1
|
Wawrzyniak P, Hartman ML. Dual role of interferon-gamma in the response of melanoma patients to immunotherapy with immune checkpoint inhibitors. Mol Cancer 2025; 24:89. [PMID: 40108693 PMCID: PMC11924818 DOI: 10.1186/s12943-025-02294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
Interferon-gamma (IFN-γ) is a cytokine produced mainly by immune cells and can affect cancer cells by modulating the activity of multiple signaling pathways, including the canonical Janus-activated kinase/signal transducer and activator of transcription (JAK/STAT) cascade. In melanoma, IFN-γ can exert both anticancer effects associated with cell-cycle arrest and cell death induction and protumorigenic activity related to immune evasion leading to melanoma progression. Notably, IFN-γ plays a crucial role in the response of melanoma patients to immunotherapy with immune checkpoint inhibitors (ICIs), which are currently used in the clinic. As these agents target programmed death-1 (PD-1) and its ligand (PD-L1), cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and lymphocyte-activation gene 3 (LAG-3), they are designed to restore the antimelanoma immune response. In this respect, IFN-γ produced by cells in the tumor microenvironment in response to ICIs has a beneficial influence on both immune and melanoma cells by increasing antigen presentation, recruiting additional T-cells to the tumor site, and inducing direct antiproliferative effects and apoptosis in melanoma cells. Therefore, IFN-γ itself and IFN-γ-related gene signatures during the response to ICIs can constitute biomarkers or predictors of the clinical outcome of melanoma patients treated with ICIs. However, owing to its multifaceted roles, IFN-γ can also contribute to developing mechanisms associated with the acquisition of resistance to ICIs. These mechanisms can be associated with either decreased IFN-γ levels in the tumor microenvironment or diminished responsiveness to IFN-γ due to changes in the melanoma phenotypes associated with affected activity of other signaling pathways or genetic alterations e.g., in JAK, which restricts the ability of melanoma cells to respond to IFN-γ. In this respect, the influence of IFN-γ on melanoma-specific regulators of the dynamic plasticity of the cell phenotype, including microphthalmia-associated transcription factor (MITF) and nerve growth factor receptor (NGFR)/CD271 can affect the clinical efficacy of ICIs. This review comprehensively discusses the role of IFN-γ in the response of melanoma patients to ICIs with respect to its positive influence and role in IFN-γ-related mechanisms of resistance to ICIs as well as the potential use of predictive markers on the basis of IFN-γ levels and signatures of IFN-γ-dependent genes.
Collapse
Affiliation(s)
- Piotr Wawrzyniak
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland
| | - Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215, Lodz, Poland.
| |
Collapse
|
2
|
Oh MS, Abascal J, Rennels AK, Salehi-Rad R, Dubinett SM, Liu B. Tumor Heterogeneity and the Immune Response in Non-Small Cell Lung Cancer: Emerging Insights and Implications for Immunotherapy. Cancers (Basel) 2025; 17:1027. [PMID: 40149360 PMCID: PMC11941341 DOI: 10.3390/cancers17061027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Resistance to immune checkpoint inhibitors (ICIs) represents a major challenge for the effective treatment of non-small cell lung cancer (NSCLC). Tumor heterogeneity has been identified as an important mechanism of treatment resistance in cancer and has been increasingly implicated in ICI resistance. The diversity and clonality of tumor neoantigens, which represent the target epitopes for tumor-specific immune cells, have been shown to impact the efficacy of immunotherapy. Advances in genomic techniques have further enhanced our understanding of clonal landscapes within NSCLC and their evolution in response to therapy. In this review, we examine the role of tumor heterogeneity during immune surveillance in NSCLC and highlight its spatial and temporal evolution as revealed by modern technologies. We explore additional sources of heterogeneity, including epigenetic and metabolic factors, that have come under greater scrutiny as potential mediators of the immune response. We finally discuss the implications of tumor heterogeneity on the efficacy of ICIs and highlight potential strategies for overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Michael S. Oh
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Jensen Abascal
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Austin K. Rennels
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
| | - Ramin Salehi-Rad
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Steven M. Dubinett
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Bin Liu
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; (M.S.O.); (J.A.); (A.K.R.); (R.S.-R.); (S.M.D.)
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Wu RZ, Sun QQ, Fu Y, Yu HN, Liu WY, Wu YH, Zhang H, Pan YL, Rui X. Fatty acid metabolism-derived prognostic model for lung adenocarcinoma: unraveling the link to survival and immune response. Front Immunol 2025; 16:1507845. [PMID: 40181976 PMCID: PMC11965909 DOI: 10.3389/fimmu.2025.1507845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Lung adenocarcinoma (LUAD) is one of the most common malignant tumors globally, characterized by poor prognosis and high mortality. Abnormal fatty acid metabolism plays a crucial role in LUAD progression. This study aims to develop a prognostic model based on fatty acid metabolism to improve the overall prognosis of LUAD. Materials and methods Bioinformatics analyses were performed using TCGA and GEO datasets, supplemented by cell experiments. A total of 309 fatty acid metabolism-related genes were identified from MsigDB. Differentially expressed genes were analyzed using the 'limma' R package. A prognostic model was constructed using LASSO regression and validated with survival analyses via the 'survminer', 'survival', and 'pROC' R packages. The analysis included somatic mutations, tumor mutation burden, clinical correlations, stemness analysis, cytokine correlations, and enrichment analysis. Protein interaction networks were constructed using STRING and Cytoscape, while immune cell infiltration and immunotherapy responses were evaluated with the 'oncoPredict' R package. Results were validated through cell experiments and immunohistochemistry staining of lung tissues. Results We identified 125 differentially expressed genes related to fatty acid metabolism, with 33 genes significantly associated with prognosis. Patients in the high-risk group had poorer overall survival and progression-free survival, and the risk score correlated with gender, N stage, clinical stage, and T stage. The risk score was also associated with cancer stem cells, with a significantly higher mRNAsi index in the high-risk group. Additionally, the risk score correlated with various cytokine expressions and showed significant enrichment in cell cycle pathways. Key genes like CDK1 were highly expressed in LUAD cell lines and validated in clinical samples. The low-risk group showed better responses to immune checkpoint inhibitors, with the risk score correlating with immune checkpoint gene expression. Conclusion This study successfully established a novel prognostic model based on fatty acid metabolism, which provides valuable insights for the treatment of LUAD.
Collapse
Affiliation(s)
- Rui-Ze Wu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Qian-Qian Sun
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yao Fu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Han-Nong Yu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Wei-Yang Liu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yong-Hui Wu
- School of Public Health, Harbin Medical University, Harbin, China
| | - Han Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yu-Lin Pan
- School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Rui
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
4
|
Kelley V, Baro M, Gasperi W, Ader N, Lea H, Lee H, Phoomak C, Kabeche L, King M, Contessa J. Loss of JAK1 Function Causes G2/M Cell Cycle Defects Vulnerable to Kif18a Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.638911. [PMID: 40060568 PMCID: PMC11888196 DOI: 10.1101/2025.02.19.638911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
To gain insight into biological mechanisms that cause resistance to DNA damage, we performed parallel pooled genetic CRISPR-Cas9 screening for survival in high risk HNSCC subtypes. Surprisingly, and in addition to ATM, DNAPK, and NFKB signaling, JAK1 was identified as a driver of tumor cell radiosensitivity. Knockout of JAK1 in HNSCC increases cell survival by enhancing the DNA damage-induced G2 arrest, and both knockout and JAK1 inhibition with abrocitinib prevent subsequent formation of radiation-induced micronuclei. Loss of JAK1 function does not affect canonical CDK1 signaling but does reduce activation of PLK1 and AURKA, kinases that regulate both G2 and M phase progression. Correspondingly, JAK1 KO was found to cause mitotic defects using both EdU labeling and live cell imaging techniques. Given this insight, we evaluated Kif18a inhibition as an approach to exacerbate mitotic stress and enhance the efficacy of radiation. These studies establish Kif18a inhibition as a novel strategy to counteract therapeutic resistance to DNA damage mediated by G2 cell cycle arrest.
Collapse
Affiliation(s)
- Vanessa Kelley
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Marta Baro
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - William Gasperi
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Nicholas Ader
- Department of Biology, University of North Carolina Greensboro, Greensboro, NC 27412 USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Hannah Lea
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Hojin Lee
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Chatchai Phoomak
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Biology, Chulalongkorn University, Bangkok 10330, Thailand
| | - Lilian Kabeche
- Department Molecular Biophysics and Biomedicine, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Megan King
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510 USA
| | - Joseph Contessa
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT 06510 USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510 USA
| |
Collapse
|
5
|
Wang T, Ma W, Zou Z, Zhong J, Lin X, Liu W, Sun W, Hu T, Xu Y, Chen Y. PD-1 blockade treatment in melanoma: Mechanism of response and tumor-intrinsic resistance. Cancer Sci 2025; 116:329-337. [PMID: 39601129 PMCID: PMC11786313 DOI: 10.1111/cas.16398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Malignant melanoma is characterized by high immunogenicity, genetic heterogeneity, and diverse pathological manifestations, affecting both skin and mucosa over the body. Pembrolizumab and nivolumab, both anti-PD-1 monoclonal antibodies, were approved by the US FDA for unresectable or metastatic melanoma in 2011 and 2014, respectively, with enduring and transformative outcomes. Despite marked clinical achievements, only a subset of patients manifested a complete response. Approximately 55% of melanoma patients exhibited primary resistance to PD-1 antibodies, with nearly 25% developing secondary resistance within 2 years of treatment. Thus, there is a critical need to comprehensively elucidate the mechanisms underlying the efficacy and resistance to PD-1 blockade. This review discusses the fundamental mechanisms of PD-1 blockade, encompassing insights from T cells and B cells, and presents resistance to anti-PD-1 with a particular focus on tumoral-intrinsic mechanisms in melanoma.
Collapse
Affiliation(s)
- Tong Wang
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Wenjie Ma
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Zijian Zou
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Jingqin Zhong
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Xinyi Lin
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Wanlin Liu
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Wei Sun
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Tu Hu
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Yu Xu
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| | - Yong Chen
- Department of Musculoskeletal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeShanghaiChina
| |
Collapse
|
6
|
Lau VWC, Mead GJ, Varyova Z, Mazet JM, Krishnan A, Roberts EW, Prota G, Gileadi U, Midwood KS, Cerundolo V, Gérard A. Remodelling of the immune landscape by IFNγ counteracts IFNγ-dependent tumour escape in mouse tumour models. Nat Commun 2025; 16:2. [PMID: 39746898 PMCID: PMC11696141 DOI: 10.1038/s41467-024-54791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Loss of IFNγ-sensitivity by tumours is thought to be a mechanism enabling evasion, but recent studies suggest that IFNγ-resistant tumours can be sensitised for immunotherapy, yet the underlying mechanism remains unclear. Here, we show that IFNγ receptor-deficient B16-F10 mouse melanoma tumours are controlled as efficiently as WT tumours despite their lower MHC class I expression. Mechanistically, IFNγ receptor deletion in B16-F10 tumours increases IFNγ availability, triggering a remodelling of the immune landscape characterised by inflammatory monocyte infiltration and the generation of 'mono-macs'. This altered myeloid compartment synergises with an increase in antigen-specific CD8+ T cells to promote anti-tumour immunity against IFNγ receptor-deficient tumours, with such an immune crosstalk observed around blood vessels. Importantly, analysis of transcriptomic datasets suggests that similar immune remodelling occurs in human tumours carrying mutations in the IFNγ pathway. Our work thus serves mechanistic insight for the crosstalk between tumour IFNγ resistance and anti-tumour immunity, and implicates this regulation for future cancer therapy.
Collapse
Affiliation(s)
- Vivian W C Lau
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Gracie J Mead
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Zofia Varyova
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Julie M Mazet
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Anagha Krishnan
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Immunodynamics Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Gennaro Prota
- MRC Translational Immune Discovery Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Uzi Gileadi
- MRC Translational Immune Discovery Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Kim S Midwood
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Vincenzo Cerundolo
- MRC Translational Immune Discovery Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Audrey Gérard
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Mei T, Ye T, Huang D, Xie Y, Xue Y, Zhou D, Wang W, Chen J. Triggering immunogenic death of cancer cells by nanoparticles overcomes immunotherapy resistance. Cell Oncol (Dordr) 2024; 47:2049-2071. [PMID: 39565509 DOI: 10.1007/s13402-024-01009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2024] [Indexed: 11/21/2024] Open
Abstract
Immunotherapy resistance poses a significant challenge in oncology, necessitating novel strategies to enhance the therapeutic efficacy. Immunogenic cell death (ICD), including necroptosis, pyroptosis and ferroptosis, triggers the release of tumor-associated antigens and numerous bioactive molecules. This release can potentiate a host immune response, thereby overcoming resistance to immunotherapy. Nanoparticles (NPs) with their biocompatible and immunomodulatory properties, are emerging as promising vehicles for the delivery of ICD-inducing agents and immune-stimulatory adjuvants to enhance immune cells tumoral infiltration and augment immunotherapy efficacy. This review explores the mechanisms underlying immunotherapy resistance, and offers an in-depth examination of ICD, including its principles and diverse modalities of cell death that contribute to it. We also provide a thorough overview of how NPs are being utilized to trigger ICD and bolster antitumor immunity. Lastly, we highlight the potential of NPs in combination with immunotherapy to revolutionize cancer treatment.
Collapse
Affiliation(s)
- Ting Mei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ting Ye
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Dingkun Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Yuxiu Xie
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China
| | - Ying Xue
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, 430022, China.
- Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Guan GF, Fu ZM, Zhang DJ, Guo YY, Guo F, Wan YN, Bai J, Zhao Y. Interferon Gamma Receptor 2 Collaborates With Circular RNA/MicroRNA to Modulate Programmed Cell Death-Ligand 1 Levels in Nasopharyngeal Carcinoma. World J Oncol 2024; 15:929-941. [PMID: 39697423 PMCID: PMC11650609 DOI: 10.14740/wjon1994] [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] [Received: 10/22/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Background The effectiveness of immune checkpoint therapy highlights the need to understand abnormal programmed cell death protein-1 (PD-1) expression in nasopharyngeal carcinoma (NPC), especially when treatments fail, or resistance develops. Interferon gamma (IFN-γ) signaling is crucial for regulating programmed cell death-ligand 1 (PD-L1) expression. Our study focuses on interferon gamma receptor 2 (IFNGR2), an essential part of the IFN-γ pathway, and its impact on malignant traits in NPC. Methods The expression levels of IFNGR2 and PD-L1 were accessed using quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). To understand the cellular phenotypic effects, small interfering RNA (siRNA)/short hairpin RNA (shRNA) knockdown techniques were used to evaluate cell viability, clonogenic survival, migration and invasion, immunohistochemistry, and tumor formation assays. The relationship between IFNGR2 and microRNAs (miRNAs)/circular RNAs (circRNAs) will be verified using methods such as circRNA stability assay, rescue, and dual-luciferase reporter assay. Results IFNGR2 was significantly overexpressed in NPC, and its expression positively correlated with PD-L1 levels. This overexpression contributed to increased cell proliferation, migration, invasion, clonogenicity, and tumor growth. Additionally, we identified an oncogenic circular RNA, circ_001377, and uncovered a novel mechanism by which upregulation of circ_001377 competitively bound to miR-498-3p. This interaction reduced miR-498-3p's ability to target IFNGR2. As a result, the diminished miR-498-3p led to increased IFNGR2 expression, which subsequently activated the IFN-γ signaling pathway and drove abnormal PD-L1 expression. Conclusions IFNGR2 is an oncogenic factor in NPC. The circ_001377/miR-498-3p interaction drives IFNGR2 upregulation and PD-L1 overexpression, suggesting that targeting this axis could improve therapeutic outcomes.
Collapse
Affiliation(s)
- Guo Fang Guan
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Ze Ming Fu
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - De Jun Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Ying Yuan Guo
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Fang Guo
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yi Ning Wan
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jie Bai
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Ying Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
9
|
Zhang Y, Xu Q, Gao Z, Zhang H, Xie X, Li M. High-throughput screening for optimizing adoptive T cell therapies. Exp Hematol Oncol 2024; 13:113. [PMID: 39538305 PMCID: PMC11562648 DOI: 10.1186/s40164-024-00580-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Adoptive T cell therapy is a pivotal strategy in cancer immunotherapy, demonstrating potent clinical efficacy. However, its limited durability often results in primary resistance. High-throughput screening technologies, which include both genetic and non-genetic approaches, facilitate the optimization of adoptive T cell therapies by enabling the selection of biologically significant targets or substances from extensive libraries. In this review, we examine advancements in high-throughput screening technologies and their applications in adoptive T cell therapies. We highlight the use of genetic screening for T cells, tumor cells, and other promising combination strategies, and elucidate the role of non-genetic screening in identifying small molecules and targeted delivery systems relevant to adoptive T cell therapies, providing guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Qinglong Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Zhifei Gao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| | - Meifang Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
10
|
Boreel DF, Sandker GGW, Ansems M, van den Bijgaart RJE, Peters JPW, Span PN, Adema GJ, Heskamp S, Bussink J. MHC-I and PD-L1 Expression is Associated with Decreased Tumor Outgrowth and is Radiotherapy-inducible in the Murine Head and Neck Squamous Cell Carcinoma Model MOC1. Mol Imaging Biol 2024; 26:835-846. [PMID: 39009951 PMCID: PMC11436446 DOI: 10.1007/s11307-024-01934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
INTRODUCTION Combined radiotherapy and immune checkpoint inhibition is a potential treatment option for head and neck squamous cell carcinoma (HNSCC). Immunocompetent mouse models can help to successfully develop radio- immunotherapy combinations and to increase our understanding of the effects of radiotherapy on the tumor microenvironment for future clinical translation. Therefore, the aim of this study was to develop a homogeneous, reproducible HNSCC model originating from the Mouse Oral Cancer 1 (MOC1) HNSCC cell line, and to explore the radiotherapy-induced changes in its tumor microenvironment, using flow cytometry and PD-L1 microSPECT/CT imaging. MATERIALS AND METHODS In vivo growing tumors originating from the parental MOC1 line were used to generate single cell derived clones. These clones were screened in vitro for their ability to induce programmed cell death ligand 1 (PD-L1) and major histocompatibility complex class I (MHC-I) following IFNγ exposure. Clones with different IFNγ sensitivity were inoculated in C57BL/6 mice and assessed for tumor outgrowth. The composition of the tumor microenvironment of a stably growing (non)irradiated MOC1-derived clone was assessed by immunohistochemistry, flow cytometry and PD-L1 microSPECT/CT. RESULTS Low in vitro inducibility of MHC-I and PD-L1 by IFNγ was associated with increased tumor outgrowth of MOC1 clones in vivo. Flow cytometry analysis of cells derived from a stable in vivo growing MOC1 clone MOC1.3D5low showed expression of MHC-I and PD-L1 on several cell populations within the tumor. Upon irradiation, MHC-I and PD-L1 increased on leukocytes (CD45.2+) and cancer associated fibroblasts (CD45.2-/EpCAM-/CD90.1+). Furthermore, PD-L1 microSPECT/CT showed increased tumor uptake of radiolabeled PD-L1 antibodies with a heterogeneous spatial distribution of the radio signal, which co-localized with PD-L1+ and CD45.2+ areas. DISCUSSION PD-L1 and MHC-I inducibility by IFNγ in vitro is associated with tumor outgrowth of MOC1 clones in vivo. In tumors originating from a stably growing MOC1-derived clone, expression of these immune-related markers was induced by irradiation shown by flow cytometry on several cell populations within the tumor microenvironment such as immune cells and cancer associated fibroblasts. PD-L1 microSPECT/CT showed increased tumor uptake following radiotherapy, and autoradiography showed correlation of uptake with areas that are heavily infiltrated by immune cells. Knowledge of radiotherapy-induced effects on the tumor microenvironment in this model can help optimize timing and dosage for radio- immunotherapy combination strategies in future research.
Collapse
Affiliation(s)
- Daan F Boreel
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands.
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands.
| | - Gerwin G W Sandker
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands
| | - Marleen Ansems
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Renske J E van den Bijgaart
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Johannes P W Peters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Paul N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboudumc, Geert Grooteplein 10, Nijmegen, 6525GA, The Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Geert Grooteplein Zuid 32, 6525GA, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Li S, Dai Y, Chen J, Yan F, Yang Y. MRI-based habitat imaging in cancer treatment: current technology, applications, and challenges. Cancer Imaging 2024; 24:107. [PMID: 39148139 PMCID: PMC11328409 DOI: 10.1186/s40644-024-00758-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Extensive efforts have been dedicated to exploring the impact of tumor heterogeneity on cancer treatment at both histological and genetic levels. To accurately measure intra-tumoral heterogeneity, a non-invasive imaging technique, known as habitat imaging, was developed. The technique quantifies intra-tumoral heterogeneity by dividing complex tumors into distinct sub- regions, called habitats. This article reviews the following aspects of habitat imaging in cancer treatment, with a focus on radiotherapy: (1) Habitat imaging biomarkers for assessing tumor physiology; (2) Methods for habitat generation; (3) Efforts to combine radiomics, another imaging quantification method, with habitat imaging; (4) Technical challenges and potential solutions related to habitat imaging; (5) Pathological validation of habitat imaging and how it can be utilized to evaluate cancer treatment by predicting treatment response including survival rate, recurrence, and pathological response as well as ongoing open clinical trials.
Collapse
Affiliation(s)
- Shaolei Li
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai, 201800, China
| | - Yongming Dai
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, 201210, China
| | - Jiayi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai, 201800, China
| | - Fuhua Yan
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai, 201800, China
- Department of Radiology, Ruijin Hospital, Shanghai, 201800, China
| | - Yingli Yang
- Institute for Medical Imaging Technology, Ruijin Hospital, Shanghai, 201800, China.
| |
Collapse
|
12
|
Hickey JW, Agmon E, Horowitz N, Tan TK, Lamore M, Sunwoo JB, Covert MW, Nolan GP. Integrating multiplexed imaging and multiscale modeling identifies tumor phenotype conversion as a critical component of therapeutic T cell efficacy. Cell Syst 2024; 15:322-338.e5. [PMID: 38636457 PMCID: PMC11030795 DOI: 10.1016/j.cels.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/07/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Cancer progression is a complex process involving interactions that unfold across molecular, cellular, and tissue scales. These multiscale interactions have been difficult to measure and to simulate. Here, we integrated CODEX multiplexed tissue imaging with multiscale modeling software to model key action points that influence the outcome of T cell therapies with cancer. The initial phenotype of therapeutic T cells influences the ability of T cells to convert tumor cells to an inflammatory, anti-proliferative phenotype. This T cell phenotype could be preserved by structural reprogramming to facilitate continual tumor phenotype conversion and killing. One takeaway is that controlling the rate of cancer phenotype conversion is critical for control of tumor growth. The results suggest new design criteria and patient selection metrics for T cell therapies, call for a rethinking of T cell therapeutic implementation, and provide a foundation for synergistically integrating multiplexed imaging data with multiscale modeling of the cancer-immune interface. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- John W Hickey
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Eran Agmon
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Center for Cell Analysis and Modeling, University of Connecticut Health, Farmington, CT 06032, USA
| | - Nina Horowitz
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Tze-Kai Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew Lamore
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - John B Sunwoo
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Otolaryngology, Head and Neck Surgery, Stanford Cancer Institute Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Markus W Covert
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.
| | - Garry P Nolan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
13
|
Xu Y, Du H, Guo W, Liu B, Yan W, Zhang C, Qin L, Huang J, Wang H, Wu S, Ren W, Zou Y, Wang J, Zhu Q, Xu Y, Gu H. Discovery of Highly Potent Small-Molecule PD-1/PD-L1 Inhibitors with a Novel Scaffold for Cancer Immunotherapy. J Med Chem 2024; 67:4083-4099. [PMID: 38348878 DOI: 10.1021/acs.jmedchem.3c02362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2024]
Abstract
Inhibition of the PD-1/PD-L1 interaction through small-molecule inhibitors is a promising therapeutic approach in cancer immunotherapy. Herein, we utilized BMS-202 as the lead compound to develop a series of novel PD-1/PD-L1 small-molecule inhibitors with a naphthyridin scaffold. Among these compounds, X14 displayed the most potent inhibitory activity for the PD-1/PD-L1 interaction (IC50 = 15.73 nM). Furthermore, X14 exhibited good binding affinity to both human PD-L1 (KD = 14.62 nM) and mouse PD-L1 (KD = 392 nM). In particular, X14 showed favorable pharmacokinetic properties (oral bioavailability, F = 58.0%). In the 4T1 (mouse breast cancer cells) syngeneic mouse model, intragastric administration of X14 at 10 mg/kg displayed significant antitumor efficacy (TGI = 66%). Mechanistic investigations revealed that X14 effectively enhanced T-cell infiltration within the tumor microenvironment. Our study demonstrates that compound X14 exhibits potential as a candidate compound for the development of orally effective small-molecule inhibitors targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Yongling Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Huijie Du
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Weibo Guo
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
- Xi'an Xintong Pharmaceutical Research Co., Ltd, Xi'an 710061, China
| | - Beibei Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Wenxin Yan
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Chi Zhang
- Xi'an Xintong Pharmaceutical Research Co., Ltd, Xi'an 710061, China
| | - Long Qin
- Xi'an Xintong Pharmaceutical Research Co., Ltd, Xi'an 710061, China
| | - Jingling Huang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Hongxia Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Shiqi Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Weijie Ren
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Zou
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Wang
- China Pharmaceutical University Center for Analysis and Testing, China Pharmaceutical University, Nanjing 211198, China
| | - Qihua Zhu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yungen Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Hongfeng Gu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
14
|
Chen A, Yu Z, Ma N, Lu X, Zhang Y, Xu W, Wang Y, Xie J, Qin Y, Mo G, Wu S, Hou J, Zhu W. Atovaquone enhances antitumor efficacy of TCR-T therapy by augmentation of ROS-induced ferroptosis in hepatocellular carcinoma. Cancer Immunol Immunother 2024; 73:49. [PMID: 38349553 PMCID: PMC10864481 DOI: 10.1007/s00262-024-03628-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/06/2024] [Indexed: 02/15/2024]
Abstract
T-cell receptor (TCR) engineered T-cell therapy has recently emerged as a promising adoptive immunotherapy approach for tumor treatment, yet hindered by tumor immune evasion resulting in poor therapeutic efficacy. The introduction of ferroptosis-targeted inducers offers a potential solution, as they empower T cells to induce ferroptosis and exert influence over the tumor microenvironment. Atovaquone (ATO) stands as a prospective pharmaceutical candidate with the potential to target ferroptosis, effectively provoking an excessive generation and accumulation of reactive oxygen species (ROS). In this study, we evaluated the effectiveness of a combination therapy comprising ATO and TCR-T cells against hepatocellular carcinoma (HCC), both in vitro and in vivo. The results of lactate dehydrogenase and cytokine assays demonstrated that ATO enhanced cytotoxicity mediated by AFP-specific TCR-T cells and promoted the release of IFN-γ in vitro. Additionally, in an established HCC xenograft mouse model, the combined therapy with low-dose ATO and TCR-T cells exhibited heightened efficacy in suppressing tumor growth, with no apparent adverse effects, comparable to the results achieved through monotherapy. The RNA-seq data unveiled a significant activation of the ferroptosis-related pathway in the combination therapy group in comparison to the TCR-T cells group. Mechanistically, the synergy between ATO and TCR-T cells augmented the release of IFN-γ by TCR-T cells, while concurrently elevating the intracellular and mitochondrial levels of ROS, expanding the labile iron pool, and impairing the integrity of the mitochondrial membrane in HepG2 cells. This multifaceted interaction culminated in the potentiation of ferroptosis within the tumor, primarily induced by an excess of ROS. In summary, the co-administration of ATO and TCR-T cells in HCC exhibited heightened vulnerability to ferroptosis. This heightened susceptibility led to the inhibition of tumor growth and the stimulation of an anti-tumor immune response. These findings suggest that repurposing atovaquone for adoptive cell therapy combination therapy holds the potential to enhance treatment outcomes in HCC.
Collapse
Affiliation(s)
- Anan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhiwu Yu
- Department of Laboratory Medicine, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Na Ma
- Department of Pathology, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yajing Zhang
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Weikang Xu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510220, China
| | - Yiyue Wang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiayi Xie
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuqi Qin
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guoheng Mo
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, 510515, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Wei Zhu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Takeuchi E, Ogino H, Kondo K, Okano Y, Ichihara S, Kunishige M, Kadota N, Machida H, Hatakeyama N, Naruse K, Nokihara H, Shinohara T, Nishioka Y. An increased relative eosinophil count as a predictive dynamic biomarker in non-small cell lung cancer patients treated with immune checkpoint inhibitors. Thorac Cancer 2024; 15:248-257. [PMID: 38087769 PMCID: PMC10803223 DOI: 10.1111/1759-7714.15191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND An increased relative eosinophil count (REC) has potential as a predictive biomarker for a beneficial clinical response and outcome to cancer immunotherapies. Therefore, the present study investigated the impact of an increased posttreatment REC on the prognosis of non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs). METHODS We retrospectively reviewed all 151 patients diagnosed with NSCLC and treated with ICI monotherapy and blood test data between March 2016 and August 2021 at National Hospital Organization Kochi Hospital and Tokushima University. RESULTS A total of 151 patients with a mean age of 69 years were included. REC after 4 weeks of initial ICI monotherapy was higher than pretreatment REC in 87 patients but not in 64. REC after 4 weeks of the ICI treatment with and without an increased REC were 4.4 and 1.8%, respectively (p < 0.001). Disease control rates (DCR) were significantly higher in patients with than in those without an increased REC (84% vs. 47%, p < 0.001). The median overall survival (OS) of lung cancer patients with or without an increased REC were 674 and 234 days, respectively. A Kaplan-Meier univariate analysis revealed a significant difference in OS between the two groups (p < 0.001). A Cox proportional regression analysis identified an increased REC as an independent predictor of OS (p = 0.003). CONCLUSION ICI-treated NSCLC patients with an increased REC after 4 weeks of treatment had a better DCR and prognosis than the other patients examined.
Collapse
Affiliation(s)
- Eiji Takeuchi
- Department of Clinical InvestigationNational Hospital Organization Kochi HospitalKochiJapan
| | - Hirokazu Ogino
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Kensuke Kondo
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Yoshio Okano
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Seiya Ichihara
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Michihiro Kunishige
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Naoki Kadota
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Hisanori Machida
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Nobuo Hatakeyama
- Department of Respiratory MedicineNational Hospital Organization Kochi HospitalKochiJapan
| | - Keishi Naruse
- Department of PathologyNational Hospital Organization Kochi HospitalKochiJapan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Tsutomu Shinohara
- Department of Community Medicine for RespirologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and RheumatologyGraduate School of Biomedical Sciences, Tokushima UniversityTokushimaJapan
| |
Collapse
|
16
|
Knopf P, Stowbur D, Hoffmann SHL, Hermann N, Maurer A, Bucher V, Poxleitner M, Tako B, Sonanini D, Krishnamachary B, Sinharay S, Fehrenbacher B, Gonzalez-Menendez I, Reckmann F, Bomze D, Flatz L, Kramer D, Schaller M, Forchhammer S, Bhujwalla ZM, Quintanilla-Martinez L, Schulze-Osthoff K, Pagel MD, Fransen MF, Röcken M, Martins AF, Pichler BJ, Ghoreschi K, Kneilling M. Acidosis-mediated increase in IFN-γ-induced PD-L1 expression on cancer cells as an immune escape mechanism in solid tumors. Mol Cancer 2023; 22:207. [PMID: 38102680 PMCID: PMC10722725 DOI: 10.1186/s12943-023-01900-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 11/12/2023] [Indexed: 12/17/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, yet the efficacy of these treatments is often limited by the heterogeneous and hypoxic tumor microenvironment (TME) of solid tumors. In the TME, programmed death-ligand 1 (PD-L1) expression on cancer cells is mainly regulated by Interferon-gamma (IFN-γ), which induces T cell exhaustion and enables tumor immune evasion. In this study, we demonstrate that acidosis, a common characteristic of solid tumors, significantly increases IFN-γ-induced PD-L1 expression on aggressive cancer cells, thus promoting immune escape. Using preclinical models, we found that acidosis enhances the genomic expression and phosphorylation of signal transducer and activator of transcription 1 (STAT1), and the translation of STAT1 mRNA by eukaryotic initiation factor 4F (elF4F), resulting in an increased PD-L1 expression. We observed this effect in murine and human anti-PD-L1-responsive tumor cell lines, but not in anti-PD-L1-nonresponsive tumor cell lines. In vivo studies fully validated our in vitro findings and revealed that neutralizing the acidic extracellular tumor pH by sodium bicarbonate treatment suppresses IFN-γ-induced PD-L1 expression and promotes immune cell infiltration in responsive tumors and thus reduces tumor growth. However, this effect was not observed in anti-PD-L1-nonresponsive tumors. In vivo experiments in tumor-bearing IFN-γ-/- mice validated the dependency on immune cell-derived IFN-γ for acidosis-mediated cancer cell PD-L1 induction and tumor immune escape. Thus, acidosis and IFN-γ-induced elevation of PD-L1 expression on cancer cells represent a previously unknown immune escape mechanism that may serve as a novel biomarker for anti-PD-L1/PD-1 treatment response. These findings have important implications for the development of new strategies to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Philipp Knopf
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dimitri Stowbur
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Sabrina H L Hoffmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Natalie Hermann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Valentina Bucher
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Marilena Poxleitner
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Bredi Tako
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
| | - Balaji Krishnamachary
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sanhita Sinharay
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | | | - Irene Gonzalez-Menendez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Felix Reckmann
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
| | - David Bomze
- Department of Dermatology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Lukas Flatz
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Daniela Kramer
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
| | - Martin Schaller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | | | - Zaver M Bhujwalla
- Division of Cancer Imaging Research, The Russell H Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Leticia Quintanilla-Martinez
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Institute of Pathology and Neuropathology, Department of Pathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, Tübingen University Hospital, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Mark D Pagel
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Martin Röcken
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - André F Martins
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10117, Berlin, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Röntgenweg 13, 72076, Tübingen, Germany.
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
17
|
Champhekar A, Heymans R, Saco J, Turon Font G, Gonzalez C, Gao A, Pham J, Lee J, Maryoung R, Medina E, Campbell KM, Karin D, Austin D, Damioseaux R, Ribas A. ERK mediates interferon gamma-induced melanoma cell death. Mol Cancer 2023; 22:165. [PMID: 37803324 PMCID: PMC10557262 DOI: 10.1186/s12943-023-01868-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Interferon-gamma (IFNγ) exerts potent growth inhibitory effects on a wide range of cancer cells through unknown signaling pathways. We pursued complementary screening approaches to characterize the growth inhibition pathway. METHODS We performed chemical genomics and whole genome targeting CRISPR/Cas9 screens using patient-derived melanoma lines to uncover essential nodes in the IFNγ-mediated growth inhibition pathway. We used transcriptomic profiling to identify cell death pathways activated upon IFNγ exposure. Live imaging experiments coupled with apoptosis assays confirmed the involvement of these pathways in IFNγ-mediated cell death. RESULTS We show that IFNγ signaling activated ERK. Blocking ERK activation rescued IFNγ-mediated apoptosis in 17 of 23 (~ 74%) cell lines representing BRAF, NRAS, NF1 mutant, and triple wild type subtypes of cutaneous melanoma. ERK signaling induced a stress response, ultimately leading to apoptosis through the activity of DR5 and NOXA proteins. CONCLUSIONS Our results provide a new understanding of the IFNγ growth inhibition pathway, which will be crucial in defining mechanisms of immunotherapy response and resistance.
Collapse
Affiliation(s)
- Ameya Champhekar
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Rachel Heymans
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Justin Saco
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - Guillem Turon Font
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Cynthia Gonzalez
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Anne Gao
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - John Pham
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - June Lee
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ryan Maryoung
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Egmidio Medina
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Katie M Campbell
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Daniel Karin
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
| | - David Austin
- California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
| | - Robert Damioseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA
- Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, CA, 90095, USA
| | - Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, Los Angeles, CA, 90095, USA.
- Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, 94129, USA.
| |
Collapse
|
18
|
Wong CW, Huang YY, Hurlstone A. The role of IFN-γ-signalling in response to immune checkpoint blockade therapy. Essays Biochem 2023; 67:991-1002. [PMID: 37503572 PMCID: PMC10539948 DOI: 10.1042/ebc20230001] [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: 03/31/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/29/2023]
Abstract
Treatment with immune checkpoint inhibitors, widely known as immune checkpoint blockade therapy (ICBT), is now the fourth pillar in cancer treatment, offering the chance of durable remission for patients with advanced disease. However, ICBT fails to induce objective responses in most cancer patients with still others progressing after an initial response. It is necessary, therefore, to elucidate the primary and acquired resistance mechanisms to ICBT to improve its efficacy. Here, we highlight the paradoxical role of the cytokine interferon-γ (IFN-γ) in ICBT response: on the one hand induction of IFN-γ signalling in the tumour microenvironment correlates with good ICBT response as it drives the cellular immune responses required for tumour destruction; nonetheless, IFN-γ signalling is implicated in ICBT acquired resistance. We address the negative feedback and immunoregulatory effects of IFN-γ signalling that promote immune evasion and resistance to ICBT and discuss how these can be targeted pharmacologically to restore sensitivity or circumvent resistance.
Collapse
Affiliation(s)
- Chun Wai Wong
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, U.K
| | - Yang Yu Huang
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, U.K
| | - Adam Hurlstone
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, U.K
- Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
19
|
Lems CM, Burger GA, Beltman JB. Tumor-mediated immunosuppression and cytokine spreading affects the relation between EMT and PD-L1 status. Front Immunol 2023; 14:1219669. [PMID: 37638024 PMCID: PMC10449452 DOI: 10.3389/fimmu.2023.1219669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/30/2023] [Indexed: 08/29/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) and immune resistance mediated by Programmed Death-Ligand 1 (PD-L1) upregulation are established drivers of tumor progression. Their bi-directional crosstalk has been proposed to facilitate tumor immunoevasion, yet the impact of immunosuppression and spatial heterogeneity on the interplay between these processes remains to be characterized. Here we study the role of these factors using mathematical and spatial models. We first designed models incorporating immunosuppressive effects on T cells mediated via PD-L1 and the EMT-inducing cytokine Transforming Growth Factor beta (TGFβ). Our models predict that PD-L1-mediated immunosuppression merely reduces the difference in PD-L1 levels between EMT states, while TGFβ-mediated suppression also causes PD-L1 expression to correlate negatively with TGFβ within each EMT phenotype. We subsequently embedded the models in multi-scale spatial simulations to explicitly describe heterogeneity in cytokine levels and intratumoral heterogeneity. Our multi-scale models show that Interferon gamma (IFNγ)-induced partial EMT of a tumor cell subpopulation can provide some, albeit limited protection to bystander tumor cells. Moreover, our simulations show that the true relationship between EMT status and PD-L1 expression may be hidden at the population level, highlighting the importance of studying EMT and PD-L1 status at the single-cell level. Our findings deepen the understanding of the interactions between EMT and the immune response, which is crucial for developing novel diagnostics and therapeutics for cancer patients.
Collapse
Affiliation(s)
| | | | - Joost B. Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
20
|
Zhang J, Xu L, Zhang J, Liu Y, Li X, Ren T, Liu H. Pan-cancer analysis of the prognostic and immunological role of matrix metalloproteinase 9. Medicine (Baltimore) 2023; 102:e34499. [PMID: 37505149 PMCID: PMC10378727 DOI: 10.1097/md.0000000000034499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/15/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Matrix metalloproteinase 9 (MMP9), a zinc ion-dependent endopeptidase, is one of the most complex matrix metalloproteinases in the gelatinase family. During tissue remodeling, MMP9 leads to gelatin and collagen degradation, which in turn promotes tumor invasion and metastasis. However, comprehensive pan-cancer analysis has not been performed for MMP9. In addition, the diagnostic and prognostic value of MMP9 as a cancer biomarker remain poorly understood, as well as the utility of MMP9 expression as a predictor of immunological responses. Based on a comprehensive analysis of bioinformatics information, we investigated MMP9 expression in different cancers, correlations between MMP9 expression and cancer prognosis and gene mutations, and relationships between MMP9 expression and immune cell infiltration. Our results indicated that MMP9 was highly expressed in most malignant cancers. MMP9 expression was significantly positively or negatively associated with the clinical prognoses of patients with different kinds of cancer. Furthermore, MMP9 expression significantly correlated with infiltrating cells and the expression levels of immune checkpoint genes. This pan-cancer analysis provides comprehensive information on the potential value of MMP9 as a theranostic biomarker.
Collapse
Affiliation(s)
- Jie Zhang
- Clinical Medical School, Chengdu Medical College, Chengdu, China
- Oncology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Key Clinical Specialty of Sichuan Province (Oncology Department), The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Lei Xu
- Clinical Medical School, Chengdu Medical College, Chengdu, China
- Gynecology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Jingjun Zhang
- Oncology Department, The People’s Hospital of Jianyang City, Chengdu, China
| | - Ying Liu
- Clinical Medical School, Chengdu Medical College, Chengdu, China
| | - Xiang Li
- Clinical Medical School, Chengdu Medical College, Chengdu, China
| | - Tao Ren
- Clinical Medical School, Chengdu Medical College, Chengdu, China
- Oncology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Key Clinical Specialty of Sichuan Province (Oncology Department), The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Hairong Liu
- Clinical Medical School, Chengdu Medical College, Chengdu, China
- Science and Technology Department, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
21
|
Gu Y, Zhang Z, Camps MG, Ossendorp F, Wijdeven RH, ten Dijke P. Genome-wide CRISPR screens define determinants of epithelial-mesenchymal transition mediated immune evasion by pancreatic cancer cells. SCIENCE ADVANCES 2023; 9:eadf9915. [PMID: 37450593 PMCID: PMC10348683 DOI: 10.1126/sciadv.adf9915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
The genetic circuits that allow cancer cells to evade immune killing via epithelial mesenchymal plasticity remain poorly understood. Here, we showed that mesenchymal-like (Mes) KPC3 pancreatic cancer cells were more resistant to cytotoxic T lymphocyte (CTL)-mediated killing than the parental epithelial-like (Epi) cells and used parallel genome-wide CRISPR screens to assess the molecular underpinnings of this difference. Core CTL-evasion genes (such as IFN-γ pathway components) were clearly evident in both types. Moreover, we identified and validated multiple Mes-specific regulators of cytotoxicity, such as Egfr and Mfge8. Both genes were significantly higher expressed in Mes cancer cells, and their depletion sensitized Mes cancer cells to CTL-mediated killing. Notably, Mes cancer cells secreted more Mfge8 to inhibit proliferation of CD8+ T cells and production of IFN-γ and TNFα. Clinically, increased Egfr and Mfge8 expression was correlated with a worse prognosis. Thus, Mes cancer cells use Egfr-mediated intrinsic and Mfge8-mediated extrinsic mechanisms to facilitate immune escape from CD8+ T cells.
Collapse
Affiliation(s)
- Yuanzhuo Gu
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, Netherlands
| | - Zhengkui Zhang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Marcel G. M. Camps
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Ferry Ossendorp
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Ruud H. Wijdeven
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, Netherlands
| | - Peter ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, Netherlands
| |
Collapse
|
22
|
Boulch M, Cazaux M, Cuffel A, Guerin MV, Garcia Z, Alonso R, Lemaître F, Beer A, Corre B, Menger L, Grandjean CL, Morin F, Thieblemont C, Caillat-Zucman S, Bousso P. Tumor-intrinsic sensitivity to the pro-apoptotic effects of IFN-γ is a major determinant of CD4 + CAR T-cell antitumor activity. NATURE CANCER 2023; 4:968-983. [PMID: 37248395 PMCID: PMC10368531 DOI: 10.1038/s43018-023-00570-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/27/2023] [Indexed: 05/31/2023]
Abstract
CD4+ T cells and CD4+ chimeric antigen receptor (CAR) T cells display highly variable antitumor activity in preclinical models and in patients; however, the mechanisms dictating how and when CD4+ T cells promote tumor regression are incompletely understood. With the help of functional intravital imaging, we report that interferon (IFN)-γ production but not perforin-mediated cytotoxicity was the dominant mechanism for tumor elimination by anti-CD19 CD4+ CAR T cells. Mechanistically, mouse or human CD4+ CAR T-cell-derived IFN-γ diffused extensively to act on tumor cells at distance selectively killing tumors sensitive to cytokine-induced apoptosis, including antigen-negative variants. In anti-CD19 CAR T-cell-treated patients exhibiting elevated CAR CD4:CD8 ratios, strong induction of serum IFN-γ was associated with increased survival. We propose that the sensitivity of tumor cells to the pro-apoptotic activity of IFN-γ is a major determinant of CD4+ CAR T-cell efficacy and may be considered to guide the use of CD4+ T cells during immunotherapy.
Collapse
Affiliation(s)
- Morgane Boulch
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Marine Cazaux
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Alexis Cuffel
- Université de Paris Cité, Hôpital Saint-Louis, AP-HP Nord, Laboratoire d'Immunologie, Paris, France
- INSERM UMR976, Institut de Recherche St-Louis, Paris, France
| | - Marion V Guerin
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Zacarias Garcia
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Ruby Alonso
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Fabrice Lemaître
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Alexander Beer
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Béatrice Corre
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Laurie Menger
- Gustave Roussy, Villejuif, France; INSERM U1015, Villejuif, France
| | - Capucine L Grandjean
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florence Morin
- Université de Paris Cité, Hôpital Saint-Louis, AP-HP Nord, Laboratoire d'Immunologie, Paris, France
| | - Catherine Thieblemont
- Service d'Hémato-Oncologie, Hôpital Saint-Louis, AP-HP, Université de Paris Cité, Paris, France
| | - Sophie Caillat-Zucman
- Université de Paris Cité, Hôpital Saint-Louis, AP-HP Nord, Laboratoire d'Immunologie, Paris, France
- INSERM UMR976, Institut de Recherche St-Louis, Paris, France
| | - Philippe Bousso
- Institut Pasteur, Université de Paris Cité, INSERM U1223, Dynamics of Immune Responses Unit, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
23
|
Tolg C, Milojevic M, Qi FW, Pavanel HA, Locke MEO, Ma J, Price M, Nelson AC, McCarthy JB, Hill KA, Turley EA. RHAMM regulates MMTV-PyMT-induced lung metastasis by connecting STING-dependent DNA damage sensing to interferon/STAT1 pro-apoptosis signaling. Breast Cancer Res 2023; 25:74. [PMID: 37349798 PMCID: PMC10286489 DOI: 10.1186/s13058-023-01652-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/28/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND RHAMM is a multifunctional protein that is upregulated in breast tumors, and the presence of strongly RHAMM+ve cancer cell subsets associates with elevated risk of peripheral metastasis. Experimentally, RHAMM impacts cell cycle progression and cell migration. However, the RHAMM functions that contribute to breast cancer metastasis are poorly understood. METHODS We interrogated the metastatic functions of RHAMM using a loss-of-function approach by crossing the MMTV-PyMT mouse model of breast cancer susceptibility with Rhamm-/- mice. In vitro analyses of known RHAMM functions were performed using primary tumor cell cultures and MMTV-PyMT cell lines. Somatic mutations were identified using a mouse genotyping array. RNA-seq was performed to identify transcriptome changes resulting from Rhamm-loss, and SiRNA and CRISPR/Cas9 gene editing was used to establish cause and effect of survival mechanisms in vitro. RESULTS Rhamm-loss does not alter initiation or growth of MMTV-PyMT-induced primary tumors but unexpectedly increases lung metastasis. Increased metastatic propensity with Rhamm-loss is not associated with obvious alterations in proliferation, epithelial plasticity, migration, invasion or genomic stability. SNV analyses identify positive selection of Rhamm-/- primary tumor clones that are enriched in lung metastases. Rhamm-/- tumor clones are characterized by an increased ability to survive with ROS-mediated DNA damage, which associates with blunted expression of interferon pathway and target genes, particularly those implicated in DNA damage-resistance. Mechanistic analyses show that ablating RHAMM expression in breast tumor cells by siRNA knockdown or CRISPR-Cas9 gene editing blunts interferon signaling activation by STING agonists and reduces STING agonist-induced apoptosis. The metastasis-specific effect of RHAMM expression-loss is linked to microenvironmental factors unique to tumor-bearing lung tissue, notably high ROS and TGFB levels. These factors promote STING-induced apoptosis of RHAMM+ve tumor cells to a significantly greater extent than RHAMM-ve comparators. As predicted by these results, colony size of Wildtype lung metastases is inversely related to RHAMM expression. CONCLUSION RHAMM expression-loss blunts STING-IFN signaling, which offers growth advantages under specific microenvironmental conditions of lung tissue. These results provide mechanistic insight into factors controlling clonal survival/expansion of metastatic colonies and has translational potential for RHAMM expression as a marker of sensitivity to interferon therapy.
Collapse
Affiliation(s)
- Cornelia Tolg
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Maja Milojevic
- Departments of Biology, Western University, London, ON, Canada
| | - Freda W Qi
- Departments of Biology, Western University, London, ON, Canada
| | | | - M Elizabeth O Locke
- Departments of Biology, Western University, London, ON, Canada
- Departments of Computer Science, Western University, London, ON, Canada
| | - Jenny Ma
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Mathew Price
- Masonic Cancer Center, Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Andrew C Nelson
- Masonic Cancer Center, Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - James B McCarthy
- Masonic Cancer Center, Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kathleen A Hill
- Departments of Biology, Western University, London, ON, Canada.
- Departments of Computer Science, Western University, London, ON, Canada.
| | - Eva A Turley
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada.
- Departments of Biochemistry, Oncology and Surgery, Western University, London, ON, Canada.
| |
Collapse
|
24
|
Vredevoogd DW, Peeper DS. Heterogeneity in functional genetic screens: friend or foe? Front Immunol 2023; 14:1162706. [PMID: 37398651 PMCID: PMC10312307 DOI: 10.3389/fimmu.2023.1162706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Functional genetic screens to uncover tumor-intrinsic nodes of immune resistance have uncovered numerous mechanisms by which tumors evade our immune system. However, due to technical limitations, tumor heterogeneity is imperfectly captured with many of these analyses. Here, we provide an overview of the nature and sources of heterogeneity that are relevant for tumor-immune interactions. We argue that this heterogeneity may actually contribute to the discovery of novel mechanisms of immune evasion, given a sufficiently large and heterogeneous set of input data. Taking advantage of tumor cell heterogeneity, we provide proof-of-concept analyses of mechanisms of TNF resistance. Thus, consideration of tumor heterogeneity is imperative to increase our understanding of immune resistance mechanisms.
Collapse
|
25
|
Abstract
Song and Chow demonstrate that while tumor-intrinsic mutations in the IFN-γ signaling pathway confer immune resistance across in vitro co-culture systems, such alterations associate with enhanced anti-tumor immunity in vivo and improved responsiveness to immune checkpoint blockade therapy in patients with cancer.
Collapse
Affiliation(s)
- Eric Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Ophthalmology, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan D Chow
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Lee ND, Kaveh K, Bozic I. Clonal interactions in cancer: integrating quantitative models with experimental and clinical data. Semin Cancer Biol 2023; 92:61-73. [PMID: 37023969 DOI: 10.1016/j.semcancer.2023.04.002] [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: 11/30/2022] [Revised: 02/16/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Tumors consist of different genotypically distinct subpopulations-or subclones-of cells. These subclones can influence neighboring clones in a process called "clonal interaction." Conventionally, research on driver mutations in cancer has focused on their cell-autonomous effects that lead to an increase in fitness of the cells containing the driver. Recently, with the advent of improved experimental and computational technologies for investigating tumor heterogeneity and clonal dynamics, new studies have shown the importance of clonal interactions in cancer initiation, progression, and metastasis. In this review we provide an overview of clonal interactions in cancer, discussing key discoveries from a diverse range of approaches to cancer biology research. We discuss common types of clonal interactions, such as cooperation and competition, its mechanisms, and the overall effect on tumorigenesis, with important implications for tumor heterogeneity, resistance to treatment, and tumor suppression. Quantitative models-in coordination with cell culture and animal model experiments-have played a vital role in investigating the nature of clonal interactions and the complex clonal dynamics they generate. We present mathematical and computational models that can be used to represent clonal interactions and provide examples of the roles they have played in identifying and quantifying the strength of clonal interactions in experimental systems. Clonal interactions have proved difficult to observe in clinical data; however, several very recent quantitative approaches enable their detection. We conclude by discussing ways in which researchers can further integrate quantitative methods with experimental and clinical data to elucidate the critical-and often surprising-roles of clonal interactions in human cancers.
Collapse
Affiliation(s)
- Nathan D Lee
- Department of Applied Mathematics, University of Washington, Seattle, WA, United States of America
| | - Kamran Kaveh
- Department of Applied Mathematics, University of Washington, Seattle, WA, United States of America
| | - Ivana Bozic
- Department of Applied Mathematics, University of Washington, Seattle, WA, United States of America; Herbold Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America.
| |
Collapse
|
27
|
Wang Z, Liu Z, Wang S, Bing X, Ji X, He D, Han M, Wei Y, Wang C, Xia Q, Yang J, Gao J, Yin X, Wang Z, Shang Z, Xu J, Xin T, Liu Q. Implantation of hydrogel-liposome nanoplatform inhibits glioblastoma relapse by inducing ferroptosis. Asian J Pharm Sci 2023. [DOI: 10.1016/j.ajps.2023.100800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
|
28
|
Xu L, Gao X, Xing J, Guo Z. Identification of a necroptosis-related gene signature as a novel prognostic biomarker of cholangiocarcinoma. Front Immunol 2023; 14:1118816. [PMID: 36936916 PMCID: PMC10017743 DOI: 10.3389/fimmu.2023.1118816] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Background Cholangiocarcinoma (CHOL) is the most prevalent type of malignancy and the second most common form of primary liver cancer, resulting in high rates of morbidity and mortality. Necroptosis is a type of regulated cell death that appears to be involved in the regulation of several aspects of cancer biology, including tumorigenesis, metastasis, and cancer immunity. This study aimed to construct a necroptosis-related gene (NRG) signature to investigate the prognosis of CHOL patients using an integrated bioinformatics analysis. Methods CHOL patient data were acquired from the Gene Expression Omnibus (GEO) (GSE89748, GSE107943) and The Cancer Genome Atlas (TCGA) databases, with NRGs data from the necroptosis pathway in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Univariate and multivariate regression analyses were performed to establish the NRG signatures. Kaplan-Meier (KM) curves were used to evaluate the prognosis of patients with CHOL. Functional enrichment analysis was performed to identify key NRG-associated biological signaling pathways. We also applied integrative multi-omics analysis to the high- and low-risk score groups. Spearman's rank correlation was used to clarify the relationship between the NRG signature and immune infiltration. Results 65 differentially expressed (DE) NRGs were screened, five of which were selected to establish the prognostic signature of NRGS based on multivariate Cox regression analysis. We observed that low-risk patients survived significantly longer than high-risk patients. We found that patients with high-risk scores experienced higher immune cell infiltration, drug resistance, and more somatic mutations than patients with low-risk scores. We further found that sensitivities to GW843682X, mitomycin C, rapamycin, and S-trityl-L-cysteine were significantly higher in the low-risk group than in the high-risk group. Finally, we validated the expression of five NRGs in CHOL tissues using the TCGA database, HPA database and our clinical data. Conclusion These findings demonstrate that the five-NRG prognostic signature for CHOL patients is reasonably accurate and valid, and it may prove to be of considerable value for the treatment and prognosis of CHOL patients in the future.
Collapse
Affiliation(s)
- Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xueping Gao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Gaotanyan, Chongqing, China
| | - Jiyuan Xing
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhixian Guo
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Zhixian Guo,
| |
Collapse
|
29
|
Ma YL, Yang YF, Wang HC, Yang CC, Yan LJ, Ding ZN, Tian BW, Liu H, Xue JS, Han CL, Tan SY, Hong JG, Yan YC, Mao XC, Wang DX, Li T. A novel prognostic scoring model based on copper homeostasis and cuproptosis which indicates changes in tumor microenvironment and affects treatment response. Front Pharmacol 2023; 14:1101749. [PMID: 36909185 PMCID: PMC9998499 DOI: 10.3389/fphar.2023.1101749] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/13/2023] [Indexed: 03/14/2023] Open
Abstract
Background: Intracellular copper homeostasis requires a complex system. It has shown considerable prospects for intervening in the tumor microenvironment (TME) by regulating copper homeostasis and provoking cuproptosis. Their relationship with hepatocellular carcinoma (HCC) remains elusive. Methods: In TCGA and ICGC datasets, LASSO and multivariate Cox regression were applied to obtain the signature on the basis of genes associated with copper homeostasis and cuproptosis. Bioinformatic tools were utilized to reveal if the signature was correlated with HCC characteristics. Single-cell RNA sequencing data analysis identified differences in tumor and T cells' pathway activity and intercellular communication of immune-related cells. Real-time qPCR analysis was conducted to measure the genes' expression in HCC and adjacent normal tissue from 21 patients. CCK8 assay, scratch assay, transwell, and colony formation were conducted to reveal the effect of genes on in vitro cell proliferation, invasion, migration, and colony formation. Results: We constructed a five-gene scoring system in relation to copper homeostasis and cuproptosis. The high-risk score indicated poor clinical prognosis, enhanced tumor malignancy, and immune-suppressive tumor microenvironment. The T cell activity was markedly reduced in high-risk single-cell samples. The high-risk HCC patients had a better expectation of ICB response and reactivity to anti-PD-1 therapy. A total of 156 drugs were identified as potential signature-related drugs for HCC treatment, and most were sensitive to high-risk patients. Novel ligand-receptor pairs such as FASLG, CCL, CD40, IL2, and IFN-Ⅱ signaling pathways were revealed as cellular communication bridges, which may cause differences in TME and immune function. All crucial genes were differentially expressed between HCC and paired adjacent normal tissue. Model-constructed genes affected the phosphorylation of mTOR and AKT in both Huh7 and Hep3B cells. Knockdown of ZCRB1 impaired the proliferation, invasion, migration, and colony formation in HCC cell lines. Conclusion: We obtained a prognostic scoring system to forecast the TME changes and assist in choosing therapy strategies for HCC patients. In this study, we combined copper homeostasis and cuproptosis to show the overall potential risk of copper-related biological processes in HCC for the first time.
Collapse
Affiliation(s)
- Yun-Long Ma
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Ya-Fei Yang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Han-Chao Wang
- Institute for Financial Studies, Shandong University, Jinan, China
| | - Chun-Cheng Yang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Bao-Wen Tian
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Hui Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Jun-Shuai Xue
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Cheng-Long Han
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Si-Yu Tan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Jian-Guo Hong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yu-Chuan Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Xin-Cheng Mao
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Dong-Xu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China.,Department of hepatobiliary surgery, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
30
|
Seferbekova Z, Lomakin A, Yates LR, Gerstung M. Spatial biology of cancer evolution. Nat Rev Genet 2022; 24:295-313. [PMID: 36494509 DOI: 10.1038/s41576-022-00553-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2022] [Indexed: 12/13/2022]
Abstract
The natural history of cancers can be understood through the lens of evolution given that the driving forces of cancer development are mutation and selection of fitter clones. Cancer growth and progression are spatial processes that involve the breakdown of normal tissue organization, invasion and metastasis. For these reasons, spatial patterns are an integral part of histological tumour grading and staging as they measure the progression from normal to malignant disease. Furthermore, tumour cells are part of an ecosystem of tumour cells and their surrounding tumour microenvironment. A range of new spatial genomic, transcriptomic and proteomic technologies offers new avenues for the study of cancer evolution with great molecular and spatial detail. These methods enable precise characterizations of the tumour microenvironment, cellular interactions therein and micro-anatomical structures. In conjunction with spatial genomics, it emerges that tumours and microenvironments co-evolve, which helps explain observable patterns of heterogeneity and offers new routes for therapeutic interventions.
Collapse
|
31
|
DNA barcoding reveals ongoing immunoediting of clonal cancer populations during metastatic progression and immunotherapy response. Nat Commun 2022; 13:6539. [PMID: 36344500 PMCID: PMC9640547 DOI: 10.1038/s41467-022-34041-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/11/2022] [Indexed: 11/09/2022] Open
Abstract
Cancers evade the immune system through the process of cancer immunoediting. While immune checkpoint inhibitors are effective for reactivating tumour immunity in some cancer types, many other solid cancers, including breast cancer, remain largely non-responsive. Understanding how non-responsive cancers evade immunity and whether this occurs at the clonal level will improve immunotherapeutic design. Here we use DNA barcoding to track murine mammary cancer cell clones during immunoediting and determine clonal transcriptional profiles that allow immune evasion following anti-PD1 plus anti-CTLA4 immunotherapy. Clonal diversity is significantly restricted by immunotherapy treatment in both primary tumours and metastases, demonstrating selection for pre-existing breast cancer cell populations and ongoing immunoediting during metastasis and treatment. Immunotherapy resistant clones express a common gene signature associated with poor survival of basal-like breast cancer patient cohorts. At least one of these genes has an existing small molecule that can potentially be used to improve immunotherapy response.
Collapse
|
32
|
John J, Woolaver RA, Popolizio V, Chen SMY, Ge H, Krinsky AL, Vashisht M, Kramer Y, Chen Z, Wang JH. Divergent outcomes of anti-PD-L1 treatment coupled with host-intrinsic differences in TCR repertoire and distinct T cell activation states in responding versus non-responding tumors. Front Immunol 2022; 13:992630. [PMID: 36330507 PMCID: PMC9624473 DOI: 10.3389/fimmu.2022.992630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/03/2022] [Indexed: 12/24/2022] Open
Abstract
Differential responses to immune checkpoint inhibitors (ICI) may be attributed to tumor-intrinsic factors or environmental cues; however, these mechanisms cannot fully explain the variable ICI responses in different individuals. Here, we investigate the potential contribution of immunological heterogeneity with a focus on differences in T-cell receptor (TCR) repertoire to ICI responses, which has not been defined previously. To reveal additional factors underlying heterogeneous responses to ICI, we employed a squamous cell carcinoma (SCC) mouse model in which tumor-bearing recipients unambiguously diverged into responders (R) or non-responders (NR) upon anti-PD-L1 treatment. Treatment efficacy absolutely required CD8 T-cells and correlated positively with effector functions of CD8 tumor-infiltrating lymphocytes (TILs). We showed that TCR repertoires exhibited a similar magnitude of clonal expansion in R vs. NR CD8 TILs. However, the top expanded TCR clonotypes appeared to be mutually exclusive between R and NR CD8 TILs, which also occurred in a recipient-specific manner, demonstrating preferential expansion of distinct TCR clonotypes against the same SCC tumor. Unexpectedly, R vs. NR CD8 TILs reached all activation clusters and did not exhibit substantial global differences in transcriptomes. By linking single-cell transcriptomic data with unique TCR clonotypes, CD8 TILs harboring top TCR clonotypes were found to occupy distinct activation clusters and upregulate genes favoring anti-tumor immunity to different extents in R vs. NR. We conclude that stochastic differences in CD8 TIL TCR repertoire and distinct activation states of top TCR clonotypes may contribute to differential anti-PD-L1 responses. Our study suggests that host-intrinsic immunological heterogeneity may offer a new explanation for differential ICI responses in different individuals, which could impact on strategies for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Jessy John
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Rachel A. Woolaver
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Vince Popolizio
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Samantha M. Y. Chen
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Huaibin Ge
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Alexandra L. Krinsky
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Monika Vashisht
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yonatan Kramer
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO, United States
| | - Zhangguo Chen
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jing H. Wang
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Division of Hematology and Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
33
|
Nagel R, Pataskar A, Champagne J, Agami R. Boosting Antitumor Immunity with an Expanded Neoepitope Landscape. Cancer Res 2022; 82:3637-3649. [PMID: 35904353 PMCID: PMC9574376 DOI: 10.1158/0008-5472.can-22-1525] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/07/2022] [Accepted: 07/21/2022] [Indexed: 01/07/2023]
Abstract
Immune-checkpoint blockade therapy has been successfully applied to many cancers, particularly tumors that harbor a high mutational burden and consequently express a high abundance of neoantigens. However, novel approaches are needed to improve the efficacy of immunotherapy for treating tumors that lack a high load of classic genetically derived neoantigens. Recent discoveries of broad classes of nongenetically encoded and inducible neoepitopes open up new avenues for therapeutic development to enhance sensitivity to immunotherapies. In this review, we discuss recent work on neoantigen discovery, with an emphasis on novel classes of noncanonical neoepitopes.
Collapse
Affiliation(s)
- Remco Nagel
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Abhijeet Pataskar
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Julien Champagne
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Erasmus MC, Rotterdam University, Rotterdam, the Netherlands
| |
Collapse
|
34
|
Ribas A, Haining WN, Schumacher TNM. When Cancer Cells Become the Enablers of an Antitumor Immune Response. Cancer Discov 2022; 12:2244-2248. [PMID: 36196573 DOI: 10.1158/2159-8290.cd-22-0706] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor-specific cytotoxic T cells unleashed by the blockade of immune checkpoints have to overcome a hostile tumor microenvironment (TME). They start from very small numbers of T cells with tumor antigen specificity and, despite expansion, likely remain at a numerical disadvantage to the tumor cells they target. To overcome these obstacles, we propose that T cells need to change the TME to make it permissive for their antitumor effects by altering the phenotype of cells beyond the cancer cells they are in physical contact with. In this process, IFNγ secreted by tumor-specific T cells plays a critical role, as it changes the expression of hundreds of genes in cancer cells and other immune cells in the TME up to 40 layers of cells away from their location, effectively turning these cells into enablers of the antitumor immune response. In this perspective, we postulate that the clinical activity of cancer immunotherapy with immune-checkpoint blocking antibodies and adoptively transferred T cells requires that cancer cells facilitate the antitumor immune response. IFNγ effectively changes the balance of power in the TME to enable the antitumor activity of tumor antigen-specific cytotoxic T cells.
Collapse
Affiliation(s)
- Antoni Ribas
- Jonsson Comprehensive Cancer Center at the University of California, Los Angeles (UCLA), Los Angeles, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | | | - Ton N M Schumacher
- Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
35
|
Dubrot J, Du PP, Lane-Reticker SK, Kessler EA, Muscato AJ, Mehta A, Freeman SS, Allen PM, Olander KE, Ockerman KM, Wolfe CH, Wiesmann F, Knudsen NH, Tsao HW, Iracheta-Vellve A, Schneider EM, Rivera-Rosario AN, Kohnle IC, Pope HW, Ayer A, Mishra G, Zimmer MD, Kim SY, Mahapatra A, Ebrahimi-Nik H, Frederick DT, Boland GM, Haining WN, Root DE, Doench JG, Hacohen N, Yates KB, Manguso RT. In vivo CRISPR screens reveal the landscape of immune evasion pathways across cancer. Nat Immunol 2022; 23:1495-1506. [PMID: 36151395 DOI: 10.1038/s41590-022-01315-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/15/2022] [Indexed: 02/04/2023]
Abstract
The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.
Collapse
Affiliation(s)
- Juan Dubrot
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Peter P Du
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | - Arnav Mehta
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Samuel S Freeman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Peter M Allen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Clara H Wolfe
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Nelson H Knudsen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | - Ian C Kohnle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hans W Pope
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Austin Ayer
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gargi Mishra
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Sarah Y Kim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Dennie T Frederick
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve M Boland
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - W Nicholas Haining
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- ArsenalBio, South San Francisco, CA, USA
| | - David E Root
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen B Yates
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Robert T Manguso
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
36
|
Mathematical modeling for the combination treatment of IFN- γ and anti-PD-1 in cancer immunotherapy. Math Biosci 2022; 353:108911. [PMID: 36150452 DOI: 10.1016/j.mbs.2022.108911] [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: 03/05/2022] [Revised: 07/12/2022] [Accepted: 09/15/2022] [Indexed: 11/21/2022]
Abstract
When the immune-checkpoint programmed death-1 (PD-1) binds to its ligand programmed death ligand 1 (PD-L1) to form the complex PD-1-PD-L1, this complex inactivates immune cells resulting in cell apoptosis, downregulation of immune reaction, and tumor evasion. The antibody, anti-PD-1 or anti-PD-L1, blocks the PD-1-PD-L1 complex formation to restore the functions of T cells. Combination of anti-PD-1 with other treatment shows promising in different types of cancer treatments. Interferon-gamma (IFN-γ) plays an important role in immune responses. It is mainly regarded as a pro-inflammatory cytokine that promotes the proliferation of CD8+ T cell and cytotoxic T cell, enhances the activation of Th1 cells and CD8+ T cells, and enhances tumor elimination. However, recent studies have been discovering many anti-inflammatory functions of IFN-γ, such as promotion of the PD-L1 expression, T cell apoptosis, and tumor metastasis, as well as inhibition of the immune recognition and the killing rates by T cells. In this work, we construct a mathematical model incorporating pro-inflammatory and anti-inflammatory functions of IFN-γ to capture tumor growth under anti-PD-1 treatment in the wild type and IFN-γ null mutant melanoma. Our simulation results qualitatively fit experimental data that IFN-γ null mutant with anti-PD-1 obtains the highest tumor reduction comparing to IFN-γ null mutant without anti-PD-1 and wild type tumor with anti-PD-1 therapy. Moreover, our synergy analysis indicates that, in the combination treatment, the tumor volume decreases as either the dosage of anti-PD-1 increases or the IFN-γ production efficiency decreases. Thus, the combination of anti-PD-1 and IFN-γ blockade improves the tumor reduction comparing to the monotherapy of anti-PD-1 or the monotherapy of IFN-γ blockade. We also find a threshold curve of the minimal dosage of anti-PD-1 corresponding to the IFN-γ production efficiency to ensure the tumor reduction under the presence of IFN-γ.
Collapse
|
37
|
Atkins MB, Abu-Sbeih H, Ascierto PA, Bishop MR, Chen DS, Dhodapkar M, Emens LA, Ernstoff MS, Ferris RL, Greten TF, Gulley JL, Herbst RS, Humphrey RW, Larkin J, Margolin KA, Mazzarella L, Ramalingam SS, Regan MM, Rini BI, Sznol M. Maximizing the value of phase III trials in immuno-oncology: A checklist from the Society for Immunotherapy of Cancer (SITC). J Immunother Cancer 2022; 10:jitc-2022-005413. [PMID: 36175037 PMCID: PMC9528604 DOI: 10.1136/jitc-2022-005413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2022] [Indexed: 11/03/2022] Open
Abstract
The broad activity of agents blocking the programmed cell death protein 1 and its ligand (the PD-(L)1 axis) revolutionized oncology, offering long-term benefit to patients and even curative responses for tumors that were once associated with dismal prognosis. However, only a minority of patients experience durable clinical benefit with immune checkpoint inhibitor monotherapy in most disease settings. Spurred by preclinical and correlative studies to understand mechanisms of non-response to the PD-(L)1 antagonists and by combination studies in animal tumor models, many drug development programs were designed to combine anti-PD-(L)1 with a variety of approved and investigational chemotherapies, tumor-targeted therapies, antiangiogenic therapies, and other immunotherapies. Several immunotherapy combinations improved survival outcomes in a variety of indications including melanoma, lung, kidney, and liver cancer, among others. This immunotherapy renaissance, however, has led to many combinations being advanced to late-stage development without definitive predictive biomarkers, limited phase I and phase II data, or clinical trial designs that are not optimized for demonstrating the unique attributes of immune-related antitumor activity-for example, landmark progression-free survival and overall survival. The decision to activate a study at an individual site is investigator-driven, and generalized frameworks to evaluate the potential for phase III trials in immuno-oncology to yield positive data, particularly to increase the number of curative responses or otherwise advance the field have thus far been lacking. To assist in evaluating the potential value to patients and the immunotherapy field of phase III trials, the Society for Immunotherapy of Cancer (SITC) has developed a checklist for investigators, described in this manuscript. Although the checklist focuses on anti-PD-(L)1-based combinations, it may be applied to any regimen in which immune modulation is an important component of the antitumor effect.
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia, USA
| | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione "G Pascale", Napoli, Italy
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, Illinois, USA
| | - Daniel S Chen
- Engenuity Life Sciences, Burlingame, California, USA
| | - Madhav Dhodapkar
- Center for Cancer Immunology, Winship Cancer Institute at Emory University, Atlanta, Georgia, USA
| | - Leisha A Emens
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Marc S Ernstoff
- DCTD/DTP-IOB, ImmunoOncology Branch, NCI, Bethesda, Maryland, USA
| | | | - Tim F Greten
- Gastrointestinal Malignancies Section, National Cancer Institue CCR Liver Program, Bethesda, Maryland, USA
| | - James L Gulley
- Center for Immuno-Oncology, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | - Kim A Margolin
- St. John's Cancer Institute, Santa Monica, California, USA
| | - Luca Mazzarella
- Experimental Oncology, New Drug Development, European Instititue of Oncology IRCCS, Milan, Italy
| | | | - Meredith M Regan
- Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Mario Sznol
- Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Khatib SA, Ma L, Dang H, Forgues M, Chung JY, Ylaya K, Hewitt SM, Chaisaingmongkol J, Rucchirawat M, Wang XW. Single-cell biology uncovers apoptotic cell death and its spatial organization as a potential modifier of tumor diversity in HCC. Hepatology 2022; 76:599-611. [PMID: 35034369 DOI: 10.1002/hep.32345] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS HCC is a highly aggressive and heterogeneous cancer type with limited treatment options. Identifying drivers of tumor heterogeneity may lead to better therapeutic options and favorable patient outcomes. We investigated whether apoptotic cell death and its spatial architecture is linked to tumor molecular heterogeneity using single-cell in situ hybridization analysis. APPROACH AND RESULTS We analyzed 254 tumor samples from two HCC cohorts using tissue microarrays. We developed a mathematical model to quantify cellular diversity among HCC samples using two tumor markers, cyclin-dependent kinase inhibitor 3 and protein regulator of cytokinesis 1 as surrogates for heterogeneity and caspase 3 (CASP3) as an apoptotic cell death marker. We further explored the impact of potential dying-cell hubs on tumor cell diversity and patient outcome by density contour mapping and spatial proximity analysis. We also developed a selectively controlled in vitro model of cell death using CRISPR/CRISPR-associated 9 to determine therapy response and growth under hypoxic conditions. We found that increasing levels of CASP3+ tumor cells are associated with higher tumor diversity. Interestingly, we discovered regions of densely populated CASP3+ , which we refer to as CASP3+ cell islands, in which the nearby cellular heterogeneity was found to be the greatest compared to cells farther away from these islands and that this phenomenon was associated with survival. Additionally, cell culture experiments revealed that higher levels of cell death, accompanied by increased CASP3 expression, led to greater therapy resistance and growth under hypoxia. CONCLUSIONS These results are consistent with the hypothesis that increased apoptotic cell death may lead to greater tumor heterogeneity and thus worse patient outcomes.
Collapse
Affiliation(s)
- Subreen A Khatib
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Department of Tumor Biology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Hien Dang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Division of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Marshonna Forgues
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Joon-Yong Chung
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Kris Ylaya
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jittporn Chaisaingmongkol
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, Thailand.,Center of Excellence on Environmental Health and Toxicology, Office of the Higher Education Commission, Ministry of Education, Bangkok, Thailand
| | - Mathuros Rucchirawat
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, Thailand.,Center of Excellence on Environmental Health and Toxicology, Office of the Higher Education Commission, Ministry of Education, Bangkok, Thailand
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.,Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
39
|
Shen H, Huang F, Zhang X, Ojo OA, Li Y, Trummell HQ, Anderson JC, Fiveash J, Bredel M, Yang ES, Willey CD, Chong Z, Bonner JA, Shi LZ. Selective suppression of melanoma lacking IFN-γ pathway by JAK inhibition depends on T cells and host TNF signaling. Nat Commun 2022; 13:5013. [PMID: 36008408 PMCID: PMC9411168 DOI: 10.1038/s41467-022-32754-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/16/2022] [Indexed: 11/09/2022] Open
Abstract
Therapeutic resistance to immune checkpoint blockers (ICBs) in melanoma patients is a pressing issue, of which tumor loss of IFN-γ signaling genes is a major underlying mechanism. However, strategies of overcoming this resistance mechanism have been largely elusive. Moreover, given the indispensable role of tumor-infiltrating T cells (TILs) in ICBs, little is known about how tumor-intrinsic loss of IFN-γ signaling (IFNγR1KO) impacts TILs. Here, we report that IFNγR1KO melanomas have reduced infiltration and function of TILs. IFNγR1KO melanomas harbor a network of constitutively active protein tyrosine kinases centered on activated JAK1/2. Mechanistically, JAK1/2 activation is mediated by augmented mTOR. Importantly, JAK1/2 inhibition with Ruxolitinib selectively suppresses the growth of IFNγR1KO but not scrambled control melanomas, depending on T cells and host TNF. Together, our results reveal an important role of tumor-intrinsic IFN-γ signaling in shaping TILs and manifest a targeted therapy to bypass ICB resistance of melanomas defective of IFN-γ signaling.
Collapse
Affiliation(s)
- Hongxing Shen
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
| | - Fengyuan Huang
- Department of Genetics and Informatics Institute, UAB-SOM, Birmingham, AL, USA
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Oluwagbemiga A Ojo
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
| | - Yuebin Li
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
| | - Hoa Quang Trummell
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
| | - Joshua C Anderson
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
| | - John Fiveash
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA
| | - Markus Bredel
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA
| | - Eddy S Yang
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA
| | - Christopher D Willey
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA
| | - Zechen Chong
- Department of Genetics and Informatics Institute, UAB-SOM, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA.
| | - James A Bonner
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA.
| | - Lewis Zhichang Shi
- Department of Radiation Oncology, Heersink School of Medicine, University of Alabama at Birmingham (UAB-SOM), Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, UAB-SOM, Birmingham, AL, USA.
- Department of Microbiology, UAB-SOM, Birmingham, AL, USA.
- Department of Pharmacology and Toxicology, UAB-SOM, Birmingham, AL, USA.
- Programs in Immunology, UAB-SOM, Birmingham, AL, USA.
| |
Collapse
|
40
|
Barkley D, Moncada R, Pour M, Liberman DA, Dryg I, Werba G, Wang W, Baron M, Rao A, Xia B, França GS, Weil A, Delair DF, Hajdu C, Lund AW, Osman I, Yanai I. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat Genet 2022; 54:1192-1201. [PMID: 35931863 PMCID: PMC9886402 DOI: 10.1038/s41588-022-01141-9] [Citation(s) in RCA: 226] [Impact Index Per Article: 75.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
Transcriptional heterogeneity among malignant cells of a tumor has been studied in individual cancer types and shown to be organized into cancer cell states; however, it remains unclear to what extent these states span tumor types, constituting general features of cancer. Here, we perform a pan-cancer single-cell RNA-sequencing analysis across 15 cancer types and identify a catalog of gene modules whose expression defines recurrent cancer cell states including 'stress', 'interferon response', 'epithelial-mesenchymal transition', 'metal response', 'basal' and 'ciliated'. Spatial transcriptomic analysis linked the interferon response in cancer cells to T cells and macrophages in the tumor microenvironment. Using mouse models, we further found that induction of the interferon response module varies by tumor location and is diminished upon elimination of lymphocytes. Our work provides a framework for studying how cancer cell states interact with the tumor microenvironment to form organized systems capable of immune evasion, drug resistance and metastasis.
Collapse
Affiliation(s)
- Dalia Barkley
- Institute for Computational Medicine, New York, NY, USA
| | | | - Maayan Pour
- Institute for Computational Medicine, New York, NY, USA
| | | | - Ian Dryg
- Department of Dermatology, NYU School of Medicine, New York, NY, USA
| | - Gregor Werba
- Department of Surgery, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Wei Wang
- Department of Surgery, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Maayan Baron
- Institute for Computational Medicine, New York, NY, USA
| | - Anjali Rao
- Institute for Computational Medicine, New York, NY, USA
| | - Bo Xia
- Institute for Computational Medicine, New York, NY, USA
| | | | - Alejandro Weil
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | | | - Cristina Hajdu
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Amanda W. Lund
- Department of Dermatology, NYU School of Medicine, New York, NY, USA,Department of Surgery, NYU School of Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA
| | - Iman Osman
- Department of Dermatology, NYU School of Medicine, New York, NY, USA,Department of Pathology, NYU School of Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA
| | - Itai Yanai
- Institute for Computational Medicine, New York, NY, USA,Perlmutter Cancer Center NYU School of Medicine, New York, NY, USA,Corresponding author:
| |
Collapse
|
41
|
Xu Y, Chen C, Guo Y, Hu S, Sun Z. Effect of CRISPR/Cas9-Edited PD-1/PD-L1 on Tumor Immunity and Immunotherapy. Front Immunol 2022; 13:848327. [PMID: 35300341 PMCID: PMC8920996 DOI: 10.3389/fimmu.2022.848327] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR-associated nuclease9 (CRISPR/Cas9) gene editing technology implements precise programming of the human genome through RNA guidance. At present, it has been widely used in the construction of animal tumor models, the study of drug resistance regulation mechanisms, epigenetic control and innovation in cancer treatment. Tumor immunotherapy restores the normal antitumor immune response by restarting and maintaining the tumor-immune cycle. CRISPR/Cas9 technology has occupied a central position in further optimizing anti-programmed cell death 1(PD-1) tumor immunotherapy. In this review, we summarize the recent progress in exploring the regulatory mechanism of tumor immune PD-1 and programmed death ligand 1(PD-L1) based on CRISPR/Cas9 technology and its clinical application in different cancer types. In addition, CRISPR genome-wide screening identifies new drug targets and biomarkers to identify potentially sensitive populations for anti-PD-1/PD-L1 therapy and maximize antitumor effects. Finally, the strong potential and challenges of CRISPR/Cas9 for future clinical applications are discussed.
Collapse
Affiliation(s)
- Yanxin Xu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaxin Guo
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Wang XR, Jiang ZB, Xu C, Meng WY, Liu P, Zhang YZ, Xie C, Xu JY, Xie YJ, Liang TL, Yan HX, Fan XX, Yao XJ, Wu QB, Leung ELH. Andrographolide suppresses non-small-cell lung cancer progression through induction of autophagy and antitumor immune response. Pharmacol Res 2022; 179:106198. [DOI: 10.1016/j.phrs.2022.106198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
43
|
Zhang A, Miao K, Sun H, Deng CX. Tumor heterogeneity reshapes the tumor microenvironment to influence drug resistance. Int J Biol Sci 2022; 18:3019-3033. [PMID: 35541919 PMCID: PMC9066118 DOI: 10.7150/ijbs.72534] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 11/18/2022] Open
Abstract
Tumor heterogeneity is one of the hallmarks of cancer and a challenge in the field of oncology. Tumor heterogeneity is the main cause of drug resistance, leading to therapeutic failure. Mechanically, tumor heterogeneity either directly affects therapeutic targets or shapes the tumor microenvironment (TME) by defining transcriptomic and phenotypic profiles to influence drug resistance. Tumor heterogeneity evolves spatially and temporally during tumor development, leading to the constant reprogramming of the TME. Advances in molecular profiling technologies and precision oncology platforms have allowed us to uncover the impact of tumor heterogeneity on drug resistance in the context of the TME. In this review, we focus on the processes during which genomic mutations drive tumor heterogeneity and the mechanisms through which tumor heterogeneity reprograms the TME to affect drug resistance and patient prognosis.
Collapse
Affiliation(s)
- Aiping Zhang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Heng Sun
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
- Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| |
Collapse
|
44
|
Kalaora S, Nagler A, Wargo JA, Samuels Y. Mechanisms of immune activation and regulation: lessons from melanoma. Nat Rev Cancer 2022; 22:195-207. [PMID: 35105962 DOI: 10.1038/s41568-022-00442-9] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
Melanoma, a skin cancer that develops from pigment cells, has been studied intensively, particularly in terms of the immune response to tumours, and has been used as a model for the development of immunotherapy. This is due, in part, to the high mutational burden observed in melanomas, which increases both their immunogenicity and the infiltration of immune cells into the tumours, compared with other types of cancers. The immune response to melanomas involves a complex set of components and interactions. As the tumour evolves, it accumulates an increasing number of genetic and epigenetic alterations, some of which contribute to the immunogenicity of the tumour cells and the infiltration of immune cells. However, tumour evolution also enables the development of resistance mechanisms, which, in turn, lead to tumour immune escape. Understanding the interactions between melanoma tumour cells and the immune system, and the evolving changes within the melanoma tumour cells, the immune system and the microenvironment, is essential for the development of new cancer therapies. However, current research suggests that other extrinsic factors, such as the microbiome, may play a role in the immune response to melanomas. Here, we review the mechanisms underlying the immune response in the tumour and discuss recent advances as well as strategies for treatment development.
Collapse
Affiliation(s)
- Shelly Kalaora
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Nagler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
45
|
Lin YN, Schmidt MO, Sharif GM, Vietsch EE, Kiliti AJ, Barefoot ME, Riegel AT, Wellstein A. Impaired CXCL12 signaling contributes to resistance of pancreatic cancer subpopulations to T cell-mediated cytotoxicity. Oncoimmunology 2022; 11:2027136. [PMID: 35127250 PMCID: PMC8816404 DOI: 10.1080/2162402x.2022.2027136] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
Pancreatic cancer remains largely unresponsive to immune modulatory therapy attributable in part to an immunosuppressive, desmoplastic tumor microenvironment. Here, we analyze mechanisms of cancer cell-autonomous resistance to T cells. We used a 3D co-culture model of cancer cell spheroids from the KPC (LSL-KrasG12D/+ /LSL-Trp53R172H/+ /p48-Cre) pancreatic ductal adenocarcinoma (PDAC) model, to examine interactions with tumor-educated T cells isolated from draining lymph nodes of PDAC-bearing mice. Subpopulations of cancer cells resistant to these tumor-educated T cells were isolated from the in vitro co-culture and their properties compared with sensitive cancer cells. In co-culture with resistant cancer cell subpopulations, tumor-educated T cells showed reduced effector T cell functionality, reduced infiltration into tumor cell spheroids and decreased induction of apoptosis. A combination of comparative transcriptomic analyses, cytometric and immunohistochemistry techniques allowed us to dissect the role of differential gene expression and signaling pathways between sensitive and resistant cells. A decreased expression of the chemokine CXCL12 (SDF-1) was revealed as a common feature in the resistant cell subpopulations. Adding back CXCL12 reversed the resistant phenotype and was inhibited by the CXCR4 inhibitor AMD3100 (plerixafor). We conclude that reduced CXCL12 signaling contributes to PDAC subpopulation resistance to T cell-mediated attack.
Collapse
Affiliation(s)
- Yuan-Na Lin
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Ghada M. Sharif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Eveline E. Vietsch
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
- Department of Surgery, Erasmus Mc, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Amber J. Kiliti
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Megan E. Barefoot
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anna T. Riegel
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
46
|
Xu SM, Shi CJ, Xia RH, Wang LZ, Tian Z, Ye WM, Liu L, Liu SL, Zhang CY, Hu YH, Zhou R, Han Y, Wang Y, Zhang ZY, Li J. Analysis of Immunological Characteristics and Genomic Alterations in HPV-Positive Oropharyngeal Squamous Cell Carcinoma Based on PD-L1 Expression. Front Immunol 2022; 12:798424. [PMID: 35145511 PMCID: PMC8821172 DOI: 10.3389/fimmu.2021.798424] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) expression has been approved as an immune checkpoint inhibitor (ICI) response predictive biomarker; however, the clinicopathological and molecular features of HPV-positive oropharyngeal squamous cell carcinoma [HPV(+)OPSCC] based on PD-L1 expression are not well studied. We aimed to characterize clinicopathological, tumor immune microenvironmental, and molecular features of HPV(+)OPSCC with different PD-L1 expression scored by combined positive score (CPS). A total of 112 cases were collected from 2008-2021 and received PD-L1 and CD8 immunohistochemistry (IHC) staining. 71 samples received DNA sequencing out of which 32 samples received RNA sequencing for immune-related gene alterations or expression analysis. The 32 samples were also subjected to analysis of CD20, CD4, CD8, CD68, Foxp3 and P16 by multiplex immunofluorescence (mIF) staining, and the immune markers were evaluated in the tumor body (TB), tumor margin (TM) and normal stroma (NS) regions separately. Our results showed that of 112 HPV(+)OPSCC tumors, high(CPS≥20), intermediate(1≤CPS<20), and low(CPS<1) PD-L1 expression was seen in 29.5%, 43.8% and 26.8% cases respectively. Non-smoking patients and patients with tumors occurring at the tonsils or having rich lymphocytes infiltration had significantly higher PD-L1 expression. Patients with CPS≥20 had significantly higher tumor mutation burden (TMB, p=0.0058), and PD-L1 expression correlated significantly with CD8+ T cells infiltration, which were ample in tumor regions than in NS in mIF. CD20+, CD4+, CD68+, Foxp3+CD4+ cells were demonstrated to infiltrate higher in TM while CD20+ and CD68+ cells were also enriched in NS and TB regions respectively. However, none of them showed correlations with PD-L1 expression. ARID1A, STK11 alterations were enriched in the low PD-L1 group significantly, while anti-viral immune associated APOBEC mutation signature and immune-related genes expression such as XCL1 and IL11 were positively associated with PD-L1 expression (p<0.05). This is a comprehensive investigation revealing immune and molecular features of HPV(+)OPSCC based on PD-L1 expression. Our study suggested that 73.2% of HPV(+)OPSCC patients may benefit from immunotherapy, and high PD-L1 expression reflects immune-active status of HPV(+)OPSCC accompanied by higher immune effect factors such as TMB, CD8+ cytotoxic T cells and immune-related genomic alterations. Our study offers valuable information for understanding the immune features of HPV(+)OPSCC.
Collapse
Affiliation(s)
- Sheng-ming Xu
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Chao-ji Shi
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Rong-hui Xia
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-zhen Wang
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Tian
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-min Ye
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Liu Liu
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shu-li Liu
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Chun-ye Zhang
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-hua Hu
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhou
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yong Han
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yu Wang
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-yuan Zhang
- Department of Oral and Maxillofacial-Head Neck Oncology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- *Correspondence: Jiang Li, ; Zhi-yuan Zhang,
| | - Jiang Li
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Pathology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jiang Li, ; Zhi-yuan Zhang,
| |
Collapse
|
47
|
Sun T, Xu J, Chen T, Tu C, Zhu L, Yan D. Self-amplified ROS-responsive chemodrug-inhibitor conjugate for multi-drug resistance tumor therapy. Biomater Sci 2022; 10:997-1007. [DOI: 10.1039/d1bm01605a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
P-glycoprotein (P-gp) overexpression caused multidrug resistance (MDR) is a main reason for the failure of cancer chemotherapy. The combined delivery of chemodrug and P-gp inhibitor is a promising pathway to...
Collapse
|
48
|
|
49
|
Lu S, Peng X, Zeng S, Deng H, Feng Z, Zeng Q, Cheng X, Hu J, Ye Z, Li M, Xu X, Lin G, Hu C. Grass carp (Ctenopharyngodon idellus) PIAS1 inhibits innate immune response via interacting with STAT1. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 125:104216. [PMID: 34331975 DOI: 10.1016/j.dci.2021.104216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 06/13/2023]
Abstract
Protein inhibitor of activated signal transducer and activator of transcription (PIAS) family protein involved in gene transcriptional regulation acts as negative regulator in Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathway. But until now, the roles of PIAS in fish are not clear. In this study, we identified the two mammalian PIAS1 orthologs from Ctenopharyngodon idellus, namely CiPIAS1a and CiPIAS1b, respectively. They can respond to the stimulation from Polyribocytidylic acid (Poly I:C), Grass Carp Reovirus (GCRV) and Lipopolysaccharides (LPS) respectively, so we suggested that they could participate in interferon (IFN)-mediated antiviral and antibacterial immune response. The subcellular localization and nuclear cytoplasm extraction showed that CiPIAS1a and CiPIAS1b were mainly distributed in the nucleus. In addition, Co-IP showed that they separately inhibited the phosphorylation of STAT1 via interacting with it, which leads to the reduction of IFN1 expression.
Collapse
Affiliation(s)
- Shina Lu
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xiaojue Peng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Shanshan Zeng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Hang Deng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Zhiqing Feng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Qing Zeng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xining Cheng
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Jihuan Hu
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Zuocheng Ye
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Meifeng Li
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xiaowen Xu
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Gang Lin
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China.
| | - Chengyu Hu
- School of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
50
|
Song TY, Long M, Zhao HX, Zou MW, Fan HJ, Liu Y, Geng CL, Song MF, Liu YF, Chen JY, Yang YL, Zhou WR, Huang DW, Peng B, Peng ZG, Cang Y. Tumor evolution selectively inactivates the core microRNA machinery for immune evasion. Nat Commun 2021; 12:7003. [PMID: 34853298 PMCID: PMC8636623 DOI: 10.1038/s41467-021-27331-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer cells acquire genetic heterogeneity to escape from immune surveillance during tumor evolution, but a systematic approach to distinguish driver from passenger mutations is lacking. Here we investigate the impact of different immune pressure on tumor clonal dynamics and immune evasion mechanism, by combining massive parallel sequencing of immune edited tumors and CRISPR library screens in syngeneic mouse tumor model and co-culture system. We find that the core microRNA (miRNA) biogenesis and targeting machinery maintains the sensitivity of cancer cells to PD-1-independent T cell-mediated cytotoxicity. Genetic inactivation of the machinery or re-introduction of ANKRD52 frequent patient mutations dampens the JAK-STAT-interferon-γ signaling and antigen presentation in cancer cells, largely by abolishing miR-155-targeted silencing of suppressor of cytokine signaling 1 (SOCS1). Expression of each miRNA machinery component strongly correlates with intratumoral T cell infiltration in nearly all human cancer types. Our data indicate that the evolutionarily conserved miRNA pathway can be exploited by cancer cells to escape from T cell-mediated elimination and immunotherapy.
Collapse
Affiliation(s)
- Tian-Yu Song
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min Long
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hai-Xin Zhao
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Miao-Wen Zou
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong-Jie Fan
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yang Liu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen-Lu Geng
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min-Fang Song
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yu-Feng Liu
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jun-Yi Chen
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu-Lin Yang
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Wen-Rong Zhou
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Da-Wei Huang
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Bo Peng
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhen-Gang Peng
- Oncology and Immunology Unit, WuXi Biology, WuXi AppTec (Shanghai) Co, Ltd, Shanghai, China
| | - Yong Cang
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|