51
|
Safarzadeh Kozani P, Safarzadeh Kozani P, Ahmadi Najafabadi M, Yousefi F, Mirarefin SMJ, Rahbarizadeh F. Recent Advances in Solid Tumor CAR-T Cell Therapy: Driving Tumor Cells From Hero to Zero? Front Immunol 2022; 13:795164. [PMID: 35634281 PMCID: PMC9130586 DOI: 10.3389/fimmu.2022.795164] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/04/2022] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor T-cells (CAR-Ts) are known as revolutionary living drugs that have turned the tables of conventional cancer treatments in certain hematologic malignancies such as B-cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL) by achieving US Food and Drug Administration (FDA) approval based on their successful clinical outcomes. However, this type of therapy has not seen the light of victory in the fight against solid tumors because of various restricting caveats including heterogeneous tumor antigen expression and the immunosuppressive tumor microenvironments (TME) that negatively affect the tumor-site accessibility, infiltration, stimulation, activation, and persistence of CAR-Ts. In this review, we explore strategic twists including boosting vaccines and designing implementations that can support CAR-T expansion, proliferation, and tumoricidal capacity. We also step further by underscoring novel strategies for triggering endogenous antitumor responses and overcoming the limitation of poor CAR-T tumor-tissue infiltration and the lack of definitive tumor-specific antigens. Ultimately, we highlight how these approaches can address the mentioned arduous hurdles.
Collapse
Affiliation(s)
- Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Milad Ahmadi Najafabadi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Yousefi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
52
|
Peng Y, Fu S, Zhao Q. 2022 update on the scientific premise and clinical trials for IL-15 agonists as cancer immunotherapy. J Leukoc Biol 2022; 112:823-834. [PMID: 35616357 DOI: 10.1002/jlb.5mr0422-506r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/19/2022] [Indexed: 11/10/2022] Open
Abstract
Diverse cytokines and their receptors on immune cells constitute a highly complex network in the immune system. Some therapeutic cytokines and their derivatives have been approved for cancer treatment. IL-15 is an immune-regulating cytokine with multiple functions, among which the function of activating the immunity of cancer patients has great potential in cancer immunotherapy. In this review, we introduce the functions of IL-15 and discuss its role in regulating the immune system in different immune cells. Meanwhile, we will address the applications of IL-15 agonists in cancer immunotherapy and provide prospects for the next generation of therapeutic designs. Although many challenges remain, IL-15 agonists offer a new therapeutic option in the future direction of cancer immunotherapy.
Collapse
Affiliation(s)
- Yingjun Peng
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shengyu Fu
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, China
| |
Collapse
|
53
|
Grimmett E, Al-Share B, Alkassab MB, Zhou RW, Desai A, Rahim MMA, Woldie I. Cancer vaccines: past, present and future; a review article. Discov Oncol 2022; 13:31. [PMID: 35576080 PMCID: PMC9108694 DOI: 10.1007/s12672-022-00491-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/27/2022] [Indexed: 11/25/2022] Open
Abstract
Immunotherapy and vaccines have revolutionized disease treatment and prevention. Vaccines against infectious diseases have been in use for several decades. In contrast, only few cancer vaccines have been approved for human use. These include preventative vaccines against infectious agents associated with cancers, and therapeutic vaccines used as immunotherapy agents to treat cancers. Challenges in developing cancer vaccines include heterogeneity within and between cancer types, screening and identification of appropriate tumour-specific antigens, and the choice of vaccine delivery platforms. Recent advances in all of these areas and the lessons learnt from COVID-19 vaccines have significantly boosted interest in cancer vaccines. Further advances in these areas are expected to facilitate development of effective novel cancer vaccines. In this review, we aim to discuss the past, the present, and the future of cancer vaccines.
Collapse
Affiliation(s)
- Eddie Grimmett
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada
| | | | | | - Ryan Weng Zhou
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada
| | - Advait Desai
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada
| | - Mir Munir A Rahim
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada.
| | - Indryas Woldie
- Department of Biomedical Sciences, University of Windsor, Windsor, ON, Canada.
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
54
|
Sia J, Neeson PJ, Haynes NM. Basic cancer immunology for radiation oncologists. J Med Imaging Radiat Oncol 2022; 66:508-518. [PMID: 35352493 PMCID: PMC9311072 DOI: 10.1111/1754-9485.13406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/07/2022] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Abstract
Although the impressive clinical responses seen with modern cancer immunotherapy are currently limited to a subset of patients, the underlying paradigm shift has resulted in now hardly a segment in oncology that has not been touched by the immuno‐oncology revolution. A growing body of data indicates that radiation therapy (RT) can modulate the tumour immune microenvironment and complement cancer immunotherapy via non‐overlapping mechanisms to reinvigorate immunity against cancer. Thus, increasingly RT is viewed as a highly unique partner for immunotherapy across the spectrum of cancer settings, as radiobiology and cancer immunology foreseeably become more intertwined. Considering these developments, this review summarises the key concepts and terminology in immunology for the radiation oncologist, with a focus on the cancer setting and with reference to important recent advances. These concepts will provide a starting point for understanding the strategies that underlie current and emerging immunotherapy trials, as well as the indirect effects of RT by which immune responses against cancer are shaped.
Collapse
Affiliation(s)
- Joseph Sia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul J Neeson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nicole M Haynes
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
55
|
Vincent MP, Navidzadeh JO, Bobbala S, Scott EA. Leveraging self-assembled nanobiomaterials for improved cancer immunotherapy. Cancer Cell 2022; 40:255-276. [PMID: 35148814 PMCID: PMC8930620 DOI: 10.1016/j.ccell.2022.01.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
Nanomaterials and targeted drug delivery vehicles improve the therapeutic index of drugs and permit greater control over their pharmacokinetics, biodistribution, and bioavailability. Here, nanotechnologies applied to cancer immunotherapy are discussed with a focus on current and next generation self-assembling drug delivery systems composed of lipids and/or polymers. Topics covered include the fundamental design, suitability, and inherent properties of nanomaterials that induce anti-tumor immune responses and support anti-cancer vaccination. Established active and passive targeting strategies as well as newer "indirect" methods are presented together with insights into how nanocarrier structure and surface chemistry can be leveraged for controlled delivery to the tumor microenvironment while minimizing off-target effects.
Collapse
Affiliation(s)
- Michael P Vincent
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Justin O Navidzadeh
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA; Interdisciplinary Biological Sciences, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA; Simpson Querrey Institute, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
56
|
Kasherman L, Siu DHW, Woodford R, Harris CA. Angiogenesis Inhibitors and Immunomodulation in Renal Cell Cancers: The Past, Present, and Future. Cancers (Basel) 2022; 14:1406. [PMID: 35326557 PMCID: PMC8946206 DOI: 10.3390/cancers14061406] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis inhibitors have been adopted into the standard armamentarium of therapies for advanced-stage renal cell carcinomas (RCC), but more recently, combination regimens with immune checkpoint inhibitors have demonstrated better outcomes. Despite this, the majority of affected patients still eventually experience progressive disease due to therapeutic resistance mechanisms, and there remains a need to develop novel therapeutic strategies. This article will review the synergistic mechanisms behind angiogenesis and immunomodulation in the tumor microenvironment and discuss the pre-clinical and clinical evidence for both clear-cell and non-clear-cell RCC, exploring opportunities for future growth in this exciting area of drug development.
Collapse
Affiliation(s)
- Lawrence Kasherman
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
- Department of Medical Oncology, Illawarra Cancer Care Centre, Wollongong, NSW 2500, Australia
| | - Derrick Ho Wai Siu
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- National Health Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| | - Rachel Woodford
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- Faculty of Medciine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Carole A. Harris
- Department of Medical Oncology, St. George Hospital, Kogarah, NSW 2217, Australia; (D.H.W.S.); (R.W.); (C.A.H.)
- St. George and Sutherland Clinical Schools, University of New South Wales, Sydney, NSW 2217, Australia
| |
Collapse
|
57
|
Zhang Y, Zhao M, Gao H, Yu G, Zhao Y, Yao F, Yang W. MAPK signalling-induced phosphorylation and subcellular translocation of PDHE1α promotes tumour immune evasion. Nat Metab 2022; 4:374-388. [PMID: 35315437 DOI: 10.1038/s42255-022-00543-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/02/2022] [Indexed: 11/09/2022]
Abstract
Tumour cells utilize multiple strategies to evade the immune system, but the underlying metabolic mechanisms remain poorly understood. The pyruvate dehydrogenase (PDH) complex converts pyruvate to acetyl-coenzyme A in mitochondria, thereby linking glycolysis to the ricarboxylic acid cycle. Here we show that the PDH complex E1 subunit α (PDHE1α) is also located in the cytosol. Cytosolic PDHE1α interacts with IKKβ and protein phosphatase 1B, thereby facilitating the inhibition of the NF-κB pathway. Cytosolic PDHE1α can be phosphorylated at S327 by ERK2 and translocated into mitochondria. Decreased cytosolic PDHE1α levels restore NF-κB signalling, whereas increased mitochondrial PDHE1α levels drive α-ketoglutarate production and promote reactive oxygen species detoxification. Synergistic activation of NF-κB and reactive oxygen species detoxification promotes tumour cell survival and enhances resistance to cytotoxic lymphocytes. Consistently, low levels of PDHE1α phosphorylation are associated with poor prognosis of patients with lung cancer. Our findings show a mechanism through which phosphorylation-dependent subcellular translocation of PDHE1α promotes tumour immune evasion.
Collapse
Affiliation(s)
- Yajuan Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ming Zhao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Guanzhen Yu
- Medical Artificial Intelligence Laboratory, Zhejiang Institute of Digital Media, Chinese Academy of Science, Shaoxing, China
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Weiwei Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
58
|
Vimalathas G, Kristensen BW. Expression, prognostic significance and therapeutic implications of PD-L1 in gliomas. Neuropathol Appl Neurobiol 2022; 48:e12767. [PMID: 34533233 PMCID: PMC9298327 DOI: 10.1111/nan.12767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 08/27/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022]
Abstract
The advent of checkpoint immunotherapy, particularly with programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors, has provided ground-breaking results in several advanced cancers. Substantial efforts are being made to extend these promising therapies to other refractory cancers such as gliomas, especially glioblastoma, which represents the most frequent and malignant glioma and carries an exceptionally grim prognosis. Thus, there is a need for new therapeutic strategies with related biomarkers. Gliomas have a profoundly immunosuppressive tumour micro-environment and evade immunological destruction by several mechanisms, one being the expression of inhibitory immune checkpoint molecules such as PD-L1. PD-L1 is recognised as an important therapeutic target and its expression has been shown to hold prognostic value in different cancers. Several clinical trials have been launched and some already completed, but PD-1/PD-L1 inhibitors have yet to show convincing clinical efficacy in gliomas. Part of the explanation may reside in the vast molecular heterogeneity of gliomas and a complex interplay within the tumour micro-environment. In parallel, critical knowledge about PD-L1 expression is beginning to accumulate including knowledge on expression levels, testing methodology, co-expression with other checkpoint molecules and prognostic and predictive value. This article reviews these aspects and points out areas where biomarker research is needed to develop more successful checkpoint-related therapeutic strategies in gliomas.
Collapse
Affiliation(s)
| | - Bjarne Winther Kristensen
- Department of PathologyOdense University HospitalOdenseDenmark
- Department of Pathology, RigshospitaletCopenhagen University HospitalCopenhagenDenmark
- Department of Clinical Medicine and Biotech Research and Innovation Center (BRIC)University of CopenhagenCopenhagenDenmark
| |
Collapse
|
59
|
Liu R, Dollinger E, Nie Q. Machine Learning of Single Cell Transcriptomic Data From anti-PD-1 Responders and Non-responders Reveals Distinct Resistance Mechanisms in Skin Cancers and PDAC. Front Genet 2022; 12:806457. [PMID: 35178072 PMCID: PMC8844526 DOI: 10.3389/fgene.2021.806457] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/16/2021] [Indexed: 01/31/2023] Open
Abstract
Immune checkpoint therapies such as PD-1 blockade have vastly improved the treatment of numerous cancers, including basal cell carcinoma (BCC). However, patients afflicted with pancreatic ductal carcinoma (PDAC), one of the deadliest malignancies, overwhelmingly exhibit negative responses to checkpoint therapy. We sought to combine data analysis and machine learning to differentiate the putative mechanisms of BCC and PDAC non-response. We discover that increased MHC-I expression in malignant cells and suppression of MHC and PD-1/PD-L expression in CD8+ T cells is associated with nonresponse to treatment. Furthermore, we leverage machine learning to predict response to PD-1 blockade on a cellular level. We confirm divergent resistance mechanisms between BCC, PDAC, and melanoma and highlight the potential for rapid and affordable testing of gene expression in BCC patients to accurately predict response to checkpoint therapies. Our findings present an optimistic outlook for the use of quantitative cross-cancer analyses in characterizing immune responses and predicting immunotherapy outcomes.
Collapse
Affiliation(s)
- Ryan Liu
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States
| | - Emmanuel Dollinger
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, United States,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States,*Correspondence: Emmanuel Dollinger, ; Qing Nie,
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States,Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States,Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, United States,NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States,*Correspondence: Emmanuel Dollinger, ; Qing Nie,
| |
Collapse
|
60
|
Iacovino ML, Miceli CC, De Felice M, Barone B, Pompella L, Chiancone F, Di Zazzo E, Tirino G, Della Corte CM, Imbimbo C, De Vita F, Crocetto F. Novel Therapeutic Opportunities in Neoadjuvant Setting in Urothelial Cancers: A New Horizon Opened by Molecular Classification and Immune Checkpoint Inhibitors. Int J Mol Sci 2022; 23:1133. [PMID: 35163064 PMCID: PMC8835066 DOI: 10.3390/ijms23031133] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022] Open
Abstract
Muscle invasive bladder cancer (MIBC) is a widespread malignancy with a worse prognosis often related to a late diagnosis. For early-stage MIBC pts, a multidisciplinary approach is mandatory to evaluate the timing of neoadjuvant chemotherapy (NAC) and surgery. The current standard therapy is platinum-based NAC (MVAC-methotrexate, vinblastine, doxorubicin, and cisplatin or Platinum-Gemcitabine regimens) followed by radical cystectomy (RC) with lymphadenectomy. However, preliminary data from Vesper trial highlighted that dose-dense NAC MVAC is endowed with a good pathological response but shows low tolerability. In the last few years, translational-based research approaches have identified several candidate biomarkers of NAC esponsiveness, such as ERCC2, ERBB2, or DNA damage response (DDR) gene alterations. Moreover, the recent consensus MIBC molecular classification identified six molecular subtypes, characterized by different sensitivity to chemo- or targeted or immunotherapy, that could open a novel procedure for patient selection and also for neoadjuvant therapies. The Italian PURE-01 phase II Trial extended data on efficacy and resistance to Immune Checkpoint Inhibitors (ICIs) in this setting. In this review, we summarize the most relevant literature data supporting NAC use in MIBC, focusing on novel therapeutic strategies such as immunotherapy, considering the better patient stratification and selection emerging from novel molecular classification.
Collapse
Affiliation(s)
- Maria Lucia Iacovino
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Chiara Carmen Miceli
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Marco De Felice
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Biagio Barone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy; (B.B.); (C.I.)
| | - Luca Pompella
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | | | - Erika Di Zazzo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, UOC Laboratorio Analisi P.O. “A. Cardarelli”, 86100 Campobasso, Italy;
| | - Giuseppe Tirino
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Carminia Maria Della Corte
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy; (B.B.); (C.I.)
| | - Ferdinando De Vita
- Department of Precision Medicine, Medical Oncology, University of Campania Luigi Vanvitelli, Via Sergio Pansini 5, 80131 Naples, Italy; (M.L.I.); (C.C.M.); (M.D.F.); (L.P.); (G.T.); (C.M.D.C.); (F.D.V.)
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy; (B.B.); (C.I.)
| |
Collapse
|
61
|
Wang L, Chen Y, Liu X, Li Z, Dai X. The Application of CRISPR/Cas9 Technology for Cancer Immunotherapy: Current Status and Problems. Front Oncol 2022; 11:704999. [PMID: 35111663 PMCID: PMC8801488 DOI: 10.3389/fonc.2021.704999] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer is one of the main causes of disease-related deaths in the world. Although cancer treatment strategies have been improved in recent years, the survival time of cancer patients is still far from satisfied. Cancer immunotherapy, such as Oncolytic virotherapy, Immune checkpoints inhibition, Chimeric antigen receptor T (CAR-T) cell therapy, Chimeric antigen receptor natural killer (CAR-NK) cell therapy and macrophages genomic modification, has emerged as an effective therapeutic strategy for different kinds of cancer. However, many patients do not respond to the cancer immunotherapy which warrants further investigation to optimize this strategy. The clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR/Cas9), as a versatile genome engineering tool, has become popular in the biology research field and it was also applied to optimize tumor immunotherapy. Moreover, CRISPR-based high-throughput screening can be used in the study of immunomodulatory drug resistance mechanism. In this review, we summarized the development as well as the application of CRISPR/Cas9 technology in the cancer immunotherapy and discussed the potential problems that may be caused by this combination.
Collapse
Affiliation(s)
- Luyao Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yurong Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xinrui Liu
- Neurosurgery Department, First Hospital, Jilin University, Changchun, China
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai,
| |
Collapse
|
62
|
Control of Tumors by Antigen-Specific CD8 + T Cells through PDL1-Targeted Delivery of Antigenic Peptide. J Immunol Res 2022; 2022:9054569. [PMID: 35028321 PMCID: PMC8752305 DOI: 10.1155/2022/9054569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
Tumor antigen-specific T cell function is limited by immune tolerance in the tumor microenvironment. In the tumor microenvironment, tumor cells upregulate PD-L1 expression to promote T cell exhaustion by PD-1/PD-L1 interactions and undergo mutations to avoid being targeted by tumor antigen-specific T cells. Thus, tumor cells escape the immune surveillance by causing immune tolerance. We reason that a chimeric molecule made of a PD-L1-specific antibody linked to a cleavable antigenic peptide can target the antigenic peptide to the tumor microenvironment, resulting in the blockade of the PD-1/PD-L1 pathway and killing tumor cells through the coating of antigenic peptide. Here, we have generated a therapeutic chimeric protein containing the PD-L1 single-chain variable fragment (scFv) linked to a cleavable model cytotoxic T lymphocyte (CTL) epitope: E7 CTL peptide. Our study demonstrated that our chimeric protein (named PDL1-scFv-Fc-RE7) can target PD-L1-expressing tumor cells and enable E7 presentation by releasing cleavable E7 CTL peptide to coat tumor cells, resulting in tumor clearance by E7-specific CD8+ T cells. The presentation of the E7 peptide by cancer cells can then render tumor cells susceptible to the killing of preexisting E7-specific CD8+ T cells and contribute to tumor clearance. Our finding suggests a synergistic approach to not only enhance antigen-specific tumor clearance but also bypass immune tolerance.
Collapse
|
63
|
Khan E, Shrestha AK, Elkhooly M, Wilson H, Ebbert M, Srivastava S, Wen S, Rollins S, Sriwastava S. CNS and PNS manifestation in immune checkpoint inhibitors: A systematic review. J Neurol Sci 2022; 432:120089. [PMID: 34942546 DOI: 10.1016/j.jns.2021.120089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Immunomodulatory therapies, including the use of immune checkpoint inhibitors (ICIs), have made a profound impact on treatment of advanced cancers in recent decades. Neurologic immune-related adverse events (irAEs) related to use of these agents are rare but potentially fatal sequelae. This systematic reviewed aimed to describe onset, clinical features, treatment, and outcome of neurological irAEs following ICI usage. METHODS A systematic literature search was conducted to identify all case reports (n = 168) and case series (n = 29) describing neurological irAEs (n = 255 patients). Patient demographics, clinical features, and clinical courses were extracted and used to assess statistical relationships between reported variables. RESULTS Of reports describing neurological irAEs related to ICI use, the majority of cases were in men (66%) and patients above the age of fifty (85%). Disorders of the peripheral nervous system (PNS, 83%) were more common than central nervous system involvement. Neuromuscular disorders were the most common type of neurological irAE (e.g. myasthenia gravis, 36%), followed by peripheral neuropathies (16%), followed by all CNS disorders combined (15%). Most cases presented within the first 5 doses of ICI treatment. Most patients improved clinically, but 24% of cases were fatal. Mortality was highest in patients with neuromuscular irAEs, such as myasthenia gravis and myositis. CONCLUSION This systematic literature review describes the largest collection of neurological irAEs to date including both CNS and PNS manifestations of ICIs. The information described herein can be used to better inform monitoring and treatment of patients undergoing treatment with ICIs.
Collapse
Affiliation(s)
- Erum Khan
- B.J. Medical College and Civil Hospital, Ahmedabad, India
| | | | | | - Hannah Wilson
- West Virginia University, School of Medicine, Morgantown, WV, United States of America
| | - Michael Ebbert
- Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States of America
| | | | - Sijin Wen
- Department of Biostatistics, West Virginia University, Morgantown, WV, United States of America
| | - Steven Rollins
- Department of Biostatistics, West Virginia University, Morgantown, WV, United States of America
| | - Shitiz Sriwastava
- West Virginia University, School of Medicine, Morgantown, WV, United States of America; Department of Neurology, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States of America; Depratment of Neurology, Wayne State University, United States of America; West Virginia Clinical and Translational Science Institute, Morgantown, WV, United States of America.
| |
Collapse
|
64
|
Regulation of Immune Cells by microRNAs and microRNA-Based Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1385:75-108. [DOI: 10.1007/978-3-031-08356-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
65
|
TIL expansion with high dose IL-2 or low dose IL-2 with anti-CD3/anti-CD28 stimulation provides different quality of TIL-expanded T cell clones. J Immunol Methods 2022; 503:113229. [DOI: 10.1016/j.jim.2022.113229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/02/2021] [Accepted: 01/24/2022] [Indexed: 11/18/2022]
|
66
|
Cytosolic Self-DNA—A Potential Source of Chronic Inflammation in Aging. Cells 2021; 10:cells10123544. [PMID: 34944052 PMCID: PMC8700131 DOI: 10.3390/cells10123544] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/28/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is the consequence of a lifelong accumulation of stochastic damage to tissues and cellular components. Advancing age closely associates with elevated markers of innate immunity and low-grade chronic inflammation, probably reflecting steady increasing incidents of cellular and tissue damage over the life course. The DNA sensing cGAS-STING signaling pathway is activated by misplaced cytosolic self-DNA, which then initiates the innate immune responses. Here, we hypothesize that the stochastic release of various forms of DNA from the nucleus and mitochondria, e.g., because of DNA damage, altered nucleus integrity, and mitochondrial damage, can result in chronic activation of inflammatory responses that characterize the aging process. This cytosolic self-DNA-innate immunity axis may perturb tissue homeostasis and function that characterizes human aging and age-associated pathology. Proper techniques and experimental models are available to investigate this axis to develop therapeutic interventions.
Collapse
|
67
|
Li Z, Sun G, Sun G, Cheng Y, Wu L, Wang Q, Lv C, Zhou Y, Xia Y, Tang W. Various Uses of PD1/PD-L1 Inhibitor in Oncology: Opportunities and Challenges. Front Oncol 2021; 11:771335. [PMID: 34869005 PMCID: PMC8635629 DOI: 10.3389/fonc.2021.771335] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
The occurrence and development of cancer are closely related to the immune escape of tumor cells and immune tolerance. Unlike previous surgical, chemotherapy, radiotherapy and targeted therapy, tumor immunotherapy is a therapeutic strategy that uses various means to stimulate and enhance the immune function of the body, and ultimately achieves the goal of controlling tumor cells.With the in-depth understanding of tumor immune escape mechanism and tumor microenvironment, and the in-depth study of tumor immunotherapy, immune checkpoint inhibitors represented by Programmed Death 1/Programmed cell Death-Ligand 1(PD-1/PD-L1) inhibitors are becoming increasingly significant in cancer medication treatment. employ a variety of ways to avoid detection by the immune system, a single strategy is not more effective in overcoming tumor immune evasion and metastasis. Combining different immune agents or other drugs can effectively address situations where immunotherapy is not efficacious, thereby increasing the chances of success and alternative access to alternative immunotherapy. Immune combination therapies for cancer have become a hot topic in cancer treatment today. In this paper, several combination therapeutic modalities of PD1/PD-L1 inhibitors are systematically reviewed. Finally, an analysis and outlook are provided in the context of the recent advances in combination therapy with PD1/PD-L1 inhibitors and the pressing issues in this field.
Collapse
Affiliation(s)
- Zhitao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ye Cheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yichan Zhou
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| |
Collapse
|
68
|
Zhu J, Yuan Y, Wan X, Yin D, Li R, Chen W, Suo C, Song H. Immunotherapy (excluding checkpoint inhibitors) for stage I to III non-small cell lung cancer treated with surgery or radiotherapy with curative intent. Cochrane Database Syst Rev 2021; 12:CD011300. [PMID: 34870327 PMCID: PMC8647093 DOI: 10.1002/14651858.cd011300.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common lung cancer, accounting for approximately 80% to 85% of all cases. For people with localised NSCLC (stages I to III), it has been speculated that immunotherapy may be helpful for reducing postoperative recurrence rates, or improving the clinical outcomes of current treatment for unresectable tumours. This is an update of a Cochrane Review first published in 2017 and it includes two new randomised controlled trials (RCTs). OBJECTIVES To assess the effectiveness and safety of immunotherapy (excluding checkpoint inhibitors) among people with localised NSCLC of stages I to III who received curative intent of radiotherapy or surgery. SEARCH METHODS We searched the following databases (from inception to 19 May 2021): CENTRAL, MEDLINE, Embase, CINAHL, and five trial registers. We also searched conference proceedings and reference lists of included trials. SELECTION CRITERIA We included RCTs conducted in adults (≥ 18 years) diagnosed with NSCLC stage I to III after surgical resection, and those with unresectable locally advanced stage III NSCLC receiving radiotherapy with curative intent. We included participants who underwent primary surgical treatment, postoperative radiotherapy or chemoradiotherapy if the same strategy was provided for both intervention and control groups. DATA COLLECTION AND ANALYSIS Two review authors independently selected eligible trials, assessed risk of bias, and extracted data. We used survival analysis to pool time-to-event data, using hazard ratios (HRs). We used risk ratios (RRs) for dichotomous data, and mean differences (MDs) for continuous data, with 95% confidence intervals (CIs). Due to clinical heterogeneity (immunotherapeutic agents with different underlying mechanisms), we combined data by applying random-effects models. MAIN RESULTS We included 11 RCTs involving 5128 participants (this included 2 new trials with 188 participants since the last search dated 20 January 2017). Participants who underwent surgical resection or received curative radiotherapy were randomised to either an immunotherapy group or a control group. The immunological interventions were active immunotherapy Bacillus Calmette-Guérin (BCG) adoptive cell transfer (i.e. transfer factor (TF), tumour-infiltrating lymphocytes (TIL), dendritic cell/cytokine-induced killer (DC/CIK), antigen-specific cancer vaccines (melanoma-associated antigen 3 (MAGE-A3) and L-BLP25), and targeted natural killer (NK) cells. Seven trials were at high risk of bias for at least one of the risk of bias domains. Three trials were at low risk of bias across all domains and one small trial was at unclear risk of bias as it provided insufficient information. We included data from nine of the 11 trials in the meta-analyses involving 4863 participants. There was no evidence of a difference between the immunotherapy agents and the controls on any of the following outcomes: overall survival (HR 0.94, 95% CI 0.84 to 1.05; P = 0.27; 4 trials, 3848 participants; high-quality evidence), progression-free survival (HR 0.94, 95% CI 0.86 to 1.03; P = 0.19; moderate-quality evidence), adverse events (RR 1.12, 95% CI 0.97 to 1.28; P = 0.11; 4 trials, 4126 evaluated participants; low-quality evidence), and severe adverse events (RR 1.14, 95% CI 0.92 to 1.40; 6 trials, 4546 evaluated participants; low-quality evidence). Survival rates at different time points showed no evidence of a difference between immunotherapy agents and the controls. Survival rate at 1-year follow-up (RR 1.02, 95% CI 0.96 to 1.08; I2 = 57%; 7 trials, 4420 participants; low-quality evidence), 2-year follow-up (RR 1.02, 95% CI 0.93 to 1.12; 7 trials, 4420 participants; moderate-quality evidence), 3-year follow-up (RR 0.99, 95% CI 0.90 to 1.09; 7 trials, 4420 participants; I2 = 22%; moderate-quality evidence) and at 5-year follow-up (RR 0.98, 95% CI 0.86 to 1.12; I2 = 0%; 7 trials, 4389 participants; moderate-quality evidence). Only one trial reported overall response rates. Two trials provided health-related quality of life results with contradicting results. AUTHORS' CONCLUSIONS: Based on this updated review, the current literature does not provide evidence that suggests a survival benefit from adding immunotherapy (excluding checkpoint inhibitors) to conventional curative surgery or radiotherapy, for people with localised NSCLC (stages I to III). Several ongoing trials with immune checkpoints inhibitors (PD-1/PD-L1) might bring new insights into the role of immunotherapy for people with stages I to III NSCLC.
Collapse
Affiliation(s)
- Jianwei Zhu
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Yun Yuan
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Wan
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Yin
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Li
- Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenwen Chen
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Suo
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
| | - Huan Song
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| |
Collapse
|
69
|
Diagnostic and Prognostic Implications of Caspase-1 and PD-L1 Co-Expression Patterns in Myelodysplastic Syndromes. Cancers (Basel) 2021; 13:cancers13225712. [PMID: 34830867 PMCID: PMC8616142 DOI: 10.3390/cancers13225712] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary Myelodysplastic syndromes (MDS) originate from mutated hematopoietic stem and progenitor cells. Despite recent advances in genetics, the mechanisms involved in clonal progression remain largely unknown. We performed an exploratory, case-control study to identify immune-related biomarkers with diagnostic and prognostic utility. Our study suggests a combined Casp1/PD-L1 assessment to distinguish reactive conditions from lower- and higher-risk MDS. These immune-related biomarkers may help to personalize immuno-therapies but require further validation in prospective studies. Abstract Background: The inflammasome plays an essential role in lower risk MDS and immune subversion, with the up-regulation of immune checkpoint molecules in the progression to higher-risk disease. In this study, we explored the utility of immune-related biomarkers for the diagnosis and prognosis of MDS. Methods: We performed an exploratory, case-control study with 20 randomly selected MDS patients and nine controls with non-inflammatory (n = 3) and inflammatory conditions (n = 6). Patients were stratified in groups of lower (n = 10) and higher risk (n = 10) using IPSS-R. For the exploration of inflammasome and immune checkpoint activities, the expression of caspase-1 (Casp1), programmed cell death protein 1 (PD-1) and its ligand (PD-L1) were assessed in bone marrow samples using immunohistochemistry. Results: In multivariate analysis, we observed significant differences for Casp1 but not PD1/PD-L1 expression in our four conditions (p = 0.003). We found a discordant co-expression of Casp1/PD-L1 in MDS (rho = −0.41, p = 0.07) compared with a concordant co-expression in controls (rho = 0.64, p = 0.06). Neutrophil counts correlated directly with Casp1 (rho = 0.57, p = 0.009) but inversely with PD-L1 expression (rho = −0.58, p = 0.007). Conclusion: We identified characteristic discordant co-expression patterns in lower- (Casp1high/PD-L1low) and higher-risk MDS (Casp1low/PD-L1high), contrasting with concordant patterns in the non-inflammatory (Casp1low/PD-L1low) and inflammatory conditions (Casp1high/PD-L1high). Further validation is warranted in larger, prospective studies.
Collapse
|
70
|
Reddy VK, Shrestha DB, Gaire S, Mir WAY, Kassem M. Long-Term Favorable Outcome With Nivolumab in a Case of Advanced Non-Small Cell Lung Cancer: A Case Report. Cureus 2021; 13:e18526. [PMID: 34765329 PMCID: PMC8575281 DOI: 10.7759/cureus.18526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 11/05/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) constitutes around 85% of lung cancer cases. Advanced non-small cell lung cancer has a poor prognosis. Immunotherapy plays a pivotal role in managing advanced non-small cell lung cancer not positive for driver mutations. Nivolumab is a monoclonal antibody against programmed death-ligand 1 (PDL1). It is approved as a second-line treatment for patients with advanced non-small cell lung cancer who progress on or after chemotherapy. We present a case of a 71-year-old female with advanced non-small cell lung cancer without any driver mutations diagnosed four years ago. Her disease progressed while on conventional chemotherapy, and she was started on nivolumab three and a half years ago. Her lung nodules resolved, she did not show signs of progression, and her performance status improved while on nivolumab. This case report highlights the current role of nivolumab in the management of NSCLC. Patients whose condition worsens while on conventional chemotherapy can respond very well to modern targeted immunotherapy.
Collapse
Affiliation(s)
| | - Dhan B Shrestha
- Department of Internal Medicine, Mount Sinai Hospital, Chicago, USA
| | - Suman Gaire
- Department of Emergency Medicine, Palpa Hospital, Palpa, NPL
| | | | - Mohammed Kassem
- Department of Hematology and Oncology, Mount Sinai Hospital, Chicago, USA
| |
Collapse
|
71
|
Subramaniam S, Anandha Rao JS, Ramdas P, Ng MH, Kannan Kutty M, Selvaduray KR, Radhakrishnan AK. Reduced infiltration of regulatory T cells in tumours from mice fed daily with gamma-tocotrienol supplementation. Clin Exp Immunol 2021; 206:161-172. [PMID: 34331768 PMCID: PMC8506134 DOI: 10.1111/cei.13650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/06/2021] [Accepted: 07/11/2021] [Indexed: 01/08/2023] Open
Abstract
Gamma-tocotrienol (γT3) is an analogue of vitamin E with beneficial effects on the immune system, including immune-modulatory properties. This study reports the immune-modulatory effects of daily supplementation of γT3 on host T helper (Th) and T regulatory cell (Treg ) populations in a syngeneic mouse model of breast cancer. Female BALB/c mice were fed with either γT3 or vehicle (soy oil) for 2 weeks via oral gavage before they were inoculated with syngeneic 4T1 mouse mammary cancer cells (4T1 cells). Supplementation continued until the mice were euthanized. Mice (n = 6) were euthanized at specified time-points for various analysis (blood leucocyte, cytokine production and immunohistochemistry). Tumour volume was measured once every 7 days. Gene expression studies were carried out on tumour-specific T lymphocytes isolated from splenic cultures. Supplementation with γT3 increased CD4+ (p < 0.05), CD8+ (p < 0.05) T-cells and natural killer cells (p < 0.05) but suppressed Treg cells (p < 0.05) in peripheral blood when compared to animals fed with the vehicle. Higher interferon (IFN)-γ and lower transforming growth factor (TGF)-ꞵ levels were noted in the γT3 fed mice. Immunohistochemistry findings revealed higher infiltration of CD4+ cells, increased expression of interleukin-12 receptor-beta-2 (IL-12ꞵ2R), interleukin (IL)-24 and reduced expression of cells that express the forkhead box P3 (FoxP3) in tumours from the γT3-fed animals. Gene expression studies showed the down-regulation of seven prominent genes in splenic CD4+ T cells isolated from γT3-fed mice. Supplementation with γT3 from palm oil-induced T cell-dependent cell-mediated immune responses and suppressed T cells in the tumour microenvironment in a syngeneic mouse model of breast cancer.
Collapse
Affiliation(s)
- Shonia Subramaniam
- School of Postgraduate StudiesInternational Medical UniversityKuala LumpurMalaysia
- Product Development and Advisory ServicesMalaysian Palm Oil BoardKajangMalaysia
| | - Jeya Seela Anandha Rao
- Pathology DivisionSchool of MedicineInternational Medical UniversityKuala LumpurMalaysia
| | - Premdass Ramdas
- Division of Applied Biomedical Sciences and BiotechnologySchool of Health SciencesInternational Medical UniversityKuala LumpurMalaysia
| | - Mei Han Ng
- Engineering and ProcessingMalaysian Palm Oil BoardKajangMalaysia
| | | | | | - Ammu Kutty Radhakrishnan
- Pathology DivisionSchool of MedicineInternational Medical UniversityKuala LumpurMalaysia
- Jeffery Cheah School of Medicine and Health SciencesMonash University MalaysiaBandar SunwaySelangorMalaysia
| |
Collapse
|
72
|
Jang A, Adler DM, Rauterkus GP, Bilen MA, Barata PC. Immunotherapies in Genitourinary Oncology: Where Are We Now? Where Are We Going? Cancers (Basel) 2021; 13:cancers13205065. [PMID: 34680214 PMCID: PMC8533722 DOI: 10.3390/cancers13205065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Genitourinary malignancies include cancers along the urinary tract and the male reproductive tract, encompassing the adrenal glands, kidneys, bladder, prostate, and testicles. Immunotherapy, which treats cancer by using the immune system to attack malignant cells, has historically been successful in treating some types of genitourinary cancers, especially of the bladder and kidney. In the past decade, a more precise method of immunotherapy, known as immune checkpoint inhibition, has gained popularity as it enhances the immune system’s ability to recognize and destroy tumor cells. Several immune checkpoint inhibitors have achieved success in patients with advanced genitourinary cancers. This review provides a brief overview of traditional immunotherapies, focuses on how immune checkpoint inhibitors have achieved success in patients with advanced cancers, and investigates the role for immunotherapy in genitourinary malignancies in the future. Abstract For decades, limited options existed to treat metastatic genitourinary cancers, including treatment options that could be classified as immunotherapy. Historically, immunotherapy centered on systemic cytokines for the treatment of metastatic kidney cancer, which had several adverse effects, as well as the Bacillus Calmette–Guérin vaccine for non-metastatic bladder cancer. Within the past decade, advances in immunotherapy have led to several approvals from the United States Food and Drug Administration, particularly in the field of immune checkpoint inhibition. Immune checkpoint inhibitors (ICIs) are now being used extensively to treat multiple solid tumors, including kidney and bladder cancers, and they are also being tested in many other cancers. Despite encouraging data from phase 2/3 clinical trials, less is known about biomarkers that may predict better response to ICIs. The effect of ICIs in genitourinary cancers is heterogeneous, with some tumor types having little clinical data available, or ICIs having limited activity in other tumors. In this review, we briefly discuss approved immunotherapy agents prior to the time of ICIs. Then, given the emergence of this class of agents, we summarize the several important ICIs and the clinical trials that led to their approval. Finally, we mention ongoing and future clinical trials.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - David M. Adler
- Section of Hematology and Medical Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | | | - Mehmet A. Bilen
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Pedro C. Barata
- Section of Hematology and Medical Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA;
- Tulane Cancer Center, New Orleans, LA 70112, USA
- Correspondence: ; Tel.: +1-504-988-1236
| |
Collapse
|
73
|
Liu L, Du X, Fang J, Zhao J, Guo Y, Zhao Y, Zou C, Yan X, Li W. Development of an Interferon Gamma Response-Related Signature for Prediction of Survival in Clear Cell Renal Cell Carcinoma. J Inflamm Res 2021; 14:4969-4985. [PMID: 34611422 PMCID: PMC8485924 DOI: 10.2147/jir.s334041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
Background Interferon plays a crucial role in the pathogenesis and progression of tumors. Clear cell renal cell carcinoma (ccRCC) represents a prevalent malignant urinary system tumor. An effective predictive model is required to evaluate the prognosis of patients to optimize treatment. Materials and Methods RNA-sequencing data and clinicopathological data from TCGA were involved in this retrospective study. The IFN-γ response genes with significantly different gene expression were screened out. Univariate Cox regression, LASSO regression and multivariate Cox regression were used to establish a new prognostic scoring model for the training group. Survival curves and ROC curves were drawn, and nomogram was constructed. At the same time, we conducted subgroup analysis and experimental verification using our own samples. Finally, we evaluated the relatedness between the prognostic signature and immune infiltration landscapes. In addition, the sensitivity of different risk groups to six drugs and immune checkpoint inhibitors was calculated. Results The IFN-γ response-related signature included 7 genes: C1S, IFI44, ST3GAL5, NUP93, TDRD7, DDX60, and ST8SIA4. The survival curves of the training and testing groups showed the model's effectiveness (P = 4.372e-11 and P = 1.08e-08, respectively), the ROC curves showed that the signature was stable, and subgroup analyses showed the wide applicability of the model (P<0.001). Multivariate Cox regression analysis showed that the risk model was an independent prognostic factor of ccRCC. A high-risk score may represent an immunosuppressive microenvironment, while the high-risk group exhibited poor sensitivity to drugs. Conclusion Our findings strongly indicate that the IFN-γ response-related signature can be used as an effective prognostic indicator of ccRCC.
Collapse
Affiliation(s)
- Lixiao Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xuedan Du
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Jintao Fang
- Department of Urinary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Jinduo Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Yong Guo
- Department of Urinary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Ye Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Chengyang Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Xiaojian Yan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Wenfeng Li
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
74
|
JNJ-64041757 (JNJ-757), a Live, Attenuated, Double-Deleted Listeria monocytogenes-Based Immunotherapy in Patients With NSCLC: Results From Two Phase 1 Studies. JTO Clin Res Rep 2021; 2:100103. [PMID: 34589981 PMCID: PMC8474274 DOI: 10.1016/j.jtocrr.2020.100103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/22/2023] Open
Abstract
Introduction JNJ-64041757 (JNJ-757) is a live, attenuated, double-deleted Listeria monocytogenes-based immunotherapy expressing human mesothelin. JNJ-757 was evaluated in patients with advanced NSCLC as monotherapy (phase 1) and in combination with nivolumab (phase 1b/2). Methods Patients with stage IIIB/IV NSCLC who had received previous therapy were treated with JNJ-757 (1 × 108 or 1 × 109 colony-forming units [CFUs]) alone (NCT02592967) or JNJ-757 (1 × 109 CFU) plus intravenous nivolumab 240 mg (NCT03371381). Study objectives included the assessment of immunogenicity, safety, and efficacy. Results In the monotherapy study, 18 patients (median age 63.5 y; women 61%) were treated with JNJ-757 (1 × 108 or 1 × 109 CFU) with a median duration of 1.4 months (range: 0-29). The most common adverse events (AEs) were pyrexia (72%) and chills (61%), which were usually mild and resolved within 48 hours. Peripheral proinflammatory cytokines and lymphocyte activation were induced posttreatment with transient mesothelin-specific T-cell responses in 10 of 13 biomarker-evaluable patients. With monotherapy, four of 18 response-evaluable patients had stable disease of 16 or more weeks, including one patient with a reduction in target lesions. In the combination study, 12 patients were enrolled (median age 63.5 y; women 33%). The most common AEs with combination therapy were pyrexia (67%) and chills (58%); six patients had grade 3 AEs or greater, including two cases of treatment-related fatal pneumonitis. The best overall response for the combination was stable disease in four of nine response-evaluable patients. Conclusions As monotherapy, JNJ-757 was immunogenic and tolerable, with mild infusion-related fever and chills. The limited efficacy of JNJ-757, alone or with nivolumab, did not warrant further investigation of the combination.
Collapse
|
75
|
Tang Y, Zhang AXJ, Chen G, Wu Y, Gu W. Prognostic and therapeutic TILs of cervical cancer-Current advances and future perspectives. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:410-430. [PMID: 34553029 PMCID: PMC8430272 DOI: 10.1016/j.omto.2021.07.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cervical cancer is a top lethal cancer for women worldwide. Although screening and vaccination programs are available in many countries, resulting in the decline of new cases, this is not true for developing countries where there are many new cases and related deaths. Cancer immunotherapy through adaptive cell therapy (ACT) has been applied in clinics, but now much attention is focused on autogenic tumor-infiltrating lymphocyte (TIL)-based therapy, which has shown more specificity and better ability to inhibit tumor growth. Data from melanoma and cervical cancers confirm that tumor-specific T cells in TILs can be expanded for more specific and effective ACT. Moreover, TILs are derived from individual patients and are ready to home back to kill tumor cells after patient infusion, aligning well with personalized and precision medicine. In addition to therapy, TIL cell types and numbers are good indicators of host immune response to the tumor, and thus they have significant values in prognosis. Because of the special relationship with human papillomavirus (HPV) infection, cervical cancer has some specialties in TIL-based prognosis and therapy. In this review, we summarize the recent advances in the prognostic significance of TILs and TIL-based therapy for cervical cancer and discuss related perspectives.
Collapse
Affiliation(s)
- Ying Tang
- Institute of Tumor, Guangzhou University of Chinese Medicine, Guangzhou, China.,Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, China
| | - Anne X J Zhang
- Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, China
| | - Guangyu Chen
- Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, China
| | - Yanheng Wu
- Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, China
| | - Wenyi Gu
- Gillion ITM Research Institute, Guangzhou Hongkeyuan, Guangzhou, China.,Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
76
|
Wang R, Guo H, Tang X, Zhang T, Liu Y, Zhang C, Yu H, Li Y. Interferon Gamma-Induced Interferon Regulatory Factor 1 Activates Transcription of HHLA2 and Induces Immune Escape of Hepatocellular Carcinoma Cells. Inflammation 2021; 45:308-330. [PMID: 34536158 DOI: 10.1007/s10753-021-01547-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/23/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
Immunosuppression developed by cancer cells remains a leading cause of treating failure of immunotherapies. This study aimed to explore the function of human endogenous retrovirus-H long terminal repeat-associating 2 (HHLA2), an immune checkpoint molecule from the B7 family, in the immune escape in hepatocellular carcinoma (HCC). Mouse models with primary HCC or with xenograft tumors were established. The portion of tumor-associated macrophages (TAMs) and the level of PD-L1 in the tumor tissues were examined. THP-1 cells were treated with PMA to obtain a macrophage-like phenotype. The PMA-treated THP-1 cells were co-cultured with the HCC cells in Transwell chambers to examine the function of HHLA2 in chemotactic migration and polarization of macrophages. HHLA2 expression was correlated with infiltration of immune cells, especially macrophages, and was linked to poor prognosis of patients with HCC. HHLA2 knockdown reduced incidence rate of primary HCC in mice. It also reduced tumor metastasis, the portion of M2 macrophages, and the expression of PD-L1 in primary and xenograft tumors. In vitro, HHLA2 upregulation increased expression of PD-L1 in HCC cells indirectly by inducing M2 polarization and chemotactic migration of macrophages. Interferon gamma (IFNG) enhanced expression of interferon regulatory factor 1 (IFR1) in HCC cells, and IFR1 bound to the promoter region of HHLA2 to activate HHLA2 expression. This study suggested that the IFNG/IFR1/HHLA2 axis in HCC induces M2 polarization and chemotactic migration of macrophages, which leads to immune escape and development of HCC.
Collapse
Affiliation(s)
- Rui Wang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Hui Guo
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Xiaotong Tang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Tiantian Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Yang Liu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Cheng Zhang
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Anhui Province Key Laboratory of Translational Cancer Research Affiliated to Bengbu Medical University, Bengbu, 233004, Anhui, People's Republic of China
| | - Hanbing Yu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Yumei Li
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.
| |
Collapse
|
77
|
Dhar R, Seethy A, Singh S, Pethusamy K, Srivastava T, Talukdar J, Rath GK, Karmakar S. Cancer immunotherapy: Recent advances and challenges. J Cancer Res Ther 2021; 17:834-844. [PMID: 34528529 DOI: 10.4103/jcrt.jcrt_1241_20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Immunotherapy is a treatment that uses specific components of a person's immune system to fight diseases. This is usually done by stimulating or assisting one's immune system is attacking the offending agent - for instance, in the case of cancer - the target of immunotherapy will be cancer cells. Some types of immunotherapy are also called biologic therapy or biotherapy. One of the fundamental challenges that a living cell encounters are to accurately copy its genetic material to daughter cells during every single cell cycle. When this process goes haywire, genomic instability ensues, and genetic alterations ranging from nucleotide changes to chromosomal translocations and aneuploidy occur. Genomic instability arising out of DNA structural changes (indels, rearrangements, etc.,) can give rise to mutations predisposing to cancer. Cancer prevention refers to actions taken to mitigate the risk of getting cancer. The past decade has encountered an explosive rate of development of anticancer therapy ranging from standard chemotherapy to novel targeted small molecules that are nearly cancer specific, thereby reducing collateral damage. However, a new class of emerging therapy aims to train the body's defense system to fight against cancer. Termed as "cancer immunotherapy" is the new approach that has gained worldwide acceptance. It includes using antibodies that bind to and inhibit the function of proteins expressed by cancer cells or engineering and boosting the person's own T lymphocytes to target cancer. In this review, we summarized the recent advances and developments in cancer immunotherapy along with their shortcoming and challenges.
Collapse
Affiliation(s)
- Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ashikh Seethy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Tryambak Srivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Joyeeta Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Goura Kishor Rath
- Department of Radiation Oncology, DRBRAIRCH, All India Institute of Medical Sciences, New Delhi; Department of Radiation Oncology, NCI, All India Institute of Medical Sciences, Jhajjar, Haryana, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
78
|
Marshall LA, Marubayashi S, Jorapur A, Jacobson S, Zibinsky M, Robles O, Hu DX, Jackson JJ, Pookot D, Sanchez J, Brovarney M, Wadsworth A, Chian D, Wustrow D, Kassner PD, Cutler G, Wong B, Brockstedt DG, Talay O. Tumors establish resistance to immunotherapy by regulating T reg recruitment via CCR4. J Immunother Cancer 2021; 8:jitc-2020-000764. [PMID: 33243932 PMCID: PMC7692993 DOI: 10.1136/jitc-2020-000764] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Checkpoint inhibitors (CPIs) such as anti-PD(L)-1 and anti-CTLA-4 antibodies have resulted in unprecedented rates of antitumor responses and extension of survival of patients with a variety of cancers. But some patients fail to respond or initially respond but later relapse as they develop resistance to immune therapy. One of the tumor-extrinsic mechanisms for resistance to immune therapy is the accumulation of regulatory T cells (Treg) in tumors. In preclinical and clinical studies, it has been suggested that tumor trafficking of Treg is mediated by CC chemokine receptor 4 (CCR4). Over 90% of human Treg express CCR4 and migrate toward CCL17 and CCL22, two major CCR4 ligands that are either high at baseline or upregulated in tumors on CPI treatment. Hence, CCR4 antagonism has the potential to be an effective antitumor treatment by reducing the accumulation of Treg into the tumor microenvironment (TME). METHODS We developed in vitro and in vivo models to assess Treg migration and antitumor efficacy using a potent and selective CCR4 antagonist, CCR4-351. We used two separate tumor models, Pan02 and CT26 mouse tumors, that have high and low CCR4 ligand expression, respectively. Tumor growth inhibition as well as the frequency of tumor-infiltrating Treg and effector T cells was assessed following the treatment with CCR4 antagonist alone or in combination with CPI. RESULTS Using a selective and highly potent, novel small molecule inhibitor of CCR4, we demonstrate that migration of CCR4+ Treg into the tumor drives tumor progression and resistance to CPI treatment. In tumor models with high baseline levels of CCR4 ligands, blockade of CCR4 reduced the number of Treg and enhanced antitumor immune activity. Notably, in tumor models with low baseline level of CCR4 ligands, treatment with immune CPIs resulted in significant increases of CCR4 ligands and Treg numbers. Inhibition of CCR4 reduced Treg frequency and potentiated the antitumor effects of CPIs. CONCLUSION Taken together, we demonstrate that CCR4-dependent Treg recruitment into the tumor is an important tumor-extrinsic mechanism for immune resistance. Blockade of CCR4 led to reduced frequency of Treg and resulted in increased antitumor activity, supporting the clinical development of CCR4 inhibitors in combination with CPI for the treatment of cancer. STATEMENT OF SIGNIFICANCE CPI upregulates CCL17 and CCL22 expression in tumors and increases Treg migration into the TME. Pharmacological antagonism of the CCR4 receptor effectively inhibits Treg recruitment and results in enhanced antitumor efficacy either as single agent in CCR4 ligandhigh tumors or in combination with CPIs in CCR4 ligandlow tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Omar Robles
- RAPT Therapeutics, South San Francisco, California, USA
| | | | | | - Deepa Pookot
- RAPT Therapeutics, South San Francisco, California, USA
| | | | | | | | - David Chian
- Lyell Immunopharma, South San Francisco, California, USA
| | - David Wustrow
- RAPT Therapeutics, South San Francisco, California, USA
| | | | - Gene Cutler
- RAPT Therapeutics, South San Francisco, California, USA
| | - Brian Wong
- RAPT Therapeutics, South San Francisco, California, USA
| | | | - Oezcan Talay
- RAPT Therapeutics, South San Francisco, California, USA
| |
Collapse
|
79
|
Loilome W, Dokduang H, Suksawat M, Padthaisong S. Therapeutic challenges at the preclinical level for targeted drug development for Opisthorchis viverrini-associated cholangiocarcinoma. Expert Opin Investig Drugs 2021; 30:985-1006. [PMID: 34292795 DOI: 10.1080/13543784.2021.1955102] [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: 10/20/2022]
Abstract
INTRODUCTION Cholangiocarcinoma (CCA) is a malignant tumor of bile duct epithelium with the highest incidence found in Thailand. Some patients are considered suitable for adjuvant therapy and surgical resection is currently the curative treatment for CCA patients. Tumor recurrence is still a hurdle after treatment; hence, finding novel therapeutic strategies to combat CCA is necessary for improving outcome for patients. AREAS COVERED We discuss targeted therapies and other novel treatment approaches which include protein kinase inhibitors, natural products, amino acid transporter-based inhibitors, immunotherapy, and drug repurposing. We also examine the challenges of tumor heterogeneity, cancer stem cells (CSCs), the tumor microenvironment, exosomes, multiomics studies, and the potential of precision medicine. EXPERT OPINION Because CCA is difficult to diagnose at the early stage, the traditional treatment approaches are not effective for many patients and most tumors recur. Consequently, researchers are exploring multi-aspect molecular carcinogenesis to uncover molecular targets for further development of novel targeted drugs.
Collapse
Affiliation(s)
- Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hasaya Dokduang
- Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Manida Suksawat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sureerat Padthaisong
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen Thailand.,Cholangiocarcinoma Screening and Care Program (CASCAP), Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
80
|
Zhao HB, Zeng YR, Han ZD, Zhuo YJ, Liang YK, Hon CT, Wan S, Wu S, Dahl D, Zhong WD, Wu CL. Novel immune-related signature for risk stratification and prognosis in prostatic adenocarcinoma. Cancer Sci 2021; 112:4365-4376. [PMID: 34252262 PMCID: PMC8486177 DOI: 10.1111/cas.15062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 06/16/2021] [Accepted: 07/06/2021] [Indexed: 12/14/2022] Open
Abstract
A substantial proportion of prostatic adenocarcinoma (PRAD) patients experience biochemical failure (BCF) after radical prostatectomy (RP). The immune microenvironment plays a vital role in carcinogenesis and the development of PRAD. This study aimed to identify a novel immune-related gene (IRG)-based signature for risk stratification and prognosis of BCF in PRAD. Weighted gene coexpression network analysis was carried out to identify a BCF-related module in a discovery cohort of patients who underwent RP at the Massachusetts General Hospital. The median follow-up time was 70.32 months. Random forest and multivariate stepwise Cox regression analyses were used to identify an IRG-based signature from the specific module. Risk plot analyses, Kaplan-Meier curves, receiver operating characteristic curves, univariate and multivariate Cox regression analyses, stratified analysis, and Harrell's concordance index were used to assess the prognostic value and predictive accuracy of the IRG-based signature in the internal discovery cohort; The Cancer Genome Atlas database was used as a validation cohort. Tumor immune estimation resource database analysis and CIBERSORT algorithm were used to assess the immunophenotype of PRAD. A novel IRG-based signature was identified from the specific module. Five IRGs (BUB1B, NDN, NID1, COL4A6, and FLRT2) were verified as components of the risk signature. The IRG-based signature showed good prognostic value and predictive accuracy in both the discovery and validation cohorts. Infiltrations of various immune cells were significantly different between low-risk and high-risk groups in PRAD. We identified a novel IRG-based signature that could function as an index for assessing tumor immune status and risk stratification in PRAD.
Collapse
Affiliation(s)
- Hai-Bo Zhao
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yan-Ru Zeng
- Department of Anesthesiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Zhao-Dong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Yang-Jia Zhuo
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Ying-Ke Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China
| | - Chi Tin Hon
- Macau Institute of Systems Engineering, Macau University of Science and Technology, Macau, China
| | - Song Wan
- Department of Urology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Shulin Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Douglas Dahl
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wei-De Zhong
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.,Department of Urology, The Second Affiliated Hospital, South China University of Technology, Guangzhou, China.,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
81
|
Ghouzlani A, Kandoussi S, Tall M, Reddy KP, Rafii S, Badou A. Immune Checkpoint Inhibitors in Human Glioma Microenvironment. Front Immunol 2021; 12:679425. [PMID: 34305910 PMCID: PMC8301219 DOI: 10.3389/fimmu.2021.679425] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Gliomas are the most common primary brain tumors in adults. Despite the fact that they are relatively rare, they cause significant morbidity and mortality. High-grade gliomas or glioblastomas are rapidly progressing tumors with a very poor prognosis. The presence of an intrinsic immune system in the central nervous system is now more accepted. During the last decade, there has been no major progress in glioma therapy. The lack of effective treatment for gliomas can be explained by the strategies that cancer cells use to escape the immune system. This being said, immunotherapy, which involves blockade of immune checkpoint inhibitors, has improved patients' survival in different cancer types. This novel cancer therapy appears to be one of the most promising approaches. In the present study, we will start with a review of the general concept of immune response within the brain and glioma microenvironment. Then, we will try to decipher the role of various immune checkpoint inhibitors within the glioma microenvironment. Finally, we will discuss some promising therapeutic pathways, including immune checkpoint blockade and the body's effective anti-glioma immune response.
Collapse
Affiliation(s)
- Amina Ghouzlani
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Sarah Kandoussi
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mariam Tall
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Konala Priyanka Reddy
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Faculty of Medicine, Medical University of Pleven, Pleven, Bulgaria
| | - Soumaya Rafii
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| |
Collapse
|
82
|
Afolabi LO, Afolabi MO, Sani MM, Okunowo WO, Yan D, Chen L, Zhang Y, Wan X. Exploiting the CRISPR-Cas9 gene-editing system for human cancers and immunotherapy. Clin Transl Immunology 2021; 10:e1286. [PMID: 34188916 PMCID: PMC8219901 DOI: 10.1002/cti2.1286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/23/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
The discovery of clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9 (CRISPR-Cas9) technology has brought advances in the genetic manipulation of eukaryotic cells, which has revolutionised cancer research and treatment options. It is increasingly being used in cancer immunotherapy, including adoptive T and natural killer (NK) cell transfer, secretion of antibodies, cytokine stimulation and overcoming immune checkpoints. CRISPR-Cas9 technology is used in autologous T cells and NK cells to express various innovative antigen designs and combinations of chimeric antigen receptors (CARs) targeted at specific antigens for haematological and solid tumors. Additionally, advanced engineering in immune cells to enhance their sensing circuits with sophisticated functionality is now possible. Intensive research on the CRISPR-Cas9 system has provided scientists with the ability to overcome the hostile tumor microenvironment and generate more products for future clinical use, especially off-the-shelf, universal cellular products, bringing exciting milestones for immunotherapy. This review discussed the application and challenges of CRISPR technology in cancer research and immunotherapy, its advances and prospects for promoting new cell-based therapeutic beyond immune oncology.
Collapse
Affiliation(s)
- Lukman O Afolabi
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
- Department of BiochemistryFaculty of ScienceFederal University DutseDutseNigeria
| | - Mariam O Afolabi
- Open FIESTA CenterTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical OncogenomicsGraduate School at ShenzhenTsinghua UniversityShenzhenChina
| | - Musbahu M Sani
- Department of BiochemistryFaculty of ScienceFederal University DutseDutseNigeria
| | - Wahab O Okunowo
- Department of BiochemistryCollege of MedicineUniversity of LagosLagosNigeria
| | - Dehong Yan
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Liang Chen
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yaou Zhang
- Open FIESTA CenterTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical OncogenomicsGraduate School at ShenzhenTsinghua UniversityShenzhenChina
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Xiaochun Wan
- Guangdong Immune Cell therapy Engineering and Technology research CenterCenter for Protein and Cell‐based DrugsInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
83
|
Kern R, Panis C. CTLA-4 Expression and Its Clinical Significance in Breast Cancer. Arch Immunol Ther Exp (Warsz) 2021; 69:16. [PMID: 34148159 DOI: 10.1007/s00005-021-00618-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer is the leading cause of women's death among all cancers. The main reason associated with this is the development of metastasis and therapy-resistant breast carcinoma (BC), which pose the main challenge of oncology nowadays. Evidence suggest that these tumors seem to have inhibitory mechanisms that may favor their progression and surveillance. Cancer cells can evade antitumor T cell responses by expressing some immune inhibitory molecules such as the cytotoxic T-lymphocyte antigen-4 (CTLA-4), whose clinical meaning has emerged in the last few years and is poorly understood in the BC context. This systematic literature review aims at identifying studies on CTLA-4 expression in BC, and address what is known about its clinical meaning. A literature search was performed in PubMed and LILACS databases, using the MESH terms "breast cancer"; "CTLA-4 Antigen/antagonists and inhibitors"; and "Lymphocytes, Tumor-Infiltrating/immunology", published in the last 10 years. In total, 12 studies were included in this review. Systematic review used the Preferred Reporting Items for Systematic Reviews and Meta-Analyses. Despite the small number of eligible studies, the literature reports some associations between CTLA-4 expression in the tumor microenvironment and worse BC outcomes, regardless of its molecular subtype. CTLA-4 expression in BC is a putative marker of clinical significance and a rationale therapeutic target in the emerging field of immunotherapy.
Collapse
Affiliation(s)
- Rodrigo Kern
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil
| | - Carolina Panis
- Laboratory of Tumor Biology, State University of West Paraná, UNIOESTE, Francisco Beltrão, Brazil.
- State University of Western Paraná, Health Sciences Center, Vitório Traiano Highway, Km 2, Francisco Beltrão, PR, Brazil.
| |
Collapse
|
84
|
George LL, Deshpande SR, Cortese MJ, Kendall EK, Chattaraj A, Shah Z, Zhao J, Anwer F. Emerging Targets and Cellular Therapy for Relapsed Refractory Multiple Myeloma: A Systematic Review. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:741-751. [PMID: 34253497 DOI: 10.1016/j.clml.2021.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022]
Abstract
Multiple myeloma is the second most common hematologic malignancy and remains incurable. Patients who fail multiple lines of therapy typically have a poor prognosis despite recent advances in myeloma treatment. Chimeric antigen receptor T (CAR T) cell treatment has emerged as a promising therapy for many hematologic malignancies, including recently approved and emerging applications for myeloma treatment. A systematic review of the available clinical trial data for CAR T therapies in multiple myeloma was undertaken. All multiple myeloma trials registered at ClinicalTrials.gov were reviewed and studies mentioning CAR T and studying relapsed/refractory multiple myeloma (R/R MM) were included. PubMed, Google Scholar, and conference proceedings were also reviewed to determine which trials had reported data. Twenty-seven registered clinical trials in humans with published data were identified as of March 10, 2021. The majority of these trials were CAR T cells targeting B-cell maturation antigen (BCMA), and many were Phase I studies. Data demonstrated promising short-term (<12 months) efficacy with low incidence of grade 3 or higher toxicities. CAR T cell therapy in R/R MM remains a promising treatment modality. While one biologic has recently received FDA-approval, the majority of products remain investigational and in early-phase trials. More investigation is needed to determine which CAR T constructs and combination therapies optimize patient outcomes.
Collapse
Affiliation(s)
- Laeth L George
- Department of Internal Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH
| | | | - Matthew J Cortese
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Ellen K Kendall
- School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Asmi Chattaraj
- Department of Internal Medicine, University of Pittsburgh Medical Center McKeesport, McKeesport, PA
| | - Zunairah Shah
- Department of Internal Medicine, Weiss Memorial Hospital, Chicago, IL
| | - Jianjun Zhao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Faiz Anwer
- Taussig Cancer Center Hematology, Oncology, Stem Cell Transplantation Multiple Myeloma Program, Cleveland, OH.
| |
Collapse
|
85
|
Yajuk O, Baron M, Toker S, Zelter T, Fainsod-Levi T, Granot Z. The PD-L1/PD-1 Axis Blocks Neutrophil Cytotoxicity in Cancer. Cells 2021; 10:cells10061510. [PMID: 34203915 PMCID: PMC8232689 DOI: 10.3390/cells10061510] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/22/2022] Open
Abstract
The PD-L1/PD-1 axis mediates immune tolerance and promotes tumor growth and progression via the inhibition of anti-tumor immunity. Blocking the interaction between PD-L1 and PD-1 was clinically shown to be beneficial in maintaining the anti-tumor functions of the adaptive immune system. Still, the consequences of blocking the PD-L1/PD-1 axis on innate immune responses remain largely unexplored. In this context, neutrophils were shown to consist of distinct subpopulations, which possess either pro- or anti-tumor properties. PD-L1-expressing neutrophils are considered pro-tumor as they are able to suppress cytotoxic T cells and are propagated with disease progression. That said, we found that PD-L1 expression is not limited to tumor promoting neutrophils, but is also evident in anti-tumor neutrophils. We show that neutrophil cytotoxicity is effectively and efficiently blocked by tumor cell-expressed PD-1. Furthermore, the blocking of either neutrophil PD-L1 or tumor cell PD-1 maintains neutrophil cytotoxicity. Importantly, we show that tumor cell PD-1 blocks neutrophil cytotoxicity and promotes tumor growth via a mechanism independent of adaptive immunity. Taken together, these findings highlight the therapeutic potential of enhancing anti-tumor innate immune responses via blocking of the PD-L1/PD-1 axis.
Collapse
|
86
|
Maiorano BA, Schinzari G, Ciardiello D, Rodriquenz MG, Cisternino A, Tortora G, Maiello E. Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities. Vaccines (Basel) 2021; 9:623. [PMID: 34207536 PMCID: PMC8228524 DOI: 10.3390/vaccines9060623] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In the last years, many new treatment options have widened the therapeutic scenario of genitourinary malignancies. Immunotherapy has shown efficacy, especially in the urothelial and renal cell carcinomas, with no particular relevance in prostate cancer. However, despite the use of immune checkpoint inhibitors, there is still high morbidity and mortality among these neoplasms. Cancer vaccines represent another way to activate the immune system. We sought to summarize the most recent advances in vaccine therapy for genitourinary malignancies with this review. METHODS We searched PubMed, Embase and Cochrane Database for clinical trials conducted in the last ten years, focusing on cancer vaccines in the prostate, urothelial and renal cancer. RESULTS Various therapeutic vaccines, including DNA-based, RNA-based, peptide-based, dendritic cells, viral vectors and modified tumor cells, have been demonstrated to induce specific immune responses in a variable percentage of patients. However, these responses rarely corresponded to significant survival improvements. CONCLUSIONS Further preclinical and clinical studies will improve the knowledge about cancer vaccines in genitourinary malignancies to optimize dosage, select targets with a driver role for tumor development and growth, and finally overcome resistance mechanisms. Combination strategies represent possibly more effective and long-lasting treatments.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
| | - Giovanni Schinzari
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Davide Ciardiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Maria Grazia Rodriquenz
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| | - Antonio Cisternino
- Urology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy;
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| |
Collapse
|
87
|
Creemers JHA, Lesterhuis WJ, Mehra N, Gerritsen WR, Figdor CG, de Vries IJM, Textor J. A tipping point in cancer-immune dynamics leads to divergent immunotherapy responses and hampers biomarker discovery. J Immunother Cancer 2021; 9:jitc-2020-002032. [PMID: 34059522 PMCID: PMC8169479 DOI: 10.1136/jitc-2020-002032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Predicting treatment response or survival of cancer patients remains challenging in immuno-oncology. Efforts to overcome these challenges focus, among others, on the discovery of new biomarkers. Despite advances in cellular and molecular approaches, only a limited number of candidate biomarkers eventually enter clinical practice. METHODS A computational modeling approach based on ordinary differential equations was used to simulate the fundamental mechanisms that dictate tumor-immune dynamics and to investigate its implications on responses to immune checkpoint inhibition (ICI) and patient survival. Using in silico biomarker discovery trials, we revealed fundamental principles that explain the diverging success rates of biomarker discovery programs. RESULTS Our model shows that a tipping point-a sharp state transition between immune control and immune evasion-induces a strongly non-linear relationship between patient survival and both immunological and tumor-related parameters. In patients close to the tipping point, ICI therapy may lead to long-lasting survival benefits, whereas patients far from the tipping point may fail to benefit from these potent treatments. CONCLUSION These findings have two important implications for clinical oncology. First, the apparent conundrum that ICI induces substantial benefits in some patients yet completely fails in others could be, to a large extent, explained by the presence of a tipping point. Second, predictive biomarkers for immunotherapy should ideally combine both immunological and tumor-related markers, as a patient's distance from the tipping point can typically not be reliably determined from solely one of these. The notion of a tipping point in cancer-immune dynamics helps to devise more accurate strategies to select appropriate treatments for patients with cancer.
Collapse
Affiliation(s)
- Jeroen H A Creemers
- Department of Tumor Immunology, Radboudumc, Nijmegen, The Netherlands.,Oncode Institute, Nijmegen, The Netherlands
| | - W Joost Lesterhuis
- School of Biomedical Sciences and Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Niven Mehra
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | | | - Carl G Figdor
- Department of Tumor Immunology, Radboudumc, Nijmegen, The Netherlands.,Oncode Institute, Nijmegen, The Netherlands
| | | | - Johannes Textor
- Department of Tumor Immunology, Radboudumc, Nijmegen, The Netherlands .,Data Science Department, Radboud University Institute for Computing and Information Sciences, Nijmegen, The Netherlands
| |
Collapse
|
88
|
Ruan H, Bao L, Tao Z, Chen K. Flightless I Homolog Reverses Enzalutamide Resistance through PD-L1-Mediated Immune Evasion in Prostate Cancer. Cancer Immunol Res 2021; 9:838-852. [PMID: 34011528 DOI: 10.1158/2326-6066.cir-20-0729] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 03/06/2021] [Accepted: 05/04/2021] [Indexed: 12/24/2022]
Abstract
Tumor cells can evade immune surveillance and immune killing during the emergence of endocrine therapy resistance in prostate cancer, but the mechanisms underlying this phenomenon are still unclear. Flightless I homolog (FLII) is a coregulator for transcription factors in several malignancies. Here, we have demonstrated that endocrine therapy resistance can induce an immunosuppressive prostate tumor microenvironment and immune evasion through FLII downregulation, which leads to activation of the YBX1/PD-L1 signaling pathway. FLII expression negatively correlated with expression of PD-L1 in tumors. Mechanism studies demonstrated that FLII physically interacted with YBX1 to inhibit nuclear localization of YBX1 and thereby suppress transcription of PDL1 in enzalutamide-resistant tumors. Restoration of FLII expression reversed enzalutamide resistance through activation of T-cell responses in the tumor microenvironment through inhibition of the YBX1/PD-L1 pathway. We also found that reversal of endocrine therapy resistance and immune evasion was mediated by proliferation of effector CD8+ T cells and inhibition of tumor infiltration by regulatory T cells and myeloid-derived suppressor cells. Taken together, our results demonstrate a functional and biological interaction between endocrine therapy resistance and immune evasion mediated through the FLII/YBX1/PD-L1 cascade. Combination therapy with FLII expression and endocrine therapy may benefit patients with prostate cancer by preventing tumor immune evasion.
Collapse
Affiliation(s)
- Hailong Ruan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Bao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Tao
- Department of Radiation Oncology and Cyberknife Center, Tianjin Medical University Cancer institute & Hospital, Tianjin, China.
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| |
Collapse
|
89
|
Lee MA. Current status of cancer immunotherapy with immune checkpoint inhibitors. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2021. [DOI: 10.5124/jkma.2021.64.5.326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The clinical outcome in advanced cancer has improved since the development of targeted therapies and immune checkpoint inhibitors. We can expect better overall survival after a combination treatment with these therapeutic agents. Classical cytotoxic chemotherapeutic agents directly kill tumor cells by destroying the cell structure and DNA of tumor cells or inhibiting their metabolism. Targeted therapy also directly affects tumor cells by inhibiting the cell growth signaling system. Conversely, immune checkpoint inhibitors can enhance immune responses by using the host immune system in the tumor microenvironment, leading to the direct killing of tumor cells. Therefore, immune checkpoint inhibitors are less toxic and have longer response durations. Even in solid tumors with nonimmunogenic backgrounds, cytotoxic chemotherapy or targeted treatment can induce immune responses to place the tumor microenvironment in an immunogenic state. Synergistic anticancer effects can be expected when immune checkpoint inhibitors are added during this state. Moreover, drug resistance may be overcome by combination therapies. If patients with cancer are treated with a combination of these therapeutic agents and the characteristics of each tumor are identified with data from next generation sequencing, personalized treatments can be tailored, making it possible to control cancers as a curable disease just like other benign chronic diseases.
Collapse
|
90
|
Chatzidionysiou K, Liapi M, Tsakonas G, Gunnarsson I, Catrina A. Treatment of rheumatic immune-related adverse events due to cancer immunotherapy with immune checkpoint inhibitors-is it time for a paradigm shift? Clin Rheumatol 2021; 40:1687-1695. [PMID: 32989505 PMCID: PMC8102438 DOI: 10.1007/s10067-020-05420-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/19/2022]
Abstract
Immunotherapy has revolutionized cancer treatment during the last years. Several monoclonal antibodies that are specific for regulatory checkpoint molecules, that is, immune checkpoint inhibitors (ICIs), have been approved and are currently in use for various types of cancer in different lines of treatment. Cancer immunotherapy aims for enhancing the immune response against cancer cells. Despite their high efficacy, ICIs are associated to a new spectrum of adverse events of autoimmune origin, often referred to as immune-related adverse events (irAEs), which limit the utility of these drugs. These irAEs are quite common and can affect almost every organ. The grade of toxicity varies from very mild to life-threatening. The pathophysiological mechanisms behind these events are not fully understood. In this review, we will summarize current evidence specifically regarding the rheumatic irAEs and we will focus on current and future treatment strategies. Treatment guidelines largely support the use of glucocorticoids as first-line therapy, when symptomatic therapy is not efficient, and for more persistent and/or moderate/severe degree of inflammation. Targeted therapies are higher up in the treatment pyramid, after inadequate response to glucocorticoids and conventional, broad immunosuppressive agents, and for severe forms of irAEs. However, preclinical data provide evidence that raise concerns regarding the potential risk of impaired antitumoral effect. This potential risk of glucocorticoids, together with the high efficacy and potential synergistic effect of newer, targeted immunomodulation, such as tumor necrosis factor and interleukin-6 blockade, could support a paradigm shift, where more targeted treatments are considered earlier in the treatment sequence.
Collapse
Affiliation(s)
- Katerina Chatzidionysiou
- Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden.
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden.
| | - Matina Liapi
- Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Georgios Tsakonas
- Thoracic Oncology Center, Karolinska University Hospital, Stockholm, Sweden
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Iva Gunnarsson
- Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Anca Catrina
- Rheumatology Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
91
|
Rosenthal R, Swanton C, McGranahan N. Understanding the impact of immune-mediated selection on lung cancer evolution. Br J Cancer 2021; 124:1615-1617. [PMID: 33623078 PMCID: PMC8110793 DOI: 10.1038/s41416-020-01232-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/20/2020] [Accepted: 12/08/2020] [Indexed: 01/31/2023] Open
Abstract
Understanding how a tumour evolves and avoids immune recognition is paramount to improving cancer immunotherapy and patient outcome. Here we examine our recent integration of multi-region genomic, transcriptomic, epigenomic, pathology, and clinical data, highlight the need for a systematic examination of immune escape mechanisms, and discuss implications for immunotherapy approaches.
Collapse
Affiliation(s)
- Rachel Rosenthal
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, University College London Cancer Institute, University College London, London, UK
| |
Collapse
|
92
|
Olivares-Hernández A, Figuero-Pérez L, Terán-Brage E, López-Gutiérrez Á, Velasco ÁT, Sarmiento RG, Cruz-Hernández JJ, Miramontes-González JP. Resistance to Immune Checkpoint Inhibitors Secondary to Myeloid-Derived Suppressor Cells: A New Therapeutic Targeting of Haematological Malignancies. J Clin Med 2021; 10:jcm10091919. [PMID: 33925214 PMCID: PMC8124332 DOI: 10.3390/jcm10091919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/17/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a set of immature myeloid lineage cells that include macrophages, granulocytes, and dendritic cell precursors. This subpopulation has been described in relation to the tumour processes at different levels, including resistance to immunotherapy, such as immune checkpoint inhibitors (ICIs). Currently, multiple studies at the preclinical and clinical levels seek to use this cell population for the treatment of different haematological neoplasms, together with ICIs. This review addresses the different points in ongoing studies of MDSCs and ICIs in haematological malignancies and their future significance in routine clinical practice.
Collapse
Affiliation(s)
- Alejandro Olivares-Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Correspondence: (A.O.-H.); (J.P.M.-G.); Tel.: +34-923-29-11-00 (A.O.-H.); +34-983-42-04-00 (J.P.M.-G.); Fax: +34-923-29-13-25 (A.O.-H.); +34-983-21-53-65 (J.P.M.-G.)
| | - Luis Figuero-Pérez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Eduardo Terán-Brage
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Álvaro López-Gutiérrez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
| | - Álvaro Tamayo Velasco
- Department of Haematology, University Hospital of Valladolid, 47003 Valladolid, Spain;
| | - Rogelio González Sarmiento
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Juan Jesús Cruz-Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.F.-P.); (E.T.-B.); (Á.L.-G.); (J.J.C.-H.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - José Pablo Miramontes-González
- Department of Internal Medicine, University Hospital Rio Hortega, 47012 Valladolid, Spain
- Department of Medicine, University of Valladolid, 45005 Valladolid, Spain
- Correspondence: (A.O.-H.); (J.P.M.-G.); Tel.: +34-923-29-11-00 (A.O.-H.); +34-983-42-04-00 (J.P.M.-G.); Fax: +34-923-29-13-25 (A.O.-H.); +34-983-21-53-65 (J.P.M.-G.)
| |
Collapse
|
93
|
Hammink R, Weiden J, Voerman D, Popelier C, Eggermont LJ, Schluck M, Figdor CG, Verdoes M. Semiflexible Immunobrushes Induce Enhanced T Cell Activation and Expansion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:16007-16018. [PMID: 33797875 PMCID: PMC8045021 DOI: 10.1021/acsami.0c21994] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
A variety of bioactive materials developed to expand T cells for adoptive transfer into cancer patients are currently evaluated in the clinic. In most cases, T cell activating biomolecules are attached to rigid surfaces or matrices and form a static interface between materials and the signaling receptors on the T cells. We hypothesized that a T cell activating polymer brush interface might better mimic the cell surface of a natural antigen-presenting cell, facilitating receptor movement and concomitant advantageous mechanical forces to provide enhanced T cell activating capacities. Here, as a proof of concept, we synthesized semiflexible polyisocyanopeptide (PIC) polymer-based immunobrushes equipped with T cell activating agonistic anti-CD3 (αCD3) and αCD28 antibodies placed on magnetic microbeads. We demonstrated enhanced efficiency of ex vivo expansion of activated primary human T cells even at very low numbers of stimulating antibodies compared to rigid beads. Importantly, the immunobrush architecture appeared crucial for this improved T cell activating capacity. Immunobrushes outperform current benchmarks by producing higher numbers of T cells exhibiting a combination of beneficial phenotypic characteristics, such as reduced exhaustion marker expression, high cytokine production, and robust expression of cytotoxic hallmarks. This study indicates that semiflexible immunobrushes have great potential in making T cell-based immunotherapies more effective.
Collapse
Affiliation(s)
- Roel Hammink
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
| | - Jorieke Weiden
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Dion Voerman
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carlijn Popelier
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
| | - Loek J. Eggermont
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Marjolein Schluck
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Carl G. Figdor
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Division
of Immunotherapy, Oncode Institute, Radboud
University Medical Center, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| | - Martijn Verdoes
- Department
of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 26, 6525 GA Nijmegen, Netherlands
- Institute
for Chemical Immunology, 6525 GA Nijmegen, Netherlands
| |
Collapse
|
94
|
Albrecht T, Brinkmann F, Albrecht M, Lonsdorf AS, Mehrabi A, Hoffmann K, Kulu Y, Charbel A, Vogel MN, Rupp C, Köhler B, Springfeld C, Schirmacher P, Roessler S, Goeppert B. Programmed Death Ligand-1 (PD-L1) Is an Independent Negative Prognosticator in Western-World Gallbladder Cancer. Cancers (Basel) 2021; 13:1682. [PMID: 33918309 PMCID: PMC8038183 DOI: 10.3390/cancers13071682] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
Inhibition of the programmed cell death protein-1/ligand-1 (PD-1/PD-L1) axis has opened a new era in the treatment of solid cancers. However, there is no data on the expression and relevance of PD-L1 in Western gallbladder cancer (GBC). We assessed PD-L1 immunohistochemically in 131 GBC patients as Tumor Proportion Score (TPS), Immune Cell Score (IC) and Combined Positivity Score (CPS). Tumor cells expressed PD-L1 in a subset of 14.7% GBC patients at a TPS cut-off of 1%. Higher PD-L1 levels above 10% and 25% TPS were reached in 4.7% and 3.1% of GBC cases, respectively. At a 10% cut-off, TPS was associated with distinct histomorphological subtypes and correlated with poor tumor differentiation. Survival analysis revealed a TPS above 10% to be a highly significant and independent negative prognosticator in GBC. PD-L1 expression was associated with increased CD4+, CD8+ and PD-1+ immune cell densities. In 14.8% of the cases, scattered immune cells expressed T-cell immunoreceptor with Ig and ITIM domains (TIGIT), which was correlated to tumoral expression of its ligand CD155. We here show that a high PD-L1 expression confers a negative prognostic value in Western-world GBC and highlight the TIGIT/CD155 immune checkpoint as a potential new target for GBC immunotherapy.
Collapse
Affiliation(s)
- Thomas Albrecht
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| | - Fritz Brinkmann
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| | - Michael Albrecht
- European Center for Angioscience (ECAS), Medical Faculty of Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Anke S. Lonsdorf
- Department of Dermatology, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Arianeb Mehrabi
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Katrin Hoffmann
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Yakup Kulu
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany;
| | - Alphonse Charbel
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| | - Monika N. Vogel
- Diagnostic and Interventional Radiology, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany;
| | - Christian Rupp
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
- Department of Internal Medicine IV, Gastroenterology and Hepatology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Bruno Köhler
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Christoph Springfeld
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
- National Center for Tumor Diseases, Department of Medical Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| | - Benjamin Goeppert
- Institute of Pathology, Heidelberg University Hospital, 69120 Heidelberg, Germany; (T.A.); (F.B.); (A.C.); (P.S.); (S.R.)
- Liver Cancer Center Heidelberg (LCCH), 69120 Heidelberg, Germany; (A.M.); (K.H.); (C.R.); (B.K.); (C.S.)
| |
Collapse
|
95
|
Zhang J, An L, Zhou X, Shi R, Wang H. Analysis of tumor mutation burden combined with immune infiltrates in endometrial cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:551. [PMID: 33987249 PMCID: PMC8105813 DOI: 10.21037/atm-20-6049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 01/03/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Tumor mutational burden (TMB) is widely regarded as a predictor of response to immunotherapy. Few researchers have focused on the activity and prognosis of TMB in endometrial cancer (EC) and immune cells. Our study aimed to identify the prognostic role of TMB in EC. METHODS We downloaded transcriptome data from The Cancer Genome Atlas (TCGA) database. Kaplan-Meier analysis with log-rank test was conducted to assess the difference in overall survival (OS) between the high and low TMB groups. The "CIBERSORT" scripts were performed to evaluate the immune compositions of EC patients. Cox regression analysis and survival analysis were used to verify the prognostic value prognosis of TMB. RESULTS We obtained the single nucleotide mutation data for 529 EC patients. A missense mutation was the most common mutation type. TMB was associated with survival outcome, tumor grades, and pathological types. We identified 10 hub TMB-related signature and found that elevated T-cell subsets infiltrating density in the high TMB group revealed improved survival outcomes. According to Kaplan-Meier analysis, T cells gamma delta and T cells regulatory were prognostic immune cells in EC samples. Moreover, many top gene set enrichment analysis (GSEA) results, including amino sugar and nucleotide sugar metabolism, nucleotide excision repair, or p53 signaling pathway, were enriched significantly with TMB level as phenotype. CONCLUSIONS TMB is an important prognostic factor for EC, and TMB-related genes may be potential therapeutic targets for EC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lanfen An
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Shi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
96
|
Rheumatic and musculoskeletal disorders induced by immune checkpoint inhibitors: Consequences on overall survival. Joint Bone Spine 2021; 88:105168. [PMID: 33675979 DOI: 10.1016/j.jbspin.2021.105168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/10/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICIs) frequently induce immune related adverse events (irAEs) that may be associated with more favorable clinical outcomes. We aimed to evaluate the impact of all types of rheumatic adverse events (AEs) on overall survival (OS) and tumor response in patients treated with ICIs. METHODS We performed a single-center retrospective observational study to analyze the OS and tumor response in patients receiving ICIs who experienced a rheumatic AE compared to those who did not experience any AE. RESULTS From December 2010 to September 2018, 264 patients with any cancer type were included. Forty-three patients (16.3%) presented with at least one rheumatic AE. The median OS of patients with rheumatic AEs was significantly higher than that of patients without AEs, with 132 weeks (95% CI [69.3-not reached]) and 42.7 weeks (95% CI [25.6-not reached]), respectively (P<0.01). This result remained significant after multivariate analysis (HR 0.54, 95% CI [0.30-0.97], P<0.05). Also, tumor response was better in patients with rheumatic AEs. CONCLUSION The occurrence of rheumatic AEs in patients treated with ICIs is associated with better survival and tumor response. Therefore, it seems essential to detect rheumatic AEs as early as possible to allow rapid and optimal management, given the long-term response potential of these patients.
Collapse
|
97
|
Luan J, Zhang Q, Song L, Wang Y, Ji C, Cong R, Zheng Q, Xu Z, Xia J, Song N. Identification and validation of a six immune-related gene signature for prediction of biochemical recurrence in localized prostate cancer following radical prostatectomy. Transl Androl Urol 2021; 10:1018-1029. [PMID: 33850736 PMCID: PMC8039594 DOI: 10.21037/tau-20-1231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Prostate cancer (PCa) is the second lethal heterogeneous cancer among males worldwide, and approximately 20% of PCa patients following radical prostatectomy (RP) will undergo biochemical recurrence (BCR). This study is aimed to identify the immune-related gene signature that can predict BCR in localized PCa following RP. Methods Expression profile of genes together with clinical parameters from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus database (GEO) and the immune-related genes from the Molecular Signatures Database v4.0 were applied to construct and validate the gene signature. The Cox regression analyses were conducted to identify the candidate genes and establish the gene signature. To estimate the prognostic power of the risk score, the time-dependent receiver operating characteristic (ROC) analysis and Harrell's index of concordance (C-index) were utilized. We also established a nomogram to forecast the probability of patients' survival. Results A total of 268 patients from the TCGA and 77 patients from GSE70770 and six immune-related genes (SCIN, THY1, TBX1, NOTCH4, MAL, BNIP3L) were eventually selected. The Kaplan-Meier analysis demonstrated that patients in the low-risk group had a significantly longer recurrence-free survival (RFS) compared to those in the high-risk group. In the multivariate Cox model, the signature was identified as an independent prognostic factor, which was significantly associated with RFS (TCGA: HR =5.232, 95% CI: 1.762-15.538, P=0.003; GSE70770: HR =2.158, 95% CI: 1.051-4.432, P=0.036). Moreover, the C-index got improved after incorporating the risk score into original clinicopathological parameters. In addition, the novel nomogram was constructed to better predict the 1-, 3- and 5-year RFS. Conclusions This signature could serve as an independent prognostic factor for BCR. Incorporation of our signature into traditional risk classification might further stratify patients with different prognosis, which could assist practitioners in developing clinical decision-making.
Collapse
Affiliation(s)
- Jiaochen Luan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qijie Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lebin Song
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yichun Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chengjian Ji
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Cong
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qitong Zheng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhenggang Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiadong Xia
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,The Affiliated Kezhou People's Hospital of Nanjing Medical University, Kezhou, China
| |
Collapse
|
98
|
Burvenich IJG, Goh YW, Guo N, Gan HK, Rigopoulos A, Cao D, Liu Z, Ackermann U, Wichmann CW, McDonald AF, Huynh N, O'Keefe GJ, Gong SJ, Scott FE, Li L, Geng W, Zutshi A, Lan Y, Scott AM. Radiolabelling and preclinical characterization of 89Zr-Df-radiolabelled bispecific anti-PD-L1/TGF-βRII fusion protein bintrafusp alfa. Eur J Nucl Med Mol Imaging 2021; 48:3075-3088. [PMID: 33608805 DOI: 10.1007/s00259-021-05251-0] [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: 07/29/2020] [Accepted: 02/08/2021] [Indexed: 12/01/2022]
Abstract
PURPOSE Τhis study aimed to optimize the 89Zr-radiolabelling of bintrafusp alfa investigational drug product and controls, and perform the in vitro and in vivo characterization of 89Zr-Df-bintrafusp alfa and 89Zr-Df-control radioconjugates. METHODS Bintrafusp alfa (anti-PD-L1 human IgG1 antibody fused to TGF-β receptor II (TGF-βRII), avelumab (anti-PD-L1 human IgG1 control antibody), isotype control (mutated inactive anti-PD-L1 IgG1 control antibody), and trap control (mutated inactive anti-PD-L1 human IgG1 fused to active TGF-βRII) were chelated with p-isothiocyanatobenzyl-desferrioxamine (Df). After radiolabelling with zirconium-89 (89Zr), radioconjugates were assessed for radiochemical purity, immunoreactivity, antigen binding affinity, and serum stability in vitro. In vivo biodistribution and imaging studies were performed with PET/CT to identify and quantitate 89Zr-Df-bintrafusp alfa tumour uptake in a PD-L1/TGF-β-positive murine breast cancer model (EMT-6). Specificity of 89Zr-Df-bintrafusp alfa was assessed via a combined biodistribution and imaging experiment in the presence of competing cold bintrafusp alfa (1 mg/kg). RESULTS Nanomolar affinities for PD-L1 were achieved with 89Zr-Df-bintrafusp alfa and 89Zr-avelumab. Biodistribution and imaging studies in PD-L1- and TGF-β-positive EMT-6 tumour-bearing BALB/c mice demonstrated the biologic similarity of 89Zr-Df-bintrafusp alfa and 89Zr-avelumab indicating the in vivo distribution pattern of bintrafusp alfa is driven by its PD-L1 binding arm. Competition study with 1 mg of unlabelled bintrafusp alfa or avelumab co-administered with trace dose of 89Zr-labelled bintrafusp alfa demonstrated the impact of dose and specificity of PD-L1 targeting in vivo. CONCLUSION Molecular imaging of 89Zr-Df-bintrafusp alfa biodistribution was achievable and allows non-invasive quantitation of tumour uptake of 89Zr-Df-bintrafusp alfa, suitable for use in bioimaging clinical trials in cancer patients.
Collapse
Affiliation(s)
- Ingrid Julienne Georgette Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Yit Wooi Goh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia
| | - Nancy Guo
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia
| | - Hui Kong Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Diana Cao
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Zhanqi Liu
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Uwe Ackermann
- School of Cancer Medicine, La Trobe University, Melbourne, Australia.,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Christian Werner Wichmann
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Alexander Franklin McDonald
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Nhi Huynh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia
| | - Graeme Joseph O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Sylvia Jie Gong
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia.,School of Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Fiona Elizabeth Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, Australia
| | - Linghui Li
- EMD Serono Research & Development Institute, Inc., a business of Merck KGaA, Darmstadt, Germany, Billerica, MA, USA
| | - Wanping Geng
- EMD Serono Research & Development Institute, Inc., a business of Merck KGaA, Darmstadt, Germany, Billerica, MA, USA
| | - Anup Zutshi
- EMD Serono Research & Development Institute, Inc., a business of Merck KGaA, Darmstadt, Germany, Billerica, MA, USA
| | - Yan Lan
- EMD Serono Research & Development Institute, Inc., a business of Merck KGaA, Darmstadt, Germany, Billerica, MA, USA
| | - Andrew Mark Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, 145 Studley Road, Heidelberg, Melbourne, Victoria, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Melbourne, Australia. .,Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Australia. .,Department of Medicine, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
99
|
Singh S, Bano A, Saraya A, Das P, Sharma R. iTRAQ-based analysis for the identification of MARCH8 targets in human esophageal squamous cell carcinoma. J Proteomics 2021; 236:104125. [PMID: 33540066 DOI: 10.1016/j.jprot.2021.104125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 12/04/2020] [Accepted: 01/19/2021] [Indexed: 12/14/2022]
Abstract
MARCH8 is an E3 ligase, primarily involved in immune-modulation. Recently, we reported its aberrant expression in human esophageal squamous cell carcinoma. However, exact mechanisms by which it regulates cancer have been poorly understood. We applied high-throughput quantitative proteomics approach to identify downstream protein targets of MARCH8. Silencing of endogenous MARCH8 in ESCC cells followed by LC-MS/MS analysis led to identification of 1,029 unique proteins showing altered expression post MARCH8 knockdown. Several previously reported MARCH8 target proteins viz. TFR1, syntaxin-4, e-cadherin and CD44 were found to be upregulated. Furthermore, new putative targets of MARCH8, including β2M, were identified in the present study. We demonstrated that MARCH8 interacts with and ubiquitinates CDH1 and β2M. Inhibiting proteasome activity with MG132 prevented CDH1 and β2M degradation, indicating that MARCH8 might be targeting CDH1 and β2M for proteasomal degradation. Further, loss of β2M and CDH1 expression significantly and inversely correlated with MARCH8 expression in ESCC tissues (r = -0.737 and - 0.651, respectively; p < 0.01). In conclusion, our present study has led to identification of new targets of MARCH8 and suggests the role of MARCH8 in regulating CDH1 and β2M turnover in esophageal cancer cells. SIGNIFICANCE: The use of quantitative proteomics carried out has led to the recognition of new targets of MARCH8. The present study gives a broad understanding of the molecular remodeling arising in the ESCC after MARCH8 knockdown. The study also solidifies the idea that role of MARCH8 is not just limited to immunomodulation as silencing of MARCH8 affects various other processes such as protein processing and localization. This study might help in understanding the regulation of MARCH8 in ESCCs and the mechanism by which MARCH8 might be facilitating cancer cells to evade immune surveillance.
Collapse
Affiliation(s)
- Shivam Singh
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector-16(C), Dwarka, New Delhi 110078, India
| | - Arjumand Bano
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector-16(C), Dwarka, New Delhi 110078, India
| | - Anoop Saraya
- Department of Gastroenterology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Rinu Sharma
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector-16(C), Dwarka, New Delhi 110078, India.
| |
Collapse
|
100
|
Immunization with alloantibodies-covered melanoma cells induces regional antitumor effects that become systemic when combined with 5-FU treatment. Cancer Lett 2021; 503:151-162. [PMID: 33545224 DOI: 10.1016/j.canlet.2021.01.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 12/26/2022]
Abstract
Alloantibodies, in particular immunoglobulin G (allo-IgG), confer a rejection advantage to tumors sharing the same major histocompatibility complex (MHC) in mice. However, when administrated intratumorally, this effect can only be achieved in combination with dendritic cells (DCs) activation. Here, we developed high titer allo-IgG by multiple rounds of immunization with allogenic B16 melanoma cells, which allows for the strong binding with B16 cells. We demonstrate that B16 cells incubated with these allo-IgG (referred to as allo-IgG-B16) become highly immunogenic, which release tumor antigens that are efficiently presented by classic DCs in lymph nodes (LNs). Injection of allo-IgG-B16 turns the tumor into an immune hot one and even elicits a systemic antitumor response when used together with 5-fluorouracil (5-FU). This systemic response is tumor-specific and relies on the critical site - LNs. Our findings provide a rationale for the use of allo-IgG in cancer treatment.
Collapse
|