101
|
Lymph-directed immunotherapy - Harnessing endogenous lymphatic distribution pathways for enhanced therapeutic outcomes in cancer. Adv Drug Deliv Rev 2020; 160:115-135. [PMID: 33039497 DOI: 10.1016/j.addr.2020.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/07/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
The advent of immunotherapy has revolutionised the treatment of some cancers. Harnessing the immune system to improve tumour cell killing is now standard clinical practice and immunotherapy is the first line of defence for many cancers that historically, were difficult to treat. A unifying concept in cancer immunotherapy is the activation of the immune system to mount an attack on malignant cells, allowing the body to recognise, and in some cases, eliminate cancer. However, in spite of a significant proportion of patients that respond well to treatment, there remains a subset who are non-responders and a number of cancers that cannot be treated with these therapies. These limitations highlight the need for targeted delivery of immunomodulators to both tumours and the effector cells of the immune system, the latter being highly concentrated in the lymphatic system. In this context, macromolecular therapies may provide a significant advantage. Macromolecules are too large to easily access blood capillaries and instead typically exhibit preferential uptake via the lymphatic system. In contexts where immune cells are the therapeutic target, particularly in cancer therapy, this may be advantageous. In this review, we examine in brief the current immunotherapy approaches in cancer and how macromolecular and nanomedicine strategies may improve the therapeutic profiles of these drugs. We subsequently discuss how therapeutics directed either by parenteral or mucosal administration, can be taken up by the lymphatics thereby accessing a larger proportion of the body's immune cells. Finally, we detail drug delivery strategies that have been successfully employed to target the lymphatics.
Collapse
|
102
|
McAvoy MB, Choi BD, Jones PS. Immune Therapy for Central Nervous System Metastasis. Neurosurg Clin N Am 2020; 31:627-639. [PMID: 32921357 DOI: 10.1016/j.nec.2020.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Brain metastases lead to substantial morbidity and mortality among patients with advanced malignancies. Although treatment options have traditionally included largely palliative measures, studies of brain metastasis response to immunotherapy are promising. Immune checkpoint inhibitors have shown efficacy in studies of patients with melanoma, renal cell carcinoma, and lung cancer brain metastases. Patients with brain metastases are more frequently included in clinical trials, ushering in a new era in immunotherapy and management for patients with brain metastases. Gaining an understanding of the molecular determination for response to immunotherapies remains a major challenge and is an active area of future research.
Collapse
Affiliation(s)
- Malia B McAvoy
- University of Washington Medical Center, Department of Neurological Surgery, Box 356470, 1959 NE Pacific Street, Seattle, WA 98195-6470, USA
| | - Bryan D Choi
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WAC 3, Boston, MA 02114, USA
| | - Pamela S Jones
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 15 Parkman Street, WAC 745, Boston, MA 02114, USA.
| |
Collapse
|
103
|
Pierini S, Tanyi JL, Simpkins F, George E, Uribe-Herranz M, Drapkin R, Burger R, Morgan MA, Facciabene A. Ovarian granulosa cell tumor characterization identifies FOXL2 as an immunotherapeutic target. JCI Insight 2020; 5:136773. [PMID: 32814714 PMCID: PMC7455139 DOI: 10.1172/jci.insight.136773] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Granulosa cell tumors (GCT) are rare ovarian malignancies. Due to the lack of effective treatment in late relapse, there is a clear unmet need for novel therapies. Forkhead Box L2 (FOXL2) is a protein mainly expressed in granulosa cells (GC) and therefore is a rational therapeutic target. Since we identified tumor infiltrating lymphocytes (TILs) as the main immune population within GCT, TILs from 11 GCT patients were expanded, and their phenotypes were interrogated to determine that T cells acquired late antigen-experienced phenotypes and lower levels of PD1 expression. Importantly, TILs maintained their functionality after ex vivo expansion as they vigorously reacted against autologous tumors (100% of patients) and against FOXL2 peptides (57.1% of patients). To validate the relevance of FOXL2 as a target for immune therapy, we developed a plasmid DNA vaccine (FoxL2–tetanus toxin; FoxL2-TT) by fusing Foxl2 cDNA with the immune-enhancing domain of TT. Mice immunization with FoxL2-TT controlled growth of FOXL2-expressing ovarian (BR5) and breast (4T1) cancers in a T cell–mediated manner. Combination of anti–PD-L1 with FoxL2-TT vaccination further reduced tumor progression and improved mouse survival without affecting the female reproductive system and pregnancy. Together, our results suggest that FOXL2 immune targeting can produce substantial long-term clinical benefits. Our study can serve as a foundation for trials testing immunotherapeutic approaches in patients with ovarian GCT. FOXL2 may serve as a immunotherapeutic target for tumor infiltrating lymphocytes in ovarian granulosa cell tumors.
Collapse
Affiliation(s)
- Stefano Pierini
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Janos L Tanyi
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fiona Simpkins
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erin George
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mireia Uribe-Herranz
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronny Drapkin
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert Burger
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark A Morgan
- Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrea Facciabene
- Department of Radiation Oncology and.,Ovarian Cancer Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
104
|
Li W, Qiu S, Chen J, Jiang S, Chen W, Jiang J, Wang F, Si W, Shu Y, Wei P, Fan G, Tian R, Wu H, Xu C, Wang H. Chimeric Antigen Receptor Designed to Prevent Ubiquitination and Downregulation Showed Durable Antitumor Efficacy. Immunity 2020; 53:456-470.e6. [PMID: 32758419 DOI: 10.1016/j.immuni.2020.07.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/21/2020] [Accepted: 07/15/2020] [Indexed: 01/01/2023]
Abstract
Clinical evidence suggests that poor persistence of chimeric antigen receptor-T cells (CAR-T) in patients limits therapeutic efficacy. Here, we designed a CAR with recyclable capability to promote in vivo persistence and to sustain antitumor activity. We showed that the engagement of tumor antigens induced rapid ubiquitination of CARs, causing CAR downmodulation followed by lysosomal degradation. Blocking CAR ubiquitination by mutating all lysines in the CAR cytoplasmic domain (CARKR) markedly repressed CAR downmodulation by inhibiting lysosomal degradation while enhancing recycling of internalized CARs back to the cell surface. Upon encountering tumor antigens, CARKR-T cells ameliorated the loss of surface CARs, which promoted their long-term killing capacity. Moreover, CARKR-T cells containing 4-1BB signaling domains displayed elevated endosomal 4-1BB signaling that enhanced oxidative phosphorylation and promoted memory T cell differentiation, leading to superior persistence in vivo. Collectively, our study provides a straightforward strategy to optimize CAR-T antitumor efficacy by redirecting CAR trafficking.
Collapse
Affiliation(s)
- Wentao Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shizhen Qiu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jian Chen
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Shutan Jiang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wendong Chen
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jingwei Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wen Si
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yilai Shu
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Ping Wei
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ruijun Tian
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Haitao Wu
- ENT institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China.
| | - Chenqi Xu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
105
|
Dual Targeting of Mesothelin and CD19 with Chimeric Antigen Receptor-Modified T Cells in Patients with Metastatic Pancreatic Cancer. Mol Ther 2020; 28:2367-2378. [PMID: 32730744 DOI: 10.1016/j.ymthe.2020.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/12/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
B cells infiltrate pancreatic ductal adenocarcinoma (PDAC) and in preclinical cancer models, can suppress T cell immunosurveillance in cancer. Here, we conducted a pilot study to assess the safety and feasibility of administering lentiviral-transduced chimeric antigen receptor (CAR)-modified autologous T cells redirected against mesothelin to target tumor cells along with CART cells redirected against CD19 to deplete B cells. Both CARs contained 4-1BB and CD3ζ signaling domains. Three patients with chemotherapy-refractory PDAC received 1.5 g/m2 cyclophosphamide prior to separate infusions of lentiviral-transduced T cells engineered to express chimeric anti-mesothelin immunoreceptor SS1 (CART-Meso, 3 × 107/m2) and chimeric anti-CD19 immunoreceptor (CART-19, 3 × 107/m2). Treatment was well tolerated without dose-limiting toxicities. Best response was stable disease (1 of 3 patients). CART-19 (compared to CART-Meso) cells showed the greatest expansion in the blood, although persistence was transient. B cells were successfully depleted in all subjects, became undetectable by 7-10 days post-infusion, and remained undetectable for at least 28 days. Together, concomitant delivery of CART-Meso and CART-19 cells in patients with PDAC is safe. CART-19 cells deplete normal B cells but at the dose tested in these 3 subjects did not improve CART-Meso cell persistence.
Collapse
|
106
|
Melaiu O, Lucarini V, Giovannoni R, Fruci D, Gemignani F. News on immune checkpoint inhibitors as immunotherapy strategies in adult and pediatric solid tumors. Semin Cancer Biol 2020; 79:18-43. [PMID: 32659257 DOI: 10.1016/j.semcancer.2020.07.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/19/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have shown unprecedented benefits in various adult cancers, and this success has prompted the exploration of ICI therapy even in childhood malignances. Although the use of ICIs as individual agents has achieved disappointing response rates, combinational therapies are likely to promise better results. However, only a subset of patients experienced prolonged clinical effects, thus suggesting the need to identify robust bio-markers that predict individual clinical response or resistance to ICI therapy as the main challenge. In this review, we focus on how the use of ICIs in adult cancers can be translated into pediatric malignances. We discuss the physiological mechanism of action of each IC, including PD-1, PD-L1 and CTLA-4 and the new emerging ones, LAG-3, TIM-3, TIGIT, B7-H3, BTLA and IDO-1, and evaluate their prognostic value in both adult and childhood tumors. Furthermore, we offer an overview of preclinical models and clinical trials currently under investigation to improve the effectiveness of cancer immunotherapies in these patients. Finally, we outline the main predictive factors that influence the efficacy of ICIs, in order to lay the basis for the development of a pan-cancer immunogenomic model, able to direct young patients towards more specific immunotherapy.
Collapse
Affiliation(s)
- Ombretta Melaiu
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Valeria Lucarini
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Doriana Fruci
- Paediatric Haematology/Oncology Department, Ospedale Pediatrico Bambino Gesù, Rome, Italy.
| | | |
Collapse
|
107
|
Abd Hamid M, Peng Y, Dong T. Human cancer germline antigen-specific cytotoxic T cell-what can we learn from patient. Cell Mol Immunol 2020; 17:684-692. [PMID: 32451453 PMCID: PMC7331575 DOI: 10.1038/s41423-020-0468-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
In this review, we will highlight the importance of cancer germline antigen-specific cytotoxic CD8+ T lymphocytes (CTL) and the factors affecting antitumor CTL responses. In light of cancer immunotherapy, we will emphasis the need to further understand the features, characteristics, and actions of modulatory receptors of human cancer germline-specific CTLs, in order to determine the optimal conditions for antitumor CTL responses.
Collapse
Affiliation(s)
- Megat Abd Hamid
- Nufield Department of Medicine, Chinese Academy of Medical Science Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Yanchun Peng
- Nufield Department of Medicine, Chinese Academy of Medical Science Oxford Institute (COI), University of Oxford, Oxford, UK
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Tao Dong
- Nufield Department of Medicine, Chinese Academy of Medical Science Oxford Institute (COI), University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
108
|
Bianchi V, Harari A, Coukos G. Neoantigen-Specific Adoptive Cell Therapies for Cancer: Making T-Cell Products More Personal. Front Immunol 2020; 11:1215. [PMID: 32695101 PMCID: PMC7333784 DOI: 10.3389/fimmu.2020.01215] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Mutation-derived neoantigens are taking central stage as a determinant in eliciting effective antitumor immune responses following adoptive T-cell therapies. These mutations are patient-specific, and their targeting calls for highly personalized pipelines. The promising clinical outcomes of tumor-infiltrating lymphocyte (TIL) therapy have spurred interest in generating T-cell infusion products that have been selectively enriched in neoantigen (or autologous tumor) reactivity. The implementation of an isolation step, prior to T-cell in vitro expansion and reinfusion, may provide a way to improve the overall response rates achieved to date by adoptive T-cell therapies in metastatic cancer patients. Here we provide an overview of the main technologies [i.e., peptide major histocompatibility complex (pMHC) multimers, cytokine capture, and activation markers] to enrich infiltrating or circulating T-cells in predefined neoantigen specificities (or tumor reactivity). The unique technical and regulatory challenges faced by such highly specialized and patient-specific manufacturing T-cell platforms are also discussed.
Collapse
Affiliation(s)
- Valentina Bianchi
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Alexandre Harari
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.,Center of Experimental Therapeutics, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital, Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
109
|
Knochelmann HM, Dwyer CJ, Smith AS, Bowers JS, Wyatt MM, Nelson MH, Rangel Rivera GO, Horton JD, Krieg C, Armeson K, Lesinski GB, Rubinstein MP, Li Z, Paulos CM. IL6 Fuels Durable Memory for Th17 Cell-Mediated Responses to Tumors. Cancer Res 2020; 80:3920-3932. [PMID: 32561531 DOI: 10.1158/0008-5472.can-19-3685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/04/2020] [Accepted: 06/11/2020] [Indexed: 12/15/2022]
Abstract
The accessibility of adoptive T-cell transfer therapies (ACT) is hindered by the cost and time required for product development. Here we describe a streamlined ACT protocol using Th17 cells expanded only 4 days ex vivo. While shortening expansion compromised cell yield, this method licensed Th17 cells to eradicate large tumors to a greater extent than cells expanded longer term. Day 4 Th17 cells engrafted, induced release of multiple cytokines including IL6, IL17, MCP-1, and GM-CSF in the tumor-bearing host, and persisted as memory cells. IL6 was a critical component for efficacy of these therapies via its promotion of long-term immunity and resistance to tumor relapse. Mechanistically, IL6 diminished engraftment of FoxP3+ donor T cells, corresponding with robust tumor infiltration by donor effector over regulatory cells for the Day 4 Th17 cell product relative to cell products expanded longer durations ex vivo. Collectively, this work describes a method to rapidly generate therapeutic T-cell products for ACT and implicates IL6 in promoting durable immunity of Th17 cells against large, established solid tumors. SIGNIFICANCE: An abbreviated, 4-day ex vivo expansion method licenses Th17 cells to confer long-lived immunity against solid malignancies via induction of systemic IL6 in the host.See related commentary by Fiering and Ho, p. 3795.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina. .,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Connor J Dwyer
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Aubrey S Smith
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Jacob S Bowers
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Megan M Wyatt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Michelle H Nelson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Guillermo O Rangel Rivera
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Joshua D Horton
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Carsten Krieg
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Kent Armeson
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Mark P Rubinstein
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina.,Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, Ohio
| | - Chrystal M Paulos
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, South Carolina. .,Department of Dermatology & Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
110
|
Boos LA, Leslie I, Larkin J. Metastatic melanoma: therapeutic agents in preclinical and early clinical development. Expert Opin Investig Drugs 2020; 29:739-753. [PMID: 32401070 DOI: 10.1080/13543784.2020.1769066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Advanced melanoma historically had a very poor outcome but targeted therapies and immune checkpoint inhibitors (IC) have changed the course of the disease and made durable responses possible. However, most patients will develop progressive disease, so further strategies to overcome treatment resistance are needed. Areas covered: Current treatment strategies and landmark trials are discussed. Novel targeted agents, immune checkpoint inhibitors, and further immune-modulatory drugs, cancer vaccines and tumor infiltrating lymphocytes and their potential role in the treatment of melanoma are described. Current trials investigating these emerging agents and treatment strategies were searched for on ClinicalTrials.gov and are presented on the background of the current literature explaining the rationale for employing these new agents and strategies. Combinations of tumor-directed agents with those causing immune augmentation as well as a new adjuvant and neoadjuvant strategies are discussed. Expert opinion: Questions regarding treatment combination, personalization, and sequence of treatment will become increasingly important and will be guided by new biomarkers. New treatment settings will broaden the patient selection and will highlight the need for further discussions regarding toxicity in long-term survivorship.
Collapse
Affiliation(s)
- Laura Amanda Boos
- Department of Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust , London, UK
| | - Isla Leslie
- Department of Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust , London, UK
| | - James Larkin
- Department of Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust , London, UK
| |
Collapse
|
111
|
Zeng H, Huang Y, Chen L, Li H, Ma X. Exploration and validation of the effects of robust co-expressed immune-related genes on immune infiltration patterns and prognosis in laryngeal cancer. Int Immunopharmacol 2020; 85:106622. [PMID: 32485354 DOI: 10.1016/j.intimp.2020.106622] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Laryngeal cancer is a common malignant tumor that originates from the larynx, yet its molecular mechanisms have not been thoroughly explored. The purpose of this study was to identify and evaluate immune-related genes in laryngeal cancer through gene co-expression networks, which may serve as biomarkers for its immunotherapy. METHODS We applied ESTIMATE to evaluate the immune-infiltration landscape of tumor microenvironment. The co-expression networks were constructed by weighted gene co expression network analysis (WGCNA) and compared with the existing human immune related genes (IRGs) to determine the co-expressed IRGs. GSVA combined with CIBERSORT and ssGSEA illustrated the correlation of hub genes and immune infiltration patterns. TIDE algorithm and Subclass mapping evaluated the function of hub genes in predicting immune function and immunotherapeutic sensitivity. The pRRophetic was employed in the sensitivity prediction of chemotherapeutic drugs. RESULTS A total of 23 co-expressed IRGs were identified and showed robust expression characteristics. These genes were significantly related to immune infiltration patterns, immune function and sensitivity prediction of immunotherapy and chemotherapeutic drugs for laryngeal cancer patients. Genetic alteration in somatic mutation level and related pathways were also revealed. CONCLUSION The 23 co-expressed IRGs may act as immunotherapeutic biomarkers and potential therapeutic targets for laryngeal cancer with certain expression robustness. The molecular mechanisms deserve further investigation, which will guide clinical treatment in the future.
Collapse
Affiliation(s)
- Hao Zeng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yeqian Huang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, China; West China School of Medicine, West China Hospital, Sichuan University, China
| | - Linyan Chen
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hui Li
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, China; West China School of Medicine, West China Hospital, Sichuan University, China
| | - Xuelei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| |
Collapse
|
112
|
Dwyer CJ, Arhontoulis DC, Rangel Rivera GO, Knochelmann HM, Smith AS, Wyatt MM, Rubinstein MP, Atkinson C, Thaxton JE, Neskey DM, Paulos CM. Ex vivo blockade of PI3K gamma or delta signaling enhances the antitumor potency of adoptively transferred CD8 + T cells. Eur J Immunol 2020; 50:1386-1399. [PMID: 32383488 PMCID: PMC7496332 DOI: 10.1002/eji.201948455] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/13/2020] [Indexed: 01/05/2023]
Abstract
Adoptive T cell transfer therapy induces objective responses in patients with advanced malignancies. Despite these results, some individuals do not respond due to the generation of terminally differentiated T cells during the expansion protocol. As the gamma and delta catalytic subunits in the PI3K pathway are abundant in leukocytes and involved in cell activation, we posited that blocking both subunits ex vivo with the inhibitor IPI‐145 would prevent their differentiation, thereby increasing antitumor activity in vivo. However, IPI‐145 treatment generated a product with reduced antitumor activity. Instead, T cells inhibited of PI3Kγ (IPI‐549) or PI3Kδ (CAL‐101 or TGR‐1202) alone were more potent in vivo. While T cells coinhibited of PI3Kγ and PI3Kδ were less differentiated, they were functionally impaired, indicated by reduced production of effector cytokines after antigenic re‐encounter and decreased persistence in vivo. Human CAR T cells expanded with either a PI3Kγ or PI3Kδ inhibitor possessed a central memory phenotype compared to vehicle cohorts. We also found that PI3Kδ‐inhibited CARs lysed human tumors in vitro more effectively than PI3Kγ‐expanded or traditionally expanded CAR T cells. Our data imply that sole blockade of PI3Kγ or PI3Kδ generates T cells with remarkable antitumor properties, a discovery that has substantial clinical implications.
Collapse
Affiliation(s)
- Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Dimitrios C Arhontoulis
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Guillermo O Rangel Rivera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Carl Atkinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Surgery, Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica E Thaxton
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Orthopedics, Medical University of South Carolina, Charleston, SC, USA
| | - David M Neskey
- Department of Otolaryngology, Head and Neck Surgery, Medical University of South Carolina, Charleston, SC, USA.,Department of Cell and Molecular Pharmacology and Developmental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
113
|
Li X, Daniyan AF, Lopez AV, Purdon TJ, Brentjens RJ. Cytokine IL-36γ improves CAR T-cell functionality and induces endogenous antitumor response. Leukemia 2020; 35:506-521. [PMID: 32447345 PMCID: PMC7680719 DOI: 10.1038/s41375-020-0874-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/28/2020] [Accepted: 05/13/2020] [Indexed: 01/04/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has shown remarkable responses in B cell malignancies. However, many patients suffer from limited response and tumor relapse due to lack of persisting CAR T cells and immune escape. These clinical challenges have compromised the long-term efficacy of CAR T cell therapy and call for the development of novel CAR designs. We demonstrated that CAR T cells secreting a cytokine interleukin-36γ (IL-36γ) showed significantly improved CAR T cell expansion and persistence, and resulted in superior tumor eradication compared to conventional CAR T cells. The enhanced cellular function by IL-36γ was mediated through an autocrine manner. In addition, activation of endogenous antigen-presenting cells (APCs) and T cells by IL-36γ aided the formation of a secondary anti-tumor response which delayed the progression of antigen-negative tumor challenge. Together, our data provide preclinical evidence to support the translation of this design for an improved CAR T cell–mediated anti-tumor response.
Collapse
Affiliation(s)
- Xinghuo Li
- Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, USA.,Department of Medicine, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anthony F Daniyan
- Department of Medicine, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Andrea V Lopez
- Department of Medicine, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Terence J Purdon
- Department of Medicine, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Renier J Brentjens
- Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, USA. .,Department of Medicine, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
114
|
Roncati L, Palmieri B. Adoptive cell transfer (ACT) of autologous tumor-infiltrating lymphocytes (TILs) to treat malignant melanoma: the dawn of a chimeric antigen receptor T (CAR-T) cell therapy from autologous donor. Int J Dermatol 2020; 59:763-769. [PMID: 32441324 DOI: 10.1111/ijd.14945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/02/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) are B, T-helper, and T-cytotoxic lymphocytes migrated from the blood or lymph stream toward a tumor with the aim to infiltrate and destroy it. They can be histologically graded as brisk, nonbrisk, or absent. Malignant melanoma has been the first malignancy found to be correlated with TILs status, being brisk TILs associated with better clinical outcomes. By the terminology of "adoptive cell transfer" (ACT), the medical oncology refers to the transfer of cells in a tumor-bearing patient from the same recipient or a healthy donor. METHODS A PubMed literature search on the topic has been performed. Additional documents known to the authors and identified from the reference list of cited publications have been included. RESULTS In the past, autologous TILs ACT was successfully tested for the treatment of malignant melanoma and, today, it is a standardized procedure in several centers around the world. It represents the first research step toward the bioengineered chimeric antigen receptor T (CAR-T) cell therapy from autologous donor. CONCLUSIONS Both autologous TILs ACT and CAR-T cell therapy from autologous donor exploit the anticancer power of targeted self-lymphocytes, but CAR-T cell technology also virtually allows treatment of those melanomas devoid of TILs or with so few cytotoxic TILs that are difficult to identify.
Collapse
Affiliation(s)
- Luca Roncati
- Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
| | - Beniamino Palmieri
- Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
| |
Collapse
|
115
|
Bagheri S, Safaie Qamsari E, Yousefi M, Riazi-Rad F, Sharifzadeh Z. Targeting the 4-1BB costimulatory molecule through single chain antibodies promotes the human T-cell response. Cell Mol Biol Lett 2020; 25:28. [PMID: 32336974 PMCID: PMC7178758 DOI: 10.1186/s11658-020-00219-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 03/27/2020] [Indexed: 11/12/2022] Open
Abstract
Background Adoptive T-cell therapy (ACT) using autologous tumor-reactive T lymphocytes has considerable potential for cancer immunotherapy. In ACT, T cells are isolated from cancer patients and then stimulated and expanded in vitro by cytokines and costimulatory molecules. 4-1BB is an important costimulatory protein belonging to the TNF receptor superfamily. It is involved in T-cell survival, proliferation and activation. Agonistic anti-4-1BB monoclonal antibodies have been introduced as appropriate tools for ACT. Methods Here, various single-chain fragment variable (scFv) antibodies were used to activate T cells isolated from peripheral blood via immune magnetic isolation. The T cells were stimulated with IL-2 and anti-CD-3 mAb and then treated with agonistic anti-4-1BB scFvs. The results showed the remarkable effects of anti-41BB scFvs on the functional properties of T cells, including their activation, proliferation and cytokine production. The flow cytometry analysis revealed a considerable increase in the expression of the T-cell activation marker CD69. Moreover, T-cell proliferation was evidenced in treated cells by CFSE labeling compared to the control groups. Result Anti-4-1BB scFvs significantly increased IFN-γ and IL-2 mRNA and protein expression in T cells, but exhibited no stimulatory effect on IL-4 expression. These findings show that anti-4-1BB scFvs could evoke a Type I immune response. Conclusions Our results demonstrate that targeting the 4-1BB molecule using agonistic scFvs could be an effective strategy for T-cell stimulation as part of an ACT approach to cancer treatment.
Collapse
Affiliation(s)
- Salman Bagheri
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.,2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,3Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Safaie Qamsari
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.,2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,3Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- 2Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Riazi-Rad
- 1Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
116
|
Poschke IC, Hassel JC, Rodriguez-Ehrenfried A, Lindner KAM, Heras-Murillo I, Appel LM, Lehmann J, Lövgren T, Wickström SL, Lauenstein C, Roth J, König AK, Haanen JBAG, van den Berg J, Kiessling R, Bergmann F, Flossdorf M, Strobel O, Offringa R. The Outcome of Ex Vivo TIL Expansion Is Highly Influenced by Spatial Heterogeneity of the Tumor T-Cell Repertoire and Differences in Intrinsic In Vitro Growth Capacity between T-Cell Clones. Clin Cancer Res 2020; 26:4289-4301. [PMID: 32303540 DOI: 10.1158/1078-0432.ccr-19-3845] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 03/15/2020] [Accepted: 04/14/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE During our efforts to develop tumor-infiltrating lymphocyte (TIL) therapy to counter the devastating recurrence rate in patients with primary resectable pancreatic ductal adenocarcinoma (PDA), we found that PDA TILs can readily be expanded in vitro and that the majority of resulting TIL cultures show reactivity against the autologous tumor. However, the fraction of tumor-reactive T cells is low. We investigated to which extent this was related to the in vitro expansion. EXPERIMENTAL DESIGN We compared the clonal composition of TIL preparations before and after in vitro expansion using T-cell receptor (TCR) deep sequencing. Our findings for PDA were benchmarked to experiments with melanoma TILs. RESULTS We found that the TIL TCR repertoire changes dramatically during in vitro expansion, leading to loss of tumor- dominant T-cell clones and overgrowth by newly emerging T-cell clones that are barely detectable in the tumor. These changes are primarily driven by differences in the intrinsic in vitro expansion capacity of T-cell clones. Single-cell experiments showed an association between poor proliferative capacity and expression of markers related to antigen experience and dysfunction. Furthermore, we found that spatial heterogeneity of the TIL repertoire resulted in TCR repertoires that are greatly divergent between TIL cultures derived from distant tumor samples of the same patient. CONCLUSIONS Culture-induced changes in clonal composition are likely to affect tumor reactivity of TIL preparations. TCR deep sequencing provides important insights into the factors that govern the outcome of in vitro TIL expansion and thereby a path toward optimization of the production of TIL preparations with high therapeutic efficacy.See related commentary by Lozano-Rabella and Gros, p. 4177.
Collapse
Affiliation(s)
- Isabel C Poschke
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany. .,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Jessica C Hassel
- Department of Dermatology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Aaron Rodriguez-Ehrenfried
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Katharina A M Lindner
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Ignacio Heras-Murillo
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Lena M Appel
- Division of Theoretical Systems Biology, German Cancer Research Center and BioQuant Center, University of Heidelberg, Heidelberg, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Johanna Lehmann
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Tanja Lövgren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stina L Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Claudia Lauenstein
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Jasmin Roth
- Department of Dermatology and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Anna-Katharina König
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - John B A G Haanen
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joost van den Berg
- Department of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Frank Bergmann
- Department of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Flossdorf
- Division of Theoretical Systems Biology, German Cancer Research Center and BioQuant Center, University of Heidelberg, Heidelberg, Germany.,Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Strobel
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
117
|
Han S, Chung DC, St Paul M, Liu ZQ, Garcia-Batres C, Elford AR, Tran CW, Chapatte L, Ohashi PS. Overproduction of IL-2 by Cbl-b deficient CD4 + T cells provides resistance against regulatory T cells. Oncoimmunology 2020; 9:1737368. [PMID: 32313719 PMCID: PMC7153846 DOI: 10.1080/2162402x.2020.1737368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells are integral to the regulation of autoimmune and anti-tumor immune responses. However, several studies have suggested that changes in T cell signaling networks can result in T cells that are resistant to the suppressive effects of regulatory T cells. Here, we investigated the role of Cbl-b, an E3 ubiquitin ligase, in establishing resistance to Treg-mediated suppression. We found that the absence of Cbl-b, a negative regulator of multiple TCR signaling pathways, rendered T cells impartial to Treg suppression by regulating cytokine networks leading to improved anti-tumor immunity despite the presence of Treg cells in the tumor. Specifically, Cbl-b KO CD4+FoxP3− T cells hyper-produced IL-2 and together with IL-2 Rα upregulation served as an essential mechanism to escape suppression by Treg cells. Furthermore, we report that IL-2 serves as the central molecule required for cytokine-induced Treg resistance. Collectively our data emphasize the role of IL-2 as a key mechanism that renders CD4+ T cells resistant to the inhibitory effects of Treg cells.
Collapse
Affiliation(s)
- SeongJun Han
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Douglas C Chung
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael St Paul
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhe Qi Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Carlos Garcia-Batres
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Alisha R Elford
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Charles W Tran
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Laurence Chapatte
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
118
|
Cytotoxic T Lymphocytes Regenerated from iPS Cells Have Therapeutic Efficacy in a Patient-Derived Xenograft Solid Tumor Model. iScience 2020; 23:100998. [PMID: 32259478 PMCID: PMC7188741 DOI: 10.1016/j.isci.2020.100998] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/20/2020] [Accepted: 03/17/2020] [Indexed: 12/27/2022] Open
Abstract
Current adoptive T cell therapies conducted in an autologous setting are costly, time consuming, and depend on the quality of the patient's T cells. To address these issues, we developed a strategy in which cytotoxic T lymphocytes (CTLs) are regenerated from iPSCs that were originally derived from T cells and succeeded in regenerating CTLs specific for the WT1 antigen, which exhibited therapeutic efficacy in a xenograft model of leukemia. In this study, we extended our strategy to solid tumors. The regenerated WT1-specific CTLs had a strong therapeutic effect in orthotopic xenograft model using a renal cell carcinoma (RCC) cell line. To make our method more generally applicable, we developed an allogeneic approach by transducing HLA-haplotype homozygous iPSCs with WT1-specific TCR α/β genes that had been tested clinically. The regenerated CTLs antigen-specifically suppressed tumor growth in a patient-derived xenograft model of RCC, demonstrating the feasibility of our strategy against solid tumors. Patient-derived xenograft of renal cell carcinoma was used in a cell-therapy model Cytotoxic T lymphocytes (CTLs) that target WT1-antigen were used as effector cells CTLs produced from iPSCs transduced with WT1-TCR genes showed efficacy in the model The present results demonstrate the feasibility of our strategy against solid tumors
Collapse
|
119
|
Glitza IC, Goff SL, Ross M, Margolin K. And Now for Something Completely Different: Immunotherapy Beyond Checkpoints in Melanoma. Am Soc Clin Oncol Educ Book 2020; 40:1-12. [PMID: 32243202 DOI: 10.1200/edbk_79437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Advances in the understanding of biology and therapy of melanoma have occurred at an astonishing pace over the past approximately 15 years, and successful melanoma therapy has led the way for similar advances in many other solid tumors that are continuing to improve outcomes for all patients with cancer. Although the 2018 Nobel Prize was awarded to two investigators who discovered that therapeutic targeting of immune checkpoints held the key to major patient benefits, there are many additional immunotherapeutic strategies that warrant further study and discussion at scientific and medical meetings. This article provides the newest information on three areas of immunotherapy that have been successfully applied to melanoma and continue to pave the way for new developments: cytokines, adoptive cell therapies (ADTs), and intratumoral injection of immunomodulatory agents.
Collapse
Affiliation(s)
- Isabella Claudia Glitza
- Department of Melanoma Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Stephanie L Goff
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Merrick Ross
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Kim Margolin
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
120
|
Brightman SE, Naradikian MS, Miller AM, Schoenberger SP. Harnessing neoantigen specific CD4 T cells for cancer immunotherapy. J Leukoc Biol 2020; 107:625-633. [PMID: 32170883 PMCID: PMC7793607 DOI: 10.1002/jlb.5ri0220-603rr] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
The goal of precision immunotherapy is to direct a patient's T cell response against the immunogenic mutations expressed on their tumors. Most immunotherapy approaches to-date have focused on MHC class I-restricted peptide epitopes by which cytotoxic CD8+ T lymphocytes (CTL) can directly recognize tumor cells. This strategy largely overlooks the critical role of MHC class II-restricted CD4+ T cells as both positive regulators of CTL and other effector cell types, and as direct effectors of antitumor immunity. In this review, we will discuss the role of neoantigen specific CD4+ T cells in cancer immunotherapy and how existing treatment modalities may be leveraged to engage this important T cell subset.
Collapse
Affiliation(s)
- Spencer E. Brightman
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Martin S. Naradikian
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Aaron M. Miller
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037
| | | |
Collapse
|
121
|
Lloyd-Williams H, Hughes DA. A systematic review of economic evaluations of advanced therapy medicinal products. Br J Clin Pharmacol 2020; 87:2428-2443. [PMID: 32154598 PMCID: PMC8247439 DOI: 10.1111/bcp.14275] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/06/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022] Open
Abstract
Aims Advanced therapy medicinal products (ATMPs) represent a new category of medicinal products with a potential for transformative improvements in health outcomes but at exceptionally high prices. Routine adoption of ATMPs requires robust evidence of their cost‐effectiveness. Methods A systematic literature review of economic evaluations of ATMPs, including gene therapies, somatic cell therapies and tissue‐engineered products, was conducted. Literature was searched using MedLine, Embase, PubMed, Cochrane Register, the NHS Economic Evaluation Database and the grey literature of health technology assessment organisations with search terms relating to ATMPs and economic evaluations. Titles were screened independently by 2 reviewers. Articles deemed to meet the inclusion criteria were screened independently on abstract, and full texts reviewed. Study findings were appraised critically. Results 4514 articles were identified, of which 23 met the inclusion criteria. There was some evidence supporting the cost‐effectiveness of: chimeric antigen receptor T‐cell therapy axicabtagene–ciloleucel (Yescarta), embryonic neural stem cells, tumour infiltrating lymphocytes, in vitro expanded myoblast, autologous chondrocyte implantation, ex vivo gene therapy (Strimvelis) and voretigene neparvovec (Luxturna). However, estimates of cost‐effectiveness were associated with significant uncertainty and high likelihood of bias, resulting from largely unknown long‐term outcomes, a paucity of evidence on health state utilities and extensive modelling assumptions. Conclusion There are critical limitations to the economic evidence for ATMPs, most notably in relation to evidence on the durability of treatment effect, and the reliability of opinion‐based assumptions necessary when evidence is absent.
Collapse
Affiliation(s)
- Huw Lloyd-Williams
- Centre for Health Economics and Medicines Evaluation, Bangor University, Wales, UK
| | - Dyfrig A Hughes
- Centre for Health Economics and Medicines Evaluation, Bangor University, Wales, UK
| |
Collapse
|
122
|
Chruściel E, Urban-Wójciuk Z, Arcimowicz Ł, Kurkowiak M, Kowalski J, Gliwiński M, Marjański T, Rzyman W, Biernat W, Dziadziuszko R, Montesano C, Bernardini R, Marek-Trzonkowska N. Adoptive Cell Therapy-Harnessing Antigen-Specific T Cells to Target Solid Tumours. Cancers (Basel) 2020; 12:E683. [PMID: 32183246 PMCID: PMC7140076 DOI: 10.3390/cancers12030683] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/26/2022] Open
Abstract
In recent years, much research has been focused on the field of adoptive cell therapies (ACT) that use native or genetically modified T cells as therapeutic tools. Immunotherapy with T cells expressing chimeric antigen receptors (CARs) demonstrated great success in the treatment of haematologic malignancies, whereas adoptive transfer of autologous tumour infiltrating lymphocytes (TILs) proved to be highly effective in metastatic melanoma. These encouraging results initiated many studies where ACT was tested as a treatment for various solid tumours. In this review, we provide an overview of the challenges of T cell-based immunotherapies of solid tumours. We describe alternative approaches for choosing the most efficient T cells for cancer treatment in terms of their tumour-specificity and phenotype. Finally, we present strategies for improvement of anti-tumour potential of T cells, including combination therapies.
Collapse
Affiliation(s)
- Elżbieta Chruściel
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Zuzanna Urban-Wójciuk
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
| | - Jacek Kowalski
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
- Department of Pathomorphology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Mateusz Gliwiński
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Tomasz Marjański
- Department of Thoracic Surgery, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (T.M.); (W.R.)
| | - Witold Rzyman
- Department of Thoracic Surgery, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (T.M.); (W.R.)
| | - Wojciech Biernat
- Department of Pathomorphology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Rafał Dziadziuszko
- Department of Oncology and Radiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Carla Montesano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy;
| | - Roberta Bernardini
- Department of Biology and Interdepartmental Center CIMETA, University of Rome "Tor Vergata", 00133 Rome, Italy;
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science (ICCVS), University of Gdańsk, 80-309 Gdańsk, Poland; (E.C.); (Z.U.-W.); (M.K.); (J.K.)
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
123
|
CTLA-4 blockade boosts the expansion of tumor-reactive CD8 + tumor-infiltrating lymphocytes in ovarian cancer. Sci Rep 2020; 10:3914. [PMID: 32127601 PMCID: PMC7054305 DOI: 10.1038/s41598-020-60738-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Adoptive cell therapy (ACT) with autologous tumor-infiltrating lymphocytes (TILs) can induce durable complete tumor regression in patients with advanced melanoma. Efforts are currently underway to expand this treatment modality to other cancer types. In the microenvironment of ovarian cancer, the engagement of co-inhibitory immune checkpoint molecules such as CTLA-4 can lead to the inactivation of TILs. Thus, approaches that directly manipulate co-inhibitory pathways within the tumor microenvironment might improve the expansion of tumor-reactive TILs. The initial expansion of TILs for ACT from tumor fragments provides a window of opportunity to manipulate an intact tumor microenvironment and improve CD8+ T-cell output and TIL tumor reactivity. To exploit this, we used a CTLA-4-blocking antibody, added during the initial TIL culture, and found that the blockade of CTLA-4 favored the propagation of CD8+ TILs from ovarian tumor fragments. Interestingly, adding the CTLA-4 blocking antibody in the initial phase of the TIL culture resulted in more potent anti-tumor TILs in comparison to standard TIL cultures. This phenotype was preserved during the rapid expansion phase. Thus, targeting CTLA-4 within the intact tumor microenvironment of tumor fragments enriches tumor-reactive TILs and may improve clinical outcome of TIL-based ACT in ovarian cancer.
Collapse
|
124
|
Chatani PD, Yang JC. Mutated RAS: Targeting the "Untargetable" with T Cells. Clin Cancer Res 2020; 26:537-544. [PMID: 31511296 PMCID: PMC7002282 DOI: 10.1158/1078-0432.ccr-19-2138] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/14/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022]
Abstract
The RAS family of proteins is at the apex of several pathways implicated in a multitude of epithelial cancers but has remained stubbornly resistant to the wave of targeted small molecules and antibodies that have revolutionized clinical oncology. KRAS, the most commonly mutated of the isoforms, represents an attractive target for treatment, given its ubiquity, central role as a driver mutation, and association with poor prognosis. This review is a comprehensive summary of the existing approaches to targeting KRAS spanning small-molecule inhibitors, cancer vaccines, and with a focus on trials in adoptive cell therapy. Here we explain how the limitations of existing drugs and nonspecific immune-based therapies are circumvented with techniques in modern immunotherapy. The successes outlined represent the most promising path to finally targeting the prototypical "undruggable" RAS oncogene family.
Collapse
Affiliation(s)
| | - James C Yang
- Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland
| |
Collapse
|
125
|
Wang J, Shen F, Yao Y, Wang LL, Zhu Y, Hu J. Adoptive Cell Therapy: A Novel and Potential Immunotherapy for Glioblastoma. Front Oncol 2020; 10:59. [PMID: 32083009 PMCID: PMC7005203 DOI: 10.3389/fonc.2020.00059] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults with very poor prognosis and few advances in its treatment. Recently, fast-growing cancer immunotherapy provides a glimmer of hope for GBM treatment. Adoptive cell therapy (ACT) aims at infusing immune cells with direct anti-tumor activity, including tumor-infiltrating lymphocyte (TIL) transfer and genetically engineered T cells transfer. For example, complete regressions in patients with melanoma and refractory lymphoma have been shown by using naturally tumor-reactive T cells and genetically engineered T cells expressing the chimeric anti-CD19 receptor, respectively. Recently, the administration of ACT showed therapeutic potentials for GBM treatment as well. In this review, we summarize the success of ACT in the treatment of cancer and provide approaches to overcome some challenges of ACT to allow its adoption for GBM treatment.
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Shen
- Department of Orthopaedic Surgery's Spine Division, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Ying Yao
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin-Lin Wang
- Department of Basic Medicine Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongjian Zhu
- Department of Neurosurgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Hu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
126
|
Considine B, Hurwitz ME. Key Factors in Clinical Protocols for Adoptive Cell Therapy in Melanoma. Methods Mol Biol 2020; 2097:309-327. [PMID: 31776935 DOI: 10.1007/978-1-0716-0203-4_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Adoptive cell therapy (ACT) with autologous tumor infiltrating lymphocytes (TIL) has been studied for patients with advanced metastatic cancers (primarily melanoma) for decades and has changed significantly during that period. Treatment with TIL includes ex vivo cell activation and expansion followed by re-infusion of these cells into the patient. After cell infusion, patients receive Interleukin-2 (IL-2). Objective response rates up to 52% have been seen in patients with metastatic melanoma. Efforts to improve TIL therapy include better selection and expansion of tumor-reactive lymphocytes, optimization of IL-2 or other T cell activating cytokine dosing, and, potentially, genetic manipulation of the immune cell product. Here we describe methods involved in the collection, expansion, and treatment with TIL for patients with metastatic melanoma.
Collapse
|
127
|
Abstract
Tumor development is characterized by the accumulation of mutational and epigenetic changes that transform normal cells and survival pathways into self-sustaining cells capable of untrammeled growth. Although multiple modalities including surgery, radiation, and chemotherapy are available for the treatment of cancer, the benefits conferred are often limited. The immune system is capable of specific, durable, and adaptable responses. However, cancers hijack immune mechanisms such as negative regulatory checkpoints that have evolved to limit inflammatory and immune responses to thwart effective antitumor immunity. The development of monoclonal antibodies against inhibitory receptors expressed by immune cells has produced durable responses in a broad array of advanced malignancies and heralded a new dawn in the cancer armamentarium. However, these remarkable responses are limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Preclinical and clinical studies with immune checkpoint blockade are exploring the therapeutic potential antibody-based therapy targeting multiple inhibitory receptors. In this chapter, we discuss the current understanding of the structure, ligand specificities, function, and signaling activities of various inhibitory receptors. Additionally, we discuss the current development status of various immune checkpoint inhibitors targeting these negative immune receptors and highlight conceptual gaps in knowledge.
Collapse
|
128
|
Hoo WPY, Siak PY, In LLA. Overview of Current Immunotherapies Targeting Mutated KRAS Cancers. Curr Top Med Chem 2019; 19:2158-2175. [PMID: 31483231 DOI: 10.2174/1568026619666190904163524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 02/07/2023]
Abstract
The occurrence of somatic substitution mutations of the KRAS proto-oncogene is highly prevalent in certain cancer types, which often leads to constant activation of proliferative pathways and subsequent neoplastic transformation. It is often seen as a gateway mutation in carcinogenesis and has been commonly deemed as a predictive biomarker for poor prognosis and relapse when conventional chemotherapeutics are employed. Additionally, its mutational status also renders EGFR targeted therapies ineffective owing to its downstream location. Efforts to discover new approaches targeting this menacing culprit have been ongoing for years without much success, and with incidences of KRAS positive cancer patients being on the rise, researchers are now turning towards immunotherapies as the way forward. In this scoping review, recent immunotherapeutic developments and advances in both preclinical and clinical studies targeting K-ras directly or indirectly via its downstream signal transduction machinery will be discussed. Additionally, some of the challenges and limitations of various K-ras targeting immunotherapeutic approaches such as vaccines, adoptive T cell therapies, and checkpoint inhibitors against KRAS positive cancers will be deliberated.
Collapse
Affiliation(s)
- Winfrey Pui Yee Hoo
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Pui Yan Siak
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Lionel L A In
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
129
|
Dafni U, Michielin O, Lluesma SM, Tsourti Z, Polydoropoulou V, Karlis D, Besser MJ, Haanen J, Svane IM, Ohashi PS, Kammula US, Orcurto A, Zimmermann S, Trueb L, Klebanoff CA, Lotze MT, Kandalaft LE, Coukos G. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol 2019; 30:1902-1913. [PMID: 31566658 DOI: 10.1093/annonc/mdz398] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) has been tested in advanced melanoma patients at various centers. We conducted a systematic review and meta-analysis to assess its efficacy on previously treated advanced metastatic cutaneous melanoma. The PubMed electronic database was searched from inception to 17 December 2018 to identify studies administering TIL-ACT and recombinant interleukin-2 (IL-2) following non-myeloablative chemotherapy in previously treated metastatic melanoma patients. Objective response rate (ORR) was the primary end point. Secondary end points were complete response rate (CRR), overall survival (OS), duration of response (DOR) and toxicity. Pooled estimates were derived from fixed or random effect models, depending on the amount of heterogeneity detected. Analysis was carried out separately for high dose (HD) and low dose (LD) IL-2. Sensitivity analyses were carried out. Among 1211 records screened, 13 studies (published 1988 - 2016) were eligible for meta-analysis. Among 410 heavily pretreated patients (some with brain metastasis), 332 received HD-IL-2 and 78 LD-IL-2. The pooled overall ORR estimate was 41% [95% confidence interval (CI) 35% to 48%], and the overall CRR was 12% (95% CI 7% to 16%). For the HD-IL-2 group, the ORR was 43% (95% CI 36% to 50%), while for the LD-IL-2 it was 35% (95% CI 25% to 45%). Corresponding pooled estimates for CRR were 14% (95% CI 7% to 20%) and 7% (95% CI 1% to 12%). The majority of HD-IL-2 complete responders (27/28) remained in remission during the extent of follow-up after CR (median 40 months). Sensitivity analyses yielded similar results. Higher number of infused cells was associated with a favorable response. The ORR for HD-IL-2 compared favorably with the nivolumab/ipilimumab combination following anti-PD-1 failure. TIL-ACT therapy, especially when combined with HD-IL-2, achieves durable clinical benefit and warrants further investigation. We discuss the current position of TIL-ACT in the therapy of advanced melanoma, particularly in the era of immune checkpoint blockade therapy, and review future opportunities for improvement of this approach.
Collapse
Affiliation(s)
- U Dafni
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| | - O Michielin
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - S Martin Lluesma
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Z Tsourti
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - V Polydoropoulou
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - D Karlis
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - M J Besser
- Ella Institute for the Treatment and Research of Melanoma and Skin Cancer, Sheba Medical Center, Tel Aviv; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I-M Svane
- Department of Hematology and Oncology, Center for Cancer Immune Therapy, Herlev Hospital, Herlev, Denmark
| | - P S Ohashi
- Department of Immunology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - U S Kammula
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh
| | - A Orcurto
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - S Zimmermann
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - L Trueb
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - C A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York; Parker Institute for Cancer Immunotherapy, New York; Weill Cornell Medical College, New York
| | - M T Lotze
- Department of Immunology, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, USA
| | - L E Kandalaft
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - G Coukos
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
130
|
Comoli P, Chabannon C, Koehl U, Lanza F, Urbano-Ispizua A, Hudecek M, Ruggeri A, Secondino S, Bonini C, Pedrazzoli P. Development of adaptive immune effector therapies in solid tumors. Ann Oncol 2019; 30:1740-1750. [PMID: 31435646 DOI: 10.1093/annonc/mdz285] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
State-of-the-art treatment strategies have drastically ameliorated the outcome of patients affected by cancer. However, resistant and recurrent solid tumors are generally nonresponsive to conventional therapies. A central factor in the sequence of events that lead to cancer is an alteration in antitumor immune surveillance, which results in failure to recognize and eliminate the transformed tumor cell. A greater understanding of the dysregulation and evasion of the immune system in the evolution and progression of cancer provides the basis for improved therapies. Targeted strategies, such as T-cell therapy, not only generally spare normal tissues, but also use alternative antineoplastic mechanisms that synergize with other therapeutics. Despite encouraging success in hematologic malignancies, adaptive cellular therapies for solid tumors face unique challenges because of the immunosuppressive tumor microenvironment, and the hurdle of T-cell trafficking within scarcely accessible tumor sites. This review provides a brief overview of current cellular therapeutic strategies for solid tumors, research carried out to increase efficacy and safety, and results from ongoing clinical trials.
Collapse
Affiliation(s)
- P Comoli
- Cell Factory and Pediatric Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - C Chabannon
- Institut Paoli-Calmettes, Aix-Marseille University, INSERM CBT 1409, Centre for Clinical Investigation in Biotherapy, Marseille, France
| | - U Koehl
- Institute of Clinical Immunology, University of Leipzig and Fraunhofer Institute for Cell Therapy and Immunology, Leipzig; Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - F Lanza
- Hematology and Stem Cell Transplant, Romagna Transplant Network, Ravenna, Italy
| | - A Urbano-Ispizua
- Department of Hematology, IDIBAPS, Institute of Research Josep Carreras, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - M Hudecek
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - A Ruggeri
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome
| | - S Secondino
- Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia
| | - C Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, University Vita-Salute San Raffaele and Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - P Pedrazzoli
- Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia.
| |
Collapse
|
131
|
Yunger S, Bar El A, Zeltzer LA, Fridman E, Raviv G, Laufer M, Schachter J, Markel G, Itzhaki O, Besser MJ. Tumor-infiltrating lymphocytes from human prostate tumors reveal anti-tumor reactivity and potential for adoptive cell therapy. Oncoimmunology 2019; 8:e1672494. [PMID: 31741775 PMCID: PMC6844325 DOI: 10.1080/2162402x.2019.1672494] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 12/11/2022] Open
Abstract
Advanced prostate cancer remains incurable and is the second leading cause of mortality in men. Immunotherapy based on the adoptive transfer of tumor-infiltrating lymphocytes (TIL) has demonstrated promising clinical results in patients with metastatic melanoma and lately also in other solid tumors. However, the ability to obtain TIL from patients with prostate cancer, considered poorly immunogenic, remains unknown. In this study, we investigate the feasibility of isolating and expanding TIL from primary prostate tumors. We collected tumor specimens from eight patients with diagnosed prostate adenocarcinoma undergoing radical prostatectomy and were able to successfully expand multiple autologous TIL cultures from all patients. Twenty-eight prostate-TIL cultures were further expanded using a standard rapid expansion procedure under Good Manufacturing Practice conditions. TIL cultures were phenotypically characterized for T cell subset composition, differentiation status and co-inhibitory/stimulatory markers such as PD-1, TIM-3, LAG-3, and CD28 and were found to have in general similarity to TIL obtained from patients with melanoma and lung carcinoma previously treated at our center. All analyzed TIL cultures were functional as determined by the capability to produce high level of IFNγ upon stimuli. Most importantly, co-culture assays of prostate-TIL with autologous tumors demonstrated anti-tumor reactivity. In conclusion, these findings demonstrate that functional and anti-tumor reactive TIL can be obtained, despite the immunosuppressive microenvironment of the cancer, thus this study supports the development of TIL therapy for prostate cancer patients.
Collapse
Affiliation(s)
- Sharon Yunger
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Assaf Bar El
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel.,Department of Urology, Sheba Medical Center, Ramat Gan, Israel
| | - Li-At Zeltzer
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Eddie Fridman
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel.,The Sackler Medical of School, Tel-Aviv University, Tel Aviv, Israel
| | - Gil Raviv
- Department of Urology, Sheba Medical Center, Ramat Gan, Israel
| | - Menachem Laufer
- Department of Urology, Sheba Medical Center, Ramat Gan, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Itzhaki
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Michal J Besser
- Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel.,Department of Clinical Microbiology and Immunology,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
132
|
Sakellariou-Thompson D, Forget MA, Hinchcliff E, Celestino J, Hwu P, Jazaeri AA, Haymaker C, Bernatchez C. Potential clinical application of tumor-infiltrating lymphocyte therapy for ovarian epithelial cancer prior or post-resistance to chemotherapy. Cancer Immunol Immunother 2019; 68:1747-1757. [PMID: 31602489 DOI: 10.1007/s00262-019-02402-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/20/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Immunotherapy has become a powerful treatment option for several solid tumor types. The presence of tumor-infiltrating lymphocytes (TIL) is correlated with better prognosis in ovarian cancer, pointing at the possibility to benefit from harnessing their anti-tumor activity. This preclinical study explores the feasibility of adoptive cell therapy (ACT) with TIL using an improved culture method. METHODS TIL from high-grade serous ovarian cancer were cultured using a combination of IL-2 with agonistic antibodies targeting 4-1BB and CD3. The cells were phenotyped using flow cytometry in the fresh tissue and after expansion. Tumor reactivity was assessed against HLA-matched ovarian cancer cell lines via IFN-γ ELISPOT. RESULTS Ovarian cancer is highly infiltrated with CD8+ TIL that are preferentially and robustly expanded with the addition of the agonistic antibodies. With a 95% success rate, the TIL are grown to ≥ 100 × 106 cells in 2-3 weeks without over differentiation. In addition, the CD8+ TIL grown with this method showed HLA-restricted tumor recognition. CONCLUSIONS These results indicate the viability of TIL ACT for refractory ovarian cancer by allowing for the large expansion of anti-tumor TIL in a short time and consistent manner.
Collapse
Affiliation(s)
- Donastas Sakellariou-Thompson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (UT MDACC), Unit 904, 7455 Fannin, Houston, TX, 77054, USA
| | - Marie-Andrée Forget
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (UT MDACC), Unit 904, 7455 Fannin, Houston, TX, 77054, USA
| | - Emily Hinchcliff
- Department of Gynecologic Oncology and Reproductive Medicine, UTMDACC, Houston, TX, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, UTMDACC, Houston, TX, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (UT MDACC), Unit 904, 7455 Fannin, Houston, TX, 77054, USA
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, UTMDACC, Houston, TX, USA
| | - Cara Haymaker
- Department of Translational Molecular Pathology, UT MDACC, Unit 2951, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA.
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (UT MDACC), Unit 904, 7455 Fannin, Houston, TX, 77054, USA. .,Department of Translational Molecular Pathology, UT MDACC, Unit 2951, 2130 W. Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
133
|
Stroncek DF, Reddy O, Highfill S, Panch SR. Advances in T-cell Immunotherapies. Hematol Oncol Clin North Am 2019; 33:825-837. [DOI: 10.1016/j.hoc.2019.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
134
|
T cell engineering for adoptive T cell therapy: safety and receptor avidity. Cancer Immunol Immunother 2019; 68:1701-1712. [PMID: 31542797 DOI: 10.1007/s00262-019-02395-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 09/10/2019] [Indexed: 12/13/2022]
Abstract
Since the first bone marrow transplantation, adoptive T cell therapy (ACT) has developed over the last 80 years to a highly efficient and specific therapy for infections and cancer. Genetic engineering of T cells with antigen-specific receptors now provides the possibility of generating highly defined and efficacious T cell products. The high sensitivity of engineered T cells towards their targets, however, also bears the risk of severe off-target toxicities. Therefore, different safety strategies for engineered T cells have been developed that enable removal of the transferred cells in case of adverse events, control of T cell activity or improvement of target selectivity. Receptor avidity is a crucial component in the balance between safety and efficacy of T cell products. In clinical trials, T cells equipped with high avidity T cell receptor (TCR)/chimeric antigen receptor (CAR) have been mostly used so far because of their faster and better response to antigen recognition. However, over-activation can trigger T cell exhaustion/death as well as side effects due to excessive cytokine production. Low avidity T cells, on the other hand, are less susceptible to over-activation and could possess better selectivity in case of tumor antigens shared with healthy tissues, but complete tumor eradication may not be guaranteed. In this review we describe how 'optimal' TCR/CAR affinity can increase the safety/efficacy balance of engineered T cells, and discuss simultaneous or sequential infusion of high and low avidity receptors as further options for efficacious but safe T cell therapy.
Collapse
|
135
|
Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L, Yang X, Sun J, Li Z, Han WD, Wang W. Genetically engineered T cells for cancer immunotherapy. Signal Transduct Target Ther 2019; 4:35. [PMID: 31637014 PMCID: PMC6799837 DOI: 10.1038/s41392-019-0070-9] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.
Collapse
Affiliation(s)
- Dan Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xue Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Wei-Lin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
- Department of Medical Oncology, Cancer Center, West China Hospital, West China Medical School, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Lin Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Jie Sun
- Department of Cell Biology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058 Zhejiang, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem cell and Immunotherapy Engineering, 310058 Zhejing, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, 200032 Shanghai, China
- CARsgen Therapeutics, 200032 Shanghai, China
| | - Wei-Dong Han
- Molecular & Immunological Department, Biotherapeutic Department, Chinese PLA General Hospital, No. 28 Fuxing Road, 100853 Beijing, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| |
Collapse
|
136
|
Harel M, Ortenberg R, Varanasi SK, Mangalhara KC, Mardamshina M, Markovits E, Baruch EN, Tripple V, Arama-Chayoth M, Greenberg E, Shenoy A, Ayasun R, Knafo N, Xu S, Anafi L, Yanovich-Arad G, Barnabas GD, Ashkenazi S, Besser MJ, Schachter J, Bosenberg M, Shadel GS, Barshack I, Kaech SM, Markel G, Geiger T. Proteomics of Melanoma Response to Immunotherapy Reveals Mitochondrial Dependence. Cell 2019; 179:236-250.e18. [PMID: 31495571 PMCID: PMC7993352 DOI: 10.1016/j.cell.2019.08.012] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/24/2019] [Accepted: 08/06/2019] [Indexed: 01/03/2023]
Abstract
Immunotherapy has revolutionized cancer treatment, yet most patients do not respond. Here, we investigated mechanisms of response by profiling the proteome of clinical samples from advanced stage melanoma patients undergoing either tumor infiltrating lymphocyte (TIL)-based or anti- programmed death 1 (PD1) immunotherapy. Using high-resolution mass spectrometry, we quantified over 10,300 proteins in total and ∼4,500 proteins across most samples in each dataset. Statistical analyses revealed higher oxidative phosphorylation and lipid metabolism in responders than in non-responders in both treatments. To elucidate the effects of the metabolic state on the immune response, we examined melanoma cells upon metabolic perturbations or CRISPR-Cas9 knockouts. These experiments indicated lipid metabolism as a regulatory mechanism that increases melanoma immunogenicity by elevating antigen presentation, thereby increasing sensitivity to T cell mediated killing both in vitro and in vivo. Altogether, our proteomic analyses revealed association between the melanoma metabolic state and the response to immunotherapy, which can be the basis for future improvement of therapeutic response.
Collapse
Affiliation(s)
- Michal Harel
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Rona Ortenberg
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Mariya Mardamshina
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Ettai Markovits
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Erez N Baruch
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Victoria Tripple
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - May Arama-Chayoth
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Eyal Greenberg
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel
| | - Anjana Shenoy
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Ruveyda Ayasun
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Naama Knafo
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Shihao Xu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Liat Anafi
- Institute of Pathology, Sheba Medical Center, Tel Hashomer 5265601, Israel
| | - Gali Yanovich-Arad
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Georgina D Barnabas
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Shira Ashkenazi
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Michal J Besser
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; The Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Marcus Bosenberg
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Comprehensive Cancer Center, New Haven, CT 06510, USA; Department of Dermatology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gerald S Shadel
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Iris Barshack
- Institute of Pathology, Sheba Medical Center, Tel Hashomer 5265601, Israel; The Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Gal Markel
- Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Tel Hashomer 5265601, Israel; Department of Clinical Immunology and Microbiology, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel.
| | - Tamar Geiger
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel-Aviv 6997801, Israel.
| |
Collapse
|
137
|
Utilizing T-cell Activation Signals 1, 2, and 3 for Tumor-infiltrating Lymphocytes (TIL) Expansion: The Advantage Over the Sole Use of Interleukin-2 in Cutaneous and Uveal Melanoma. J Immunother 2019; 41:399-405. [PMID: 29757889 DOI: 10.1097/cji.0000000000000230] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, we address one of the major critiques for tumor-infiltrating lymphocyte (TIL) therapy-the time needed for proper expansion of a suitable product. We postulated that T-cell receptor activation in the first phase of expansion combined with an agonistic stimulation of CD137/4-1BB and interleukin-2 would favor preferential expansion of CD8 TIL. Indeed, this novel 3-signal approach for optimal T-cell activation resulted in faster and more consistent expansion of CD8CD3 TIL. This new method allowed for successful expansion of TIL from cutaneous and uveal melanoma tumors in 100% of the cultures in <3 weeks. Finally, providing the 3 signals attributed to optimal T-cell activation led to expansion of TIL capable of recognizing their tumor counterpart in cutaneous and uveal melanoma. This new methodology for the initial phase of TIL expansion brings a new opportunity for translation of TIL therapy in challenging malignancies such as uveal melanoma.
Collapse
|
138
|
Ioannidou K, Randin O, Semilietof A, Maby-El Hajjami H, Baumgaertner P, Vanhecke D, Speiser DE. Low Avidity T Cells Do Not Hinder High Avidity T Cell Responses Against Melanoma. Front Immunol 2019; 10:2115. [PMID: 31555299 PMCID: PMC6742971 DOI: 10.3389/fimmu.2019.02115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/02/2022] Open
Abstract
The efficacy of T cells depends on their functional avidity, i. e., the strength of T cell interaction with cells presenting cognate antigen. The overall T cell response is composed of multiple T cell clonotypes, involving different T cell receptors and variable levels of functional avidity. Recently, it has been proposed that the presence of low avidity tumor antigen-specific CD8 T cells hinder their high avidity counterparts to protect from tumor growth. Here we analyzed human cytotoxic CD8 T cells specific for the melanoma antigen Melan-A/MART-1. We found that the presence of low avidity T cells did not result in reduced cytotoxicity of tumor cells, nor reduced cytokine production, by high avidity T cells. In vivo in NSG-HLA-A2 mice, the anti-tumor effect of high avidity T cells was similar in presence or absence of low avidity T cells. These data indicate that low avidity T cells are not hindering anti-tumor T cell responses, a finding that is reassuring because low avidity T cells are an integrated part of natural T cell responses.
Collapse
Affiliation(s)
- Kalliopi Ioannidou
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Olivier Randin
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Aikaterini Semilietof
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Hélène Maby-El Hajjami
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Petra Baumgaertner
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Dominique Vanhecke
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV), Lausanne, Switzerland
| |
Collapse
|
139
|
Ollé Hurtado M, Wolbert J, Fisher J, Flutter B, Stafford S, Barton J, Jain N, Barone G, Majani Y, Anderson J. Tumor infiltrating lymphocytes expanded from pediatric neuroblastoma display heterogeneity of phenotype and function. PLoS One 2019; 14:e0216373. [PMID: 31398192 PMCID: PMC6688820 DOI: 10.1371/journal.pone.0216373] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/18/2019] [Indexed: 01/06/2023] Open
Abstract
Adoptive transfer of ex vivo expanded tumor infiltrating lymphocytes (TILs) has led to clinical benefit in some patients with melanoma but has not demonstrated convincing efficacy in other solid cancers. Whilst the presence of TILs in many types of cancer is often associated with better clinical prognosis, their function has not been systematically evaluated across cancer types. Responses to immunological checkpoint inhibitors in a wide range of cancers, including those for which adoptive transfer of expanded TILs has not shown clinical benefit, has clearly delineated a number of tumor type associated with tumor-reactive lymphocytes capable of effecting tumor remissions. Neuroblastoma is an aggressive childhood solid cancer in which immunotherapy with GD2-directed antibodies confers a proven survival advantage through incompletely understood mechanisms. We therefore evaluated the feasibility of ex vivo expansion of TILs from freshly resected neuroblastoma tumors and the potential therapeutic utility of TIL expansions. TILs were successfully expanded from both tumor biopsies or resections. Significant numbers of NKT and γδT cells were identified alongside the mixed population of cytotoxic (CD8+) and helper (CD4+) T cells of both effector and central memory phenotypes. Isolated TILs were broadly non-reactive against autologous tumor and neuroblastoma cell lines, so enhancement of neuroblastoma killing was attained by transducing TILs with a second-generation chimeric antigen receptor (CAR) targeting GD2. CAR-TILs demonstrated antigen-specific cytotoxicity against tumor cell lines. This study is the first to show reproducible expansion of TILs from pediatric neuroblastoma, the high proportion of innate-like lymphocytes, and the feasibility to use CAR-TILs therapeutically.
Collapse
Affiliation(s)
- Marina Ollé Hurtado
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jolien Wolbert
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jonathan Fisher
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Barry Flutter
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Sian Stafford
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Jack Barton
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - Neha Jain
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
| | - Giuseppe Barone
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
| | - Yvonne Majani
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, England, United Kingdom
- Department of Oncology, Great Ormond Street Hospital, London, England, United Kingdom
- * E-mail:
| |
Collapse
|
140
|
Zhang J, Zhang Q, Chen X, Zhang N. Management of neoplastic pericardial disease. Herz 2019; 45:46-51. [PMID: 31297544 DOI: 10.1007/s00059-019-4833-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022]
Abstract
At present, there is no accurate and effective method for treating neoplastic pericardial effusion. This study analyzed the current literature on the treatment of neoplastic pericardial effusion to provide advice and guidance for clinical treatment. Surgical treatments include pericardial puncture, extension of catheter drainage, pericardial window, and surgical pericardiotomy. Each surgical procedure has a corresponding indication, and the best treatment is selected according to the patient's specific conditions. Systemic chemotherapy is effective in lymphoma and small cell lung cancer that are sensitive to chemotherapeutic drugs. Although pericardial injection of drugs is effective for pericardial tamponade and recurrent pericardial effusion, these methods can only temporarily relieve symptoms and cannot prolong the life of patients. In recent years, immunotherapy, especially adoptive immunotherapy, has achieved good results in the treatment of neoplastic pericardial effusion, thus providing a novel treatment option for neoplastic pericardial effusion.
Collapse
Affiliation(s)
- J Zhang
- Department of Cardiology, the Fourth Affiliated Hospital of Hebei Medical University, 050011, Shijiazhuang, Hebei Province, China
| | - Q Zhang
- Department of Clinical Medicine, Basic Medical College of Seven Years (2014), Hebei Medical University, 050017, Shijiazhuang, Hebei Province, China
| | - X Chen
- Department of Clinical Medicine, Basic Medical College of Seven Years (2014), Hebei Medical University, 050017, Shijiazhuang, Hebei Province, China
| | - N Zhang
- Department of Cardiology, the Fourth Affiliated Hospital of Hebei Medical University, 050011, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
141
|
Martinez O, Sosabowski J, Maher J, Papa S. New Developments in Imaging Cell-Based Therapy. J Nucl Med 2019; 60:730-735. [PMID: 30979822 PMCID: PMC6581223 DOI: 10.2967/jnumed.118.213348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/19/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy is now established as a central therapeutic pillar in hematologic oncology. Cell-based therapies, with or without genetic modification ex vivo, have reached the clinic as the standard of care in limited indications and remain the subject of intense preclinical and translational development. Expanding on this, related therapeutic approaches are in development for solid-tumor and nonmalignant indications, broadening the scope of this technology. It has long been recognized that in vivo tracking of infused cellular therapies would provide unique opportunities to optimize their efficacy and aid in the assessment and management of toxicity. Recently, we have witnessed the introduction of novel tracers for passive labeling of cell products and advances in the introduction and use of reporter genes to enable longitudinal imaging. This review highlights the key developments over the last 5 y.
Collapse
Affiliation(s)
- Olivier Martinez
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
| | - Jane Sosabowski
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, Eastbourne, United Kingdom; and
| | - Sophie Papa
- ImmunoEngineering Group, School of Cancer and Pharmaceutical Sciences, King's Health Partners Integrated Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
- Department of Medical Oncology, Guy's and St. Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
142
|
Goff SL, Rosenberg SA. BRAF Inhibition: Bridge or Boost to T-cell Therapy? Clin Cancer Res 2019; 25:2682-2684. [PMID: 30824585 DOI: 10.1158/1078-0432.ccr-19-0286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/14/2019] [Accepted: 02/26/2019] [Indexed: 01/22/2023]
Abstract
The concept of treatment-refractory disease has evolved as checkpoint modulation has changed the therapeutic landscape for patients with metastatic melanoma. Developing meaningful salvage strategies will involve the exploration of combination therapies and new immunotherapeutics, including adoptive transfer of tumor-infiltrating lymphocytes or other T-cell-based therapy.See related article by Atay et al., p. 2783.
Collapse
Affiliation(s)
- Stephanie L Goff
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
143
|
Nguyen LT, Saibil SD, Sotov V, Le MX, Khoja L, Ghazarian D, Bonilla L, Majeed H, Hogg D, Joshua AM, Crump M, Franke N, Spreafico A, Hansen A, Al-Habeeb A, Leong W, Easson A, Reedijk M, Goldstein DP, McCready D, Yasufuku K, Waddell T, Cypel M, Pierre A, Zhang B, Boross-Harmer S, Cipollone J, Nelles M, Scheid E, Fyrsta M, Lo CS, Nie J, Yam JY, Yen PH, Gray D, Motta V, Elford AR, DeLuca S, Wang L, Effendi S, Ellenchery R, Hirano N, Ohashi PS, Butler MO. Phase II clinical trial of adoptive cell therapy for patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and low-dose interleukin-2. Cancer Immunol Immunother 2019; 68:773-785. [PMID: 30747243 PMCID: PMC11028227 DOI: 10.1007/s00262-019-02307-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/17/2019] [Indexed: 02/03/2023]
Abstract
Adoptive cell therapy using autologous tumor-infiltrating lymphocytes (TIL) has shown significant clinical benefit, but is limited by toxicities due to a requirement for post-infusion interleukin-2 (IL-2), for which high dose is standard. To assess a modified TIL protocol using lower dose IL-2, we performed a single institution phase II protocol in unresectable, metastatic melanoma. The primary endpoint was response rate. Secondary endpoints were safety and assessment of immune correlates following TIL infusion. Twelve metastatic melanoma patients were treated with non-myeloablative lymphodepleting chemotherapy, TIL, and low-dose subcutaneous IL-2 (125,000 IU/kg/day, maximum 9-10 doses over 2 weeks). All but one patient had previously progressed after treatment with immune checkpoint inhibitors. No unexpected adverse events were observed, and patients received an average of 6.8 doses of IL-2. By RECIST v1.1, two patients experienced a partial response, one patient had an unconfirmed partial response, and six had stable disease. Biomarker assessment confirmed an increase in IL-15 levels following lymphodepleting chemotherapy as expected and a lack of peripheral regulatory T-cell expansion following protocol treatment. Interrogation of the TIL infusion product and monitoring of the peripheral blood following infusion suggested engraftment of TIL. In one responding patient, a population of T cells expressing a T-cell receptor Vβ chain that was dominant in the infusion product was present at a high percentage in peripheral blood more than 2 years after TIL infusion. This study shows that this protocol of low-dose IL-2 following adoptive cell transfer of TIL is feasible and clinically active. (ClinicalTrials.gov identifier NCT01883323.).
Collapse
Affiliation(s)
- Linh T Nguyen
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Samuel D Saibil
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Valentin Sotov
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Michael X Le
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Leila Khoja
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Danny Ghazarian
- Department of Laboratory Medicine, University Health Network, Toronto, Canada
| | - Luisa Bonilla
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Habeeb Majeed
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - David Hogg
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Anthony M Joshua
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
- Kinghorn Cancer Centre, St. Vincent's Hospital, Sydney, Australia
| | - Michael Crump
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Norman Franke
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Anna Spreafico
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Aaron Hansen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Ayman Al-Habeeb
- Department of Laboratory Medicine, University Health Network, Toronto, Canada
| | - Wey Leong
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Alexandra Easson
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Michael Reedijk
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - David P Goldstein
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Otolaryngology, Head and Neck Surgery, Princess Margaret Cancer Centre, Toronto, Canada
| | - David McCready
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Kazuhiro Yasufuku
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Thomas Waddell
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Marcelo Cypel
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Andrew Pierre
- Department of Surgical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Bianzheng Zhang
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Sarah Boross-Harmer
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jane Cipollone
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Megan Nelles
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Elizabeth Scheid
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Michael Fyrsta
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Charlotte S Lo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jessica Nie
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jennifer Y Yam
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Pei Hua Yen
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Diana Gray
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Vinicius Motta
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Alisha R Elford
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Stephanie DeLuca
- Department of Pharmacy, Princess Margaret Cancer Centre, Toronto, Canada
| | - Lisa Wang
- Drug Development Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Stephanie Effendi
- Drug Development Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Ragitha Ellenchery
- Drug Development Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Naoto Hirano
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Pamela S Ohashi
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Marcus O Butler
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, Canada.
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada.
- Department of Immunology, University of Toronto, Toronto, Canada.
- University Health Network, Princess Margaret Cancer Centre, 9-622, 610 University Avenue, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|
144
|
Sacchetti B, Botticelli A, Pierelli L, Nuti M, Alimandi M. CAR-T with License to Kill Solid Tumors in Search of a Winning Strategy. Int J Mol Sci 2019; 20:E1903. [PMID: 30999624 PMCID: PMC6514830 DOI: 10.3390/ijms20081903] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/11/2019] [Accepted: 04/13/2019] [Indexed: 02/06/2023] Open
Abstract
Artificial receptors designed for adoptive immune therapies need to absolve dual functions: antigen recognition and abilities to trigger the lytic machinery of reprogrammed effector T lymphocytes. In this way, CAR-T cells deliver their cytotoxic hit to cancer cells expressing targeted tumor antigens, bypassing the limitation of HLA-restricted antigen recognition. Expanding technologies have proposed a wide repertoire of soluble and cellular "immunological weapons" to kill tumor cells; they include monoclonal antibodies recognizing tumor associated antigens on tumor cells and immune cell checkpoint inhibition receptors expressed on tumor specific T cells. Moreover, a wide range of formidable chimeric antigen receptors diversely conceived to sustain quality, strength and duration of signals delivered by engineered T cells have been designed to specifically target tumor cells while minimize off-target toxicities. The latter immunological weapons have shown distinct efficacy and outstanding palmarès in curing leukemia, but limited and durable effects for solid tumors. General experience with checkpoint inhibitors and CAR-T cell immunotherapy has identified a series of variables, weaknesses and strengths, influencing the clinical outcome of the oncologic illness. These aspects will be shortly outlined with the intent of identifying the still "missing strategy" to combat epithelial cancers.
Collapse
Affiliation(s)
| | - Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Luca Pierelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marianna Nuti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| | - Maurizio Alimandi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
145
|
Han S, Toker A, Liu ZQ, Ohashi PS. Turning the Tide Against Regulatory T Cells. Front Oncol 2019; 9:279. [PMID: 31058083 PMCID: PMC6477083 DOI: 10.3389/fonc.2019.00279] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022] Open
Abstract
Regulatory T (Treg) cells play crucial roles in health and disease through their immunosuppressive properties against various immune cells. In this review we will focus on the inhibitory role of Treg cells in anti-tumor immunity. We outline how Treg cells restrict T cell function based on our understanding of T cell biology, and how we can shift the equilibrium against regulatory T cells. To date, numerous strategies have been proposed to limit the suppressive effects of Treg cells, including Treg cell neutralization, destabilizing Treg cells and rendering T cells resistant to Treg cells. Here, we focus on key mechanisms which render T cells resistant to the suppressive effects of Treg cells. Lastly, we also examine current limitations and caveats of overcoming the inhibitory activity of Treg cells, and briefly discuss the potential to target Treg cell resistance in the context of anti-tumor immunity.
Collapse
Affiliation(s)
- SeongJun Han
- Princess Margaret Cancer Centre, Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Aras Toker
- Princess Margaret Cancer Centre, Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
| | - Zhe Qi Liu
- Princess Margaret Cancer Centre, Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Pamela S. Ohashi
- Princess Margaret Cancer Centre, Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, ON, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
146
|
Abstract
Immunotherapy has revolutionized the treatment of melanoma, with implications for the surgical management of this disease. Surgeons must be aware of the impact of various immunotherapies on patients with resectable and unresectable disease, and how surgical decision-making should progress as a result. We expect that current and developing immunotherapies will increase surgeon involvement for resection of metastatic melanoma, whether for tumor harvests to generate autologous lymphocytes or for consolidating control of disease beyond what immunotherapies alone can achieve. Despite remarkable advancements in the field, significant work is needed to optimize the immuno-modulation that targets cancers while minimizing toxicity for patients.
Collapse
|
147
|
Cellular therapy approaches harnessing the power of the immune system for personalized cancer treatment. Semin Immunol 2019; 42:101306. [DOI: 10.1016/j.smim.2019.101306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
|
148
|
Monette A, Bergeron D, Ben Amor A, Meunier L, Caron C, Mes-Masson AM, Kchir N, Hamzaoui K, Jurisica I, Lapointe R. Immune-enrichment of non-small cell lung cancer baseline biopsies for multiplex profiling define prognostic immune checkpoint combinations for patient stratification. J Immunother Cancer 2019; 7:86. [PMID: 30922393 PMCID: PMC6437930 DOI: 10.1186/s40425-019-0544-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/22/2019] [Indexed: 12/21/2022] Open
Abstract
Background Permanence of front-line management of lung cancer by immunotherapies requires predictive companion diagnostics identifying immune-checkpoints at baseline, challenged by the size and heterogeneity of biopsy specimens. Methods An innovative, tumor heterogeneity reducing, immune-enriched tissue microarray was constructed from baseline biopsies, and multiplex immunofluorescence was used to profile 25 immune-checkpoints and immune-antigens. Results Multiple immune-checkpoints were ranked, correlated with antigen presenting and cytotoxic effector lymphocyte activity, and were reduced with advancing disease. Immune-checkpoint combinations on TILs were associated with a marked survival advantage. Conserved combinations validated on more than 11,000 lung, breast, gastric and ovarian cancer patients demonstrate the feasibility of pan-cancer companion diagnostics. Conclusions In this hypothesis-generating study, deepening our understanding of immune-checkpoint biology, comprehensive protein-protein interaction and pathway mapping revealed that redundant immune-checkpoint interactors associate with positive outcomes, providing new avenues for the deciphering of molecular mechanisms behind effects of immunotherapeutic agents targeting immune-checkpoints analyzed. Electronic supplementary material The online version of this article (10.1186/s40425-019-0544-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Monette
- Institut du cancer de Montréal, Montréal, Québec, Canada. .,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada. .,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| | - Derek Bergeron
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - Amira Ben Amor
- Medicine Faculty of Tunis, Department of Immunology and Histology, Tunis El Manar University, Tunis, Tunisia
| | - Liliane Meunier
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada
| | - Christine Caron
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada
| | - Anne-Marie Mes-Masson
- Institut du cancer de Montréal, Montréal, Québec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada.,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada
| | | | - Kamel Hamzaoui
- Medicine Faculty of Tunis, Department of Immunology and Histology, Tunis El Manar University, Tunis, Tunisia.,Abderrahmen Mami Hospital, Homeostasis and cell immune dysfunction Research Unit, Ariana, Tunisia
| | - Igor Jurisica
- Krembil Research Institute, UHN, 60 Leonard Avenue, Toronto, Ontario, M5T 0S8, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Réjean Lapointe
- Institut du cancer de Montréal, Montréal, Québec, Canada. .,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 rue St-Denis, Tour Viger, Room R10-432, Montréal, Québec, H2X 0A9, Canada. .,Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, Canada.
| |
Collapse
|
149
|
Chen C, Gao FH. Th17 Cells Paradoxical Roles in Melanoma and Potential Application in Immunotherapy. Front Immunol 2019; 10:187. [PMID: 30800130 PMCID: PMC6375889 DOI: 10.3389/fimmu.2019.00187] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/22/2019] [Indexed: 12/24/2022] Open
Abstract
The progressive infiltration of immune cells is associated with the progression of melanoma. Specifically, Th17 cells in melanoma microenvironment have both antitumor and protumor effects. It is now necessary to understand the contradictory data associated with how Th17 cells play a role in melanoma. This review will summarize the current knowledge regarding the potential mechanisms that may be involved in the effects of Th17 cells in melanoma progression. Currently, since adoptive transferring Th17 cells has been successful in eradicating melanoma in mice, it offers promise for next-generation adoptive cell transfer, as ex vivo expanded stemness-like memory Th17 cells which are induced by distinct cytokines or pharmacologic reagents may be infused into melanoma patients to potentiate treatment outcome.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
150
|
Moran A, Azghadi S, Maverakis EM, Christensen S, Dyer BA. Combined Immune Checkpoint Blockade and Stereotactic Ablative Radiotherapy Can Stimulate Response to Immunotherapy in Metastatic Melanoma: A Case Report. Cureus 2019; 11:e4038. [PMID: 31011500 PMCID: PMC6456359 DOI: 10.7759/cureus.4038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Skin cancer is the most commonly diagnosed malignancy in the United States, and invasive cutaneous melanoma is responsible for the vast majority of skin cancer-related deaths. Treatment options for patients with regional nodal disease, in-transit metastases, or locally advanced or distant metastatic disease are challenging. Historically survival rates in this patient population are dismal. Improved systemic control is possible using targeted agents and checkpoint inhibitors have redefined treatment outcomes. Furthermore, multi-modal therapy incorporating radiation may improve survival outcomes by priming the immune system for antigen release and help in reversing T-cell exhaustion. Herein, we describe a patient with widespread metastatic melanoma with progressive systemic disease while receiving checkpoint inhibition therapy that was reversed after combined immunoradiotherapy. The patient is now more than 41 months from diagnosis with durable, stable systemic disease.
Collapse
Affiliation(s)
- Angel Moran
- Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, USA
| | - Soheila Azghadi
- Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, USA
| | - Emanual M Maverakis
- Dermatology, University of California Davis Comprehensive Cancer Center, Sacramento, USA
| | - Scott Christensen
- Hematology & Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, USA
| | - Brandon A Dyer
- Radiation Oncology, University of California Davis Comprehensive Cancer Center, Sacramento, USA
| |
Collapse
|