7 dimer. Since the T cell clones were derived from separate precursors of circulating cytotoxic T lymphocytes (CTL), the V alpha 17/V beta 7 TCR was well represented in the peripheral blood lymphocytes of this patient. In summary, we show that melanoma cells presented their own antigens to stimulate the proliferation of melanoma-reactive CD8+ CTL. CTL with a range of melanoma specificities and different TCR alpha beta dimers were encountered in this patient, perhaps as a result of hyperimmunization.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
MESH Headings
- Adult
- Antigens, CD/analysis
- Base Sequence
- CD8 Antigens/immunology
- Cell Separation
- Clone Cells
- Cytotoxicity, Immunologic
- DNA, Neoplasm/analysis
- DNA, Neoplasm/chemistry
- Female
- Flow Cytometry
- Histocompatibility Antigens Class I/analysis
- Histocompatibility Antigens Class II/analysis
- Humans
- Immunophenotyping
- Immunotherapy, Active
- Melanoma/immunology
- Melanoma/therapy
- Molecular Sequence Data
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Vaccines, Synthetic/therapeutic use
Collapse
Affiliation(s)
- J Kan-Mitchell
- Department of Pathology, University of Southern California School of Medicine, Los Angeles 90033
| | | | | | | | | | | | | | | | | |
Collapse
|