1
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2024:10.1038/s41568-024-00761-z. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
2
|
Xu L, Cao P, Wang J, Zhang P, Hu S, Cheng C, Wang H. IL-22: A key inflammatory mediator as a biomarker and potential therapeutic target for lung cancer. Heliyon 2024; 10:e35901. [PMID: 39263114 PMCID: PMC11387261 DOI: 10.1016/j.heliyon.2024.e35901] [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: 08/13/2023] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024] Open
Abstract
Lung cancer, one of the most prevalent cancers worldwide, stands as the primary cause of cancer-related deaths. As is well-known, the utmost crucial risk factor contributing to lung cancer is smoking. In recent years, remarkable progress has been made in treating lung cancer, particularly non-small cell lung cancer (NSCLC). Nevertheless, the absence of effective and accurate biomarkers for diagnosing and treating lung cancer remains a pressing issue. Interleukin 22 (IL-22) is a member of the IL-10 cytokine family. It exerts biological functions (including induction of proliferation and anti-apoptotic signaling pathways, enhancement of tissue regeneration and immunity defense) by binding to heterodimeric receptors containing type 1 receptor chain (R1) and type 2 receptor chain (R2). IL-22 has been identified as a pro-cancer factor since dysregulation of the IL-22-IL-22R system has been implicated in the development of different cancers, including lung, breast, gastric, pancreatic, and colon cancers. In this review, we discuss the differential expression, regulatory role, and potential clinical significance of IL-22 in lung cancer, while shedding light on innovative approaches for the future.
Collapse
Affiliation(s)
- Ling Xu
- Department of Interventional Pulmonary Diseases, The Anhui Chest Hospital, Hefei, China
| | - Peng Cao
- Department of Interventional Pulmonary Diseases, The Anhui Chest Hospital, Hefei, China
| | - Jianpeng Wang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Peng Zhang
- Department of Interventional Pulmonary Diseases, The Anhui Chest Hospital, Hefei, China
| | - Shuhui Hu
- Department of Interventional Pulmonary Diseases, The Anhui Chest Hospital, Hefei, China
| | - Chao Cheng
- Department of Interventional Pulmonary Diseases, The Anhui Chest Hospital, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Zhang Z, Chen Y, Pan X, Li P, Ren Z, Wang X, Chen Y, Shen S, Wang T, Lin A. IL-1β mediates Candida tropicalis-induced immunosuppressive function of MDSCs to foster colorectal cancer. Cell Commun Signal 2024; 22:408. [PMID: 39164774 PMCID: PMC11337875 DOI: 10.1186/s12964-024-01771-y] [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: 04/23/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND There is increasing evidence that gut fungi dysbiosis plays a crucial role in the development and progression of colorectal cancer (CRC). It has been reported that gut fungi exacerbate the severity of CRC by regulating tumor immunity. Our previous studies have shown that the opportunistic pathogenic fungal pathogen, Candida tropicalis (C. tropicalis) promotes CRC progression by enhancing the immunosuppressive function of MDSCs and activating the NLRP3 inflammasome of MDSCs. However, the relationship between IL-1β produced by NLRP3 inflammasome activation and the immunosuppressive function of MDSCs enhanced by C. tropicalis in CRC remains unclear. METHODS The TCGA database was used to analyze the relationship between IL-1β and genes related to immunosuppressive function of MDSCs in human CRC. The expression of IL-1β in human CRC tissues was detected by immunofluorescence staining. The proteomic analysis was performed on the culture supernatant of C. tropicalis-stimulated MDSCs. The experiments of supplementing and blocking IL-1β as well as inhibiting the NLRP3 inflammasome activation were conducted. A mouse colon cancer xenograft model was established by using MC38 colon cancer cell line. RESULTS Analysis of CRC clinical samples showed that the high expression of IL-1β was closely related to the immunosuppressive function of tumor-infiltrated MDSCs. The results of in vitro experiments revealed that IL-1β was the most secreted cytokine of MDSCs stimulated by C. tropicalis. In vitro supplementation of IL-1β further enhanced the immunosuppressive function of C. tropicalis-stimulated MDSCs and NLRP3-IL-1β axis mediated the immunosuppressive function of MDSCs enhanced by C. tropicalis. Finally, blockade of IL-1β secreted by MDSCs augmented antitumor immunity and mitigated C. tropicalis-associated colon cancer. CONCLUSIONS C. tropicalis promotes excessive secretion of IL-1β from MDSCs via the NLRP3 inflammasome. IL-1β further enhances the immunosuppressive function of MDSCs to inhibit antitumor immunity, thus promoting the progression of CRC. Therefore, targeting IL-1β secreted by MDSCs may be a potential immunotherapeutic strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Zhiyong Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Ying Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xinyi Pan
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Pengfei Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Zhengqian Ren
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Xiuzhu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuxi Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
| | - Aihua Lin
- Department of Critical Care Medicine, Sucheng District, Suqian Hospital of Nanjing Drum Tower Hospital Group, 138 Huanghe South Road, Suqian City, China.
| |
Collapse
|
4
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [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: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
5
|
Benitez DA, Cumplido-Laso G, Olivera-Gómez M, Del Valle-Del Pino N, Díaz-Pizarro A, Mulero-Navarro S, Román-García A, Carvajal-Gonzalez JM. p53 Genetics and Biology in Lung Carcinomas: Insights, Implications and Clinical Applications. Biomedicines 2024; 12:1453. [PMID: 39062026 PMCID: PMC11274425 DOI: 10.3390/biomedicines12071453] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The TP53 gene is renowned as a tumor suppressor, playing a pivotal role in overseeing the cell cycle, apoptosis, and maintaining genomic stability. Dysregulation of p53 often contributes to the initiation and progression of various cancers, including lung cancer (LC) subtypes. The review explores the intricate relationship between p53 and its role in the development and progression of LC. p53, a crucial tumor suppressor protein, exists in various isoforms, and understanding their distinct functions in LC is essential for advancing our knowledge of this deadly disease. This review aims to provide a comprehensive literature overview of p53, its relevance to LC, and potential clinical applications.
Collapse
Affiliation(s)
- Dixan A. Benitez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| | | | | | | | | | | | | | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| |
Collapse
|
6
|
Alanazi M, Weng T, McLeod L, Gearing LJ, Smith JA, Kumar B, Saad MI, Jenkins BJ. Cytosolic DNA sensor AIM2 promotes KRAS-driven lung cancer independent of inflammasomes. Cancer Sci 2024; 115:1834-1850. [PMID: 38594840 PMCID: PMC11145135 DOI: 10.1111/cas.16171] [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: 08/17/2023] [Revised: 02/10/2024] [Accepted: 03/23/2024] [Indexed: 04/11/2024] Open
Abstract
Constitutively active KRAS mutations are among the major drivers of lung cancer, yet the identity of molecular co-operators of oncogenic KRAS in the lung remains ill-defined. The innate immune cytosolic DNA sensor and pattern recognition receptor (PRR) Absent-in-melanoma 2 (AIM2) is best known for its assembly of multiprotein inflammasome complexes and promoting an inflammatory response. Here, we define a role for AIM2, independent of inflammasomes, in KRAS-addicted lung adenocarcinoma (LAC). In genetically defined and experimentally induced (nicotine-derived nitrosamine ketone; NNK) LAC mouse models harboring the KrasG12D driver mutation, AIM2 was highly upregulated compared with other cytosolic DNA sensors and inflammasome-associated PRRs. Genetic ablation of AIM2 in KrasG12D and NNK-induced LAC mouse models significantly reduced tumor growth, coincident with reduced cellular proliferation in the lung. Bone marrow chimeras suggest a requirement for AIM2 in KrasG12D-driven LAC in both hematopoietic (immune) and non-hematopoietic (epithelial) cellular compartments, which is supported by upregulated AIM2 expression in immune and epithelial cells of mutant KRAS lung tissues. Notably, protection against LAC in AIM2-deficient mice is associated with unaltered protein levels of mature Caspase-1 and IL-1β inflammasome effectors. Moreover, genetic ablation of the key inflammasome adapter, ASC, did not suppress KrasG12D-driven LAC. In support of these in vivo findings, AIM2, but not mature Caspase-1, was upregulated in human LAC patient tumor biopsies. Collectively, our findings reveal that endogenous AIM2 plays a tumor-promoting role, independent of inflammasomes, in mutant KRAS-addicted LAC, and suggest innate immune DNA sensing may provide an avenue to explore new therapeutic strategies in lung cancer.
Collapse
Affiliation(s)
- Mohammad Alanazi
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Teresa Weng
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Linden J. Gearing
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Julian A. Smith
- Department of Surgery, School of Clinical Sciences/Monash HealthMonash UniversityClaytonVictoriaAustralia
| | - Beena Kumar
- Department of Anatomical PathologyMonash HealthClaytonVictoriaAustralia
| | - Mohamed I. Saad
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Brendan J. Jenkins
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
- South Australian immunoGENomics Cancer Institute (SAiGENCI)The University of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
7
|
Jiang B, Zhang W, Zhang X, Sun Y. Targeting senescent cells to reshape the tumor microenvironment and improve anticancer efficacy. Semin Cancer Biol 2024; 101:58-73. [PMID: 38810814 DOI: 10.1016/j.semcancer.2024.05.002] [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: 01/17/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 05/31/2024]
Abstract
Cancer is daunting pathology with remarkable breadth and scope, spanning genetics, epigenetics, proteomics, metalobomics and cell biology. Cellular senescence represents a stress-induced and essentially irreversible cell fate associated with aging and various age-related diseases, including malignancies. Senescent cells are characterized of morphologic alterations and metabolic reprogramming, and develop a highly active secretome termed as the senescence-associated secretory phenotype (SASP). Since the first discovery, senescence has been understood as an important barrier to tumor progression, as its induction in pre-neoplastic cells limits carcinogenesis. Paradoxically, senescent cells arising in the tumor microenvironment (TME) contribute to tumor progression, including augmented therapeutic resistance. In this article, we define typical forms of senescent cells commonly observed within the TME and how senescent cells functionally remodel their surrounding niche, affect immune responses and promote cancer evolution. Furthermore, we highlight the recently emerging pipelines of senotherapies particularly senolytics, which can selectively deplete senescent cells from affected organs in vivo and impede tumor progression by restoring therapeutic responses and securing anticancer efficacies. Together, co-targeting cancer cells and their normal but senescent counterparts in the TME holds the potential to achieve increased therapeutic benefits and restrained disease relapse in future clinical oncology.
Collapse
Affiliation(s)
- Birong Jiang
- School of Pharmacy, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Wei Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuguang Zhang
- Mengniu Institute of Nutrition Science, Global R&D Innovation Center, Shanghai 200124, China
| | - Yu Sun
- School of Pharmacy, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
8
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [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: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
9
|
Li J, Wu J, Xie Y, Yu X. Bone marrow adipocytes and lung cancer bone metastasis: unraveling the role of adipokines in the tumor microenvironment. Front Oncol 2024; 14:1360471. [PMID: 38571500 PMCID: PMC10987778 DOI: 10.3389/fonc.2024.1360471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024] Open
Abstract
Bone is a common site of metastasis for lung cancer. The "seed and soil" hypothesis suggests that the bone marrow microenvironment ("soil") may provide a conducive survival environment for metastasizing tumor cells ("seeds"). The bone marrow microenvironment, comprising a complex array of cells, includes bone marrow adipocytes (BMAs), which constitute about 70% of the adult bone marrow volume and may play a significant role in tumor bone metastasis. BMAs can directly provide energy for tumor cells, promoting their proliferation and migration. Furthermore, BMAs participate in the tumor microenvironment's osteogenesis regulation, osteoclast(OC) regulation, and immune response through the secretion of adipokines, cytokines, and inflammatory factors. However, the precise mechanisms of BMAs in lung cancer bone metastasis remain largely unclear. This review primarily explores the role of BMAs and their secreted adipokines (leptin, adiponectin, Nesfatin-1, Resistin, chemerin, visfatin) in lung cancer bone metastasis, aiming to provide new insights into the mechanisms and clinical treatment of lung cancer bone metastasis.
Collapse
Affiliation(s)
- Jian Li
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology and Metabolism, Shandong Second Provincial General Hospital, Jinan, China
| | - Jialu Wu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yanni Xie
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism/Department of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Chen KY, Liu SY, Tang JJ, Liu MK, Chen XY, Liu ZP, Ferrandon D, Lai KF, Li Z. NLRP3 knockout in mice provided protection against Serratia marcescens-induced acute pneumonia by decreasing PD-L1 and PD-1 expression in macrophages. Int Immunopharmacol 2024; 129:111559. [PMID: 38330794 DOI: 10.1016/j.intimp.2024.111559] [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: 11/12/2023] [Revised: 01/05/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024]
Abstract
Antibiotic-resistant Serratia marcescens (Sm) is known to cause bloodstream infections, pneumonia, etc. The nod-like receptor family, pyrin domain-containing 3 (NLRP3), has been implicated in various lung infections. Yet, its role in Sm-induced pneumonia was not well understood. In our study, we discovered that deletion of Nlrp3 in mice significantly improved Sm-induced survival rates, reduced bacterial loads in the lungs, bronchoalveolar lavage fluid (BALF), and bloodstream, and mitigated the severity of acute lung injury (ALI) compared to wild-type (WT) mice. Mechanistically, we observed that 24 h post-Sm infection, NLRP3 inflammasome activation occurred, leading to gasdermin D NH2-terminal (GSDMD-NT)-induced pyroptosis in macrophages and IL-1β secretion. The NLRP3 or NLRP3 inflammasome influenced the expression PD-L1 and PD-1, as well as the count of PD-L1 or PD-1-expressing macrophages, alveolar macrophages, interstitial macrophages, PD-L1-expressing neutrophils, and the count of macrophage receptors with collagenous structure (MARCO)-expressing macrophages, particularly MARCO+ alveolar macrophages. The frequency of MARCO+ alveolar macrophages, PD-1 expression, particularly PD-1+ interstitial macrophages were negatively or positively correlated with the Sm load, respectively. Additionally, IL-1β levels in BALF correlated with three features of acute lung injury: histologic score, protein concentration and neutrophil count in BALF. Consequently, our findings suggest that Nlrp3 deletion offers protection agaisnt acute Sm pneumonia in mice by inhibiting inflammasome activation and reducing Sm infection-induced PD-L1/PD-1 or MARCO expression, particularly in macrophages. This highlights potential therapeutic targets for Sm and other gram-negative bacteria-induced acute pneumonia.
Collapse
Affiliation(s)
- Kan-Yao Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory, Guangdong Provincial People's Hospital Zhuhai Hospital, Zhuhai, China
| | - Shu-Yan Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Juan-Juan Tang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Meng-Ke Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xu-Yang Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhi-Peng Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Dominique Ferrandon
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China; Université de Strasbourg, RIDI UPR9022 du CNRS, F-67000 Strasbourg, France
| | - Ke-Fang Lai
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China.
| | - Zi Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, China; The Second Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Yang Y, Li J, Li D, Zhou W, Yan F, Wang W. Humanized mouse models: A valuable platform for preclinical evaluation of human cancer. Biotechnol Bioeng 2024; 121:835-852. [PMID: 38151887 DOI: 10.1002/bit.28618] [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: 07/19/2023] [Accepted: 11/26/2023] [Indexed: 12/29/2023]
Abstract
Animal models are routinely employed to assess the treatments for human cancer. However, due to significant differences in genetic backgrounds, traditional animal models are unable to meet bioresearch needs. To overcome this restriction, researchers have generated and optimized immunodeficient mice, and then engrafted human genes, cells, tissues, or organs in mice so that the responses in the model mice could provide a more reliable reference for treatments. As a bridge connecting clinical application and basic research, humanized mice are increasingly used in the preclinical evaluation of cancer treatments, particularly after gene interleukin 2 receptor gamma mutant mice were generated. Human cancer models established in humanized mice support exploration of the mechanism of cancer occurrence and provide an efficient platform for drug screening. However, it is undeniable that the further application of humanized mice still faces multiple challenges. This review summarizes the construction approaches for humanized mice and their existing limitations. We also report the latest applications of humanized mice in preclinical evaluation for the treatment of cancer and point out directions for future optimization of these models.
Collapse
Affiliation(s)
- Yuening Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqian Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Weilin Zhou
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Feiyang Yan
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Sahu P, Mitra A, Ganguly A. Targeting KRAS and SHP2 signaling pathways for immunomodulation and improving treatment outcomes in solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:167-222. [PMID: 38782499 DOI: 10.1016/bs.ircmb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Historically, KRAS has been considered 'undruggable' inspite of being one of the most frequently altered oncogenic proteins in solid tumors, primarily due to the paucity of pharmacologically 'druggable' pockets within the mutant isoforms. However, pioneering developments in drug design capable of targeting the mutant KRAS isoforms especially KRASG12C-mutant cancers, have opened the doors for emergence of combination therapies comprising of a plethora of inhibitors targeting different signaling pathways. SHP2 signaling pathway, primarily known for activation of intracellular signaling pathways such as KRAS has come up as a potential target for such combination therapies as it emerged to be the signaling protein connecting KRAS and the immune signaling pathways and providing the link for understanding the overlapping regions of RAS/ERK/MAPK signaling cascade. Thus, SHP2 inhibitors having potent tumoricidal activity as well as role in immunomodulation have generated keen interest in researchers to explore its potential as combination therapy in KRAS mutant solid tumors. However, the excitement with these combination therapies need to overcome challenges thrown up by drug resistance and enhanced toxicity. In this review, we will discuss KRAS and SHP2 signaling pathways and their roles in immunomodulation and regulation of tumor microenvironment and also analyze the positive effects and drawbacks of the different combination therapies targeted at these signaling pathways along with their present and future potential to treat solid tumors.
Collapse
Affiliation(s)
- Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar, Jharkhand, India.
| |
Collapse
|
13
|
Zhang Z, Li M, Tai Y, Xing Y, Zuo H, Jin X, Ma J. ZNF70 regulates IL-1β secretion of macrophages to promote the proliferation of HCT116 cells via activation of NLRP3 inflammasome and STAT3 pathway in colitis-associated colorectal cancer. Cell Signal 2024; 114:110979. [PMID: 38000525 DOI: 10.1016/j.cellsig.2023.110979] [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: 06/20/2023] [Revised: 10/02/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023]
Abstract
Chronic inflammation is a key driver for colitis-associated colorectal cancer (CAC). It has been reported that inflammatory cytokines, such as IL-1β, could promote CAC. Zinc finger protein 70 (ZNF70) is involved in multiple biological processes. Here, we identified a previously unknown role for ZNF70 regulates macrophages IL-1β secretion to promote HCT116 proliferation in CAC, and investigated its underlying mechanism. We showed ZNF70 is much higher expressed in CAC tumor tissues compared with adjacent normal tissues in clinical CAC samples. Further experiments showed ZNF70 promoted macrophages IL-1β secretion and HCT116 proliferation. In LPS/ATP-stimulated THP-1 cells, we found ZNF70 activated NLRP3 inflammasome, resulting in robust IL-1β secretion. Interestingly, we discovered the ZnF domain of ZNF70 could interact with NLRP3 and decrease the K48-linked ubiquitination of NLRP3. Moreover, ZNF70 could activate STAT3, thereby promoting IL-1β synthesis. Noteworthy, ZNF70 enhanced proliferation by upregulating STAT3 activation in HCT116 cells cultured in the conditioned medium of THP-1 macrophages treated with LPS/ATP. Finally, the vivo observations were confirmed using AAV-mediated ZNF70 knockdown, which improved colitis-associated colorectal cancer in the AOM/DSS model. The correlation between ZNF70 expression and overall survival/IL-1β expression in colorectal cancer was verified by TCGA database. Taken together, ZNF70 regulates macrophages IL-1β secretion to promote the HCT116 cells proliferation via activation of NLRP3 inflammasome and STAT3 pathway, suggesting that ZNF70 may be a promising preventive target for treating in CAC.
Collapse
Affiliation(s)
- Zhihong Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China; Department of Pharmacology, College of Pharmacy, Beihua University, East Binjiang Road, 3999, Jilin, China
| | - Mingyue Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Yi Tai
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Yue Xing
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hongxiang Zuo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Xuejun Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| | - Juan Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China.
| |
Collapse
|
14
|
Tan DSW, Felip E, de Castro G, Solomon BJ, Greystoke A, Cho BC, Cobo M, Kim TM, Ganguly S, Carcereny E, Paz-Ares L, Bennouna J, Garassino MC, Schenker M, Kim SW, Brase JC, Bury-Maynard D, Passos VQ, Deudon S, Dharan B, Song Y, Caparica R, Johnson BE. Canakinumab Versus Placebo in Combination With First-Line Pembrolizumab Plus Chemotherapy for Advanced Non-Small-Cell Lung Cancer: Results From the CANOPY-1 Trial. J Clin Oncol 2024; 42:192-204. [PMID: 38039427 DOI: 10.1200/jco.23.00980] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/14/2023] [Accepted: 10/03/2023] [Indexed: 12/03/2023] Open
Abstract
PURPOSE The addition of checkpoint inhibitors to first-line treatment has prolonged survival of patients with non-small-cell lung cancer (NSCLC), but prognosis remains poor, with new treatment options needed. Canakinumab, a human, monoclonal anti-interleukin (IL)-1β antibody, has potential to enhance the activity of PD-L1 inhibitors and chemotherapy (CT) by inhibiting protumor inflammation. METHODS CANOPY-1 was a phase III, randomized, double-blind study comparing canakinumab (200 mg subcutaneously once every 3 weeks) versus placebo, both combined with pembrolizumab (200 mg intravenously once every 3 weeks) and platinum-based doublet CT, as first-line treatment for advanced/metastatic NSCLC without EGFR or ALK mutations. The primary end points were progression-free survival (PFS) and overall survival (OS). The secondary endpoints included overall response rate, safety, and patient-reported outcomes. RESULTS Overall, 643 patients were randomly assigned to canakinumab (n = 320) or placebo (n = 323). With a median study follow-up of 6.5 months, the median PFS was 6.8 months with canakinumab versus 6.8 months with placebo (hazard ratio [HR], 0.85; 95% CI, 0.67 to 1.09; P = .102). With a median study follow-up of 21.2 months, the median OS was 20.8 months with canakinumab versus 20.2 months with placebo (HR, 0.87; 95% CI, 0.70 to 1.10; P = .123). No unexpected safety signals were observed for canakinumab combination. Infection rates were comparable between treatment and control arms. A higher frequency of neutropenia and ALT increase (grade ≤2) were reported in the treatment arm. Higher baseline C-reactive protein and IL-6 levels were associated with shorter PFS and OS. Patients treated with canakinumab had clinically meaningful delays in deterioration of lung cancer symptoms, including chest pain and coughing per LC13 and dyspnea per LC13 and C30. CONCLUSION The addition of canakinumab to first-line pembrolizumab and CT did not prolong PFS or OS in patients with NSCLC.
Collapse
Affiliation(s)
- Daniel S W Tan
- National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | - Alastair Greystoke
- Northern Centre for Cancer Care, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Manuel Cobo
- Medical Oncology Intercenter Unit, Regional University Hospital and Virgen de la Victoria University Hospital, IBIMA, Málaga, Spain
| | - Tae Min Kim
- Seoul National University Hospital, Seoul, Republic of Korea
| | | | - Enric Carcereny
- Catalan Institute of Oncology, Badalona Applied Research Group in Oncology (B-ARGO), Barcelona, Spain
| | | | - Jaafar Bennouna
- Department of Medical Oncology, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Marina Chiara Garassino
- Department of Medicine, Section Hematology Oncology, Thoracic Oncology program, The University of Chicago, Chicago, IL
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michael Schenker
- Sf Nectarie Oncology Center Craiova and the University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Sang-We Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Yuanbo Song
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | | | | |
Collapse
|
15
|
Garon EB, Lu S, Goto Y, De Marchi P, Paz-Ares L, Spigel DR, Thomas M, Yang JCH, Ardizzoni A, Barlesi F, Orlov S, Yoshioka H, Mountzios G, Khanna S, Bossen C, Carbini M, Turri S, Myers A, Cho BC. Canakinumab as Adjuvant Therapy in Patients With Completely Resected Non-Small-Cell Lung Cancer: Results From the CANOPY-A Double-Blind, Randomized Clinical Trial. J Clin Oncol 2024; 42:180-191. [PMID: 37788412 DOI: 10.1200/jco.23.00910] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/28/2023] [Accepted: 08/09/2023] [Indexed: 10/05/2023] Open
Abstract
PURPOSE Effective treatments for resectable non-small-cell lung cancer (NSCLC) are limited and relapse rates are high. The interleukin (IL)-1β pathway has been linked with tumor development and progression, including in the Canakinumab Anti-Inflammatory Thrombosis Outcomes cardiovascular study in which IL-1β pathway inhibition with canakinumab reduced lung cancer incidence and mortality in an exploratory analysis. METHODS CANOPY-A (ClinicalTrials.gov identifier: NCT03447769) is a phase III, randomized, double-blind, multicenter study of canakinumab versus placebo for adult patients with stage II-IIIA or IIIB (T >5 cm, N2-positives II-IIIB; American Joint Committee on Cancer/Union for International Cancer Control version 8), completely resected NSCLC who had received adjuvant cisplatin-based chemotherapy. The primary end point was disease-free survival (DFS) and the key secondary end point was overall survival (OS). RESULTS In total, 1,382 patients were randomized to 200 mg canakinumab (n = 693) or placebo (n = 689) once every 3 weeks for 18 cycles. Grade ≥3 adverse events (AEs) were reported in 20.8% and 19.6% of patients receiving canakinumab and placebo, respectively; AEs led to discontinuation in 4.3% and 4.1% of patients in these groups, respectively. This study did not meet its primary end point. Median DFS was 35.0 months (canakinumab arm) and 29.7 months (placebo arm; hazard ratio, 0.94; 95% CI, 0.78 to 1.14; one-sided P = .258). DFS subgroup analyses did not show any meaningful differences between arms. As expected, because of canakinumab-driven IL-1β pathway inhibition, C-reactive protein and IL-6 levels decreased in the canakinumab arm versus placebo arm, but had no correlation with differential clinical outcomes. OS was not formally tested as DFS was not statistically significant. CONCLUSION CANOPY-A did not show a DFS benefit of adding canakinumab after surgery and adjuvant cisplatin-based chemotherapy in patients with resected, stage II-III NSCLC. No new safety signals were identified with canakinumab.
Collapse
Affiliation(s)
- Edward B Garon
- David Geffen School of Medicine at UCLA/TRIO-US/TRIO-Global Network, Los Angeles, CA
| | - Shun Lu
- Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | - Luis Paz-Ares
- University Hospital 12 de Octubre, CNIO-H120 Lung Cancer Unit, Completense University and Ciberonc, Madrid, Spain
| | | | - Michael Thomas
- Thoraxklinik and National Center for Tumor Diseases at Heidelberg University Hospital, Translational Lung Research Center Heidelberg (TLRH-C), German Center for Lung Research (DZL), Heidelberg, Germany
| | - James Chih-Hsin Yang
- National Taiwan University Cancer Center and National Taiwan University Hospital, Taipei, Taiwan
| | - Andrea Ardizzoni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Fabrice Barlesi
- Medical Oncology Department, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Bicêtre, France
| | - Sergey Orlov
- Saint Petersburg Electrotechnical University, Saint Petersburg, Russia
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University, Hirakata, Japan
| | - Giannis Mountzios
- Fourth Oncology Department and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | | | | | | | | | - Andrea Myers
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
16
|
Jiao Z, Zhang J. Interplay between inflammasomes and PD-1/PD-L1 and their implications in cancer immunotherapy. Carcinogenesis 2023; 44:795-808. [PMID: 37796835 DOI: 10.1093/carcin/bgad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/22/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023] Open
Abstract
The inflammasomes play crucial roles in inflammation and cancer development, while the PD-1/PD-L1 pathway is critical for immune suppression in the tumor microenvironment (TME). Recent research indicates a reciprocal regulatory relationship between inflammasomes and PD-1/PD-L1 signaling in cancer development and PD-1 blockade treatment. By activating in diverse cells in tumor tissues, inflammasome upregulates PD-L1 level in the TME. Moreover, the regulation of PD-1/PD-L1 activity by inflammasome activation involves natural killer cells, tumor-associated macrophages and myeloid-derived suppressor cells. Conversely, PD-1 blockade can activate the inflammasome, potentially influencing treatment outcomes. The interplay between inflammasomes and PD-1/PD-L1 has profound and intricate effects on cancer development and treatment. In this review, we discuss the crosstalk between inflammasomes and PD-1/PD-L1 in cancers, exploring their implications for tumorigenesis, metastasis and immune checkpoint inhibitor (ICI) resistance. The combined therapeutic strategies targeting both inflammasomes and checkpoint molecules hold promising potential as treatments for cancer.
Collapse
Affiliation(s)
- Zhongyu Jiao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology (Peking University), Peking University Health Science Center, Beijing 100191, P.R. China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology (Peking University), Peking University Health Science Center, Beijing 100191, P.R. China
| |
Collapse
|
17
|
He Y, Li R, She W, Ai Y, Li K, Kumeria T, Jiang Z, Shao Q, Zou C, Albashari AA, Duan X, Ye Q. Inhibitory effects of the nanoscale lysate derived from xenogenic dental pulp stem cells in lung cancer models. J Nanobiotechnology 2023; 21:488. [PMID: 38105218 PMCID: PMC10726628 DOI: 10.1186/s12951-023-02218-1] [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] [Accepted: 11/17/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Lung cancer is a highly prevalent malignancy and has the highest mortality rate among all tumors due to lymph node metastasis. Bone marrow and umbilical cord-derived mesenchymal stem cells (MSCs) have demonstrated tumor-suppressive effects on lung cancer. This study investigated the effects of DPSC lysate on proliferation, apoptosis, migration and invasion of cancer cells were studied in vivo and in vitro. METHODS The proliferation, apoptosis, and migration/metastasis were evaluated by cell counting kit-8 assay, Annexin-V and propidium iodide staining, and the transwell assay, respectively. The expression levels of apoptosis-, cell cycle-, migration-, and adhesion-related mRNA and proteins were measured by qRT-PCR and western blot. The level and mRNA expression of tumor markers carcino embryonic antigen (CEA), neuron-specific enolase (NSE), and squamous cell carcinoma (SCC) were measured by Enzyme-linked immunosorbent assay (ELISA) and qRT-PCR. Finally, a tumor-bearing mouse model was constructed to observe the tumor-suppressive effect of DPSC lysate after intraperitoneal injection. RESULTS DPSC lysate decreased the viability of A549 cells and induced apoptosis in lung cancer cells. Western blot confirmed that levels of Caspase-3, Bax, and Bad were increased, and Bcl-2 protein levels were decreased in A549 cells treated with DPSC lysate. In addition, DPSC lysate inhibited the migration and invasion of A549 cells; downregulated key genes of the cell cycle, migration, and adhesion; and significantly suppressed tumor markers. Xenograft results showed that DPSC lysate inhibited tumor growth and reduced tumor weight. CONCLUSIONS DPSC lysate inhibited proliferation, invasion, and metastasis; promoted apoptosis in lung cancer cells; and suppressed tumor growth- potentially providing a cell-based alternative therapy for lung cancer treatment.
Collapse
Affiliation(s)
- Yan He
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China
- Institute for Regenerative and Translational Research, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, 460030, Hubei, China
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 324025, Zhejiang, China
| | - Ruohan Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China
| | - Wenting She
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China
| | - Yilong Ai
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528000, Guangdong, China
| | - Kesheng Li
- Institute for Regenerative and Translational Research, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, 460030, Hubei, China
| | - Tushar Kumeria
- School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW, 2052, Australia
| | - Ziran Jiang
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528000, Guangdong, China
| | - Qing Shao
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528000, Guangdong, China
| | - Chen Zou
- Foshan Stomatological Hospital, School of Medicine, Foshan University, Foshan, 528000, Guangdong, China.
| | | | - Xingxiang Duan
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China.
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China.
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 324025, Zhejiang, China.
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 460030, Hubei, China.
- Department of Oral and Maxillofacial Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
18
|
Li M, Jiang P, Yang Y, Xiong L, Wei S, Wang J, Li C. The role of pyroptosis and gasdermin family in tumor progression and immune microenvironment. Exp Hematol Oncol 2023; 12:103. [PMID: 38066523 PMCID: PMC10704735 DOI: 10.1186/s40164-023-00464-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/29/2023] [Indexed: 06/29/2024] Open
Abstract
Pyroptosis, an inflammatory programmed cell death, distinguishes itself from apoptosis and necroptosis and has drawn increasing attention. Recent studies have revealed a correlation between the expression levels of many pyroptosis-related genes and both tumorigenesis and progression. Despite advancements in cancer treatments such as surgery, radiotherapy, chemotherapy, and immunotherapy, the persistent hallmark of cancer enables malignant cells to elude cell death and develop resistance to therapy. Recent findings indicate that pyroptosis can overcome apoptosis resistance amplify treatment-induced tumor cell death. Moreover, pyroptosis triggers antitumor immunity by releasing pro-inflammatory cytokines, augmenting macrophage phagocytosis, and activating cytotoxic T cells and natural killer cells. Additionally, it transforms "cold" tumors into "hot" tumors, thereby enhancing the antitumor effects of various treatments. Consequently, pyroptosis is intricately linked to tumor development and holds promise as an effective strategy for boosting therapeutic efficacy. As the principal executive protein of pyroptosis, the gasdermin family plays a pivotal role in influencing pyroptosis-associated outcomes in tumors and can serve as a regulatory target. This review provides a comprehensive summary of the relationship between pyroptosis and gasdermin family members, discusses their roles in tumor progression and the tumor immune microenvironment, and analyses the underlying therapeutic strategies for tumor treatment based on pyroptotic cell death.
Collapse
Affiliation(s)
- Mengyuan Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Yuhan Yang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Liting Xiong
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Shuhua Wei
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
19
|
Olivera I, Luri-Rey C, Teijeira A, Eguren-Santamaria I, Gomis G, Palencia B, Berraondo P, Melero I. Facts and Hopes on Neutralization of Protumor Inflammatory Mediators in Cancer Immunotherapy. Clin Cancer Res 2023; 29:4711-4727. [PMID: 37522874 DOI: 10.1158/1078-0432.ccr-22-3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
In cancer pathogenesis, soluble mediators are responsible for a type of inflammation that favors the progression of tumors. The mechanisms chiefly involve changes in the cellular composition of the tumor tissue stroma and in the functional modulation of myeloid and lymphoid leukocytes. Active immunosuppression, proangiogenesis, changes in leukocyte traffic, extracellular matrix remodeling, and alterations in tumor-antigen presentation are the main mechanisms linked to the inflammation that fosters tumor growth and metastasis. Soluble inflammatory mediators and their receptors are amenable to various types of inhibitors that can be combined with other immunotherapy approaches. The main proinflammatory targets which can be interfered with at present and which are under preclinical and clinical development are IL1β, IL6, the CXCR1/2 chemokine axis, TNFα, VEGF, leukemia inhibitory factor, CCL2, IL35, and prostaglandins. In many instances, the corresponding neutralizing agents are already clinically available and can be repurposed as a result of their use in other areas of medicine such as autoimmune diseases and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Irene Olivera
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Iñaki Eguren-Santamaria
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Gabriel Gomis
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Belen Palencia
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Yi M, Li T, Niu M, Mei Q, Zhao B, Chu Q, Dai Z, Wu K. Exploiting innate immunity for cancer immunotherapy. Mol Cancer 2023; 22:187. [PMID: 38008741 PMCID: PMC10680233 DOI: 10.1186/s12943-023-01885-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 11/28/2023] Open
Abstract
Immunotherapies have revolutionized the treatment paradigms of various types of cancers. However, most of these immunomodulatory strategies focus on harnessing adaptive immunity, mainly by inhibiting immunosuppressive signaling with immune checkpoint blockade, or enhancing immunostimulatory signaling with bispecific T cell engager and chimeric antigen receptor (CAR)-T cell. Although these agents have already achieved great success, only a tiny percentage of patients could benefit from immunotherapies. Actually, immunotherapy efficacy is determined by multiple components in the tumor microenvironment beyond adaptive immunity. Cells from the innate arm of the immune system, such as macrophages, dendritic cells, myeloid-derived suppressor cells, neutrophils, natural killer cells, and unconventional T cells, also participate in cancer immune evasion and surveillance. Considering that the innate arm is the cornerstone of the antitumor immune response, utilizing innate immunity provides potential therapeutic options for cancer control. Up to now, strategies exploiting innate immunity, such as agonists of stimulator of interferon genes, CAR-macrophage or -natural killer cell therapies, metabolic regulators, and novel immune checkpoint blockade, have exhibited potent antitumor activities in preclinical and clinical studies. Here, we summarize the latest insights into the potential roles of innate cells in antitumor immunity and discuss the advances in innate arm-targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ming Yi
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China
| | - Bin Zhao
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People's Republic of China.
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
21
|
Liu NN, Yi CX, Wei LQ, Zhou JA, Jiang T, Hu CC, Wang L, Wang YY, Zou Y, Zhao YK, Zhang LL, Nie YT, Zhu YJ, Yi XY, Zeng LB, Li JQ, Huang XT, Ji HB, Kozlakidis Z, Zhong L, Heeschen C, Zheng XQ, Chen C, Zhang P, Wang H. The intratumor mycobiome promotes lung cancer progression via myeloid-derived suppressor cells. Cancer Cell 2023; 41:1927-1944.e9. [PMID: 37738973 DOI: 10.1016/j.ccell.2023.08.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/08/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023]
Abstract
Although polymorphic microbiomes have emerged as hallmarks of cancer, far less is known about the role of the intratumor mycobiome as living microorganisms in cancer progression. Here, using fungi-enriched DNA extraction and deep shotgun metagenomic sequencing, we have identified enriched tumor-resident Aspergillus sydowii in patients with lung adenocarcinoma (LUAD). By three different syngeneic lung cancer mice models, we find that A. sydowii promotes lung tumor progression via IL-1β-mediated expansion and activation of MDSCs, resulting in suppressed activity of cytotoxic T lymphocyte cells and accumulation of PD-1+ CD8+ T cells. This is mediated by IL-1β secretion via β-glucan/Dectin-1/CARD9 pathway. Analysis of human samples confirms that enriched A. sydowii is associated with immunosuppression and poor patient outcome. Our findings suggest that intratumor mycobiome, albeit at low biomass, promotes lung cancer progression and could be targeted at the strain level to improve patients with LUAD outcome.
Collapse
Affiliation(s)
- Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Cheng-Xiang Yi
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lu-Qi Wei
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jin-An Zhou
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tong Jiang
- Shanghai Institute of Immunity and Infection, Chinese Academy of Science, (Past Name: Institut Pasteur of Shanghai, Chinese Academy of Sciences), Shanghai 200031, China; Laboratory Services and Biobanking, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Cong-Cong Hu
- Department of Mathematics, Shanghai Normal University, Shanghai 200234, China
| | - Lu Wang
- Department of Mathematics, Shanghai Normal University, Shanghai 200234, China
| | - Yuan-Yuan Wang
- Shanghai Institute of Immunity and Infection, Chinese Academy of Science, (Past Name: Institut Pasteur of Shanghai, Chinese Academy of Sciences), Shanghai 200031, China
| | - Yun Zou
- Shanghai Institute of Immunity and Infection, Chinese Academy of Science, (Past Name: Institut Pasteur of Shanghai, Chinese Academy of Sciences), Shanghai 200031, China
| | - Yi-Kai Zhao
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Le-Le Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ya-Ting Nie
- Department of Mathematics, Shanghai Normal University, Shanghai 200234, China
| | - Yi-Jing Zhu
- Department of Mathematics, Shanghai Normal University, Shanghai 200234, China
| | - Xin-Yao Yi
- Department of Mathematics, Shanghai Normal University, Shanghai 200234, China
| | - Ling-Bing Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330052, China
| | - Jing-Quan Li
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Tian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang 330052, China
| | - Hong-Bin Ji
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zisis Kozlakidis
- Laboratory Services and Biobanking, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Lin Zhong
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Christopher Heeschen
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao-Qi Zheng
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Changbin Chen
- Shanghai Institute of Immunity and Infection, Chinese Academy of Science, (Past Name: Institut Pasteur of Shanghai, Chinese Academy of Sciences), Shanghai 200031, China; Nanjing Advanced Academy of Life and Health, Nanjing 211135, China.
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
22
|
Aboushousha R, van der Velden J, Hamilton N, Peng Z, MacPherson M, Erickson C, White S, Wouters EFM, Reynaert NL, Seward DJ, Li J, Janssen-Heininger YMW. Glutaredoxin attenuates glutathione levels via deglutathionylation of Otub1 and subsequent destabilization of system x C. SCIENCE ADVANCES 2023; 9:eadi5192. [PMID: 37703360 PMCID: PMC10499329 DOI: 10.1126/sciadv.adi5192] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/10/2023] [Indexed: 09/15/2023]
Abstract
Glutathione (GSH) is a critical component of the cellular redox system that combats oxidative stress. The glutamate-cystine antiporter, system xC-, is a key player in GSH synthesis that allows for the uptake of cystine, the rate-limiting building block of GSH. It is unclear whether GSH or GSH-dependent protein oxidation [protein S-glutathionylation (PSSG)] regulates the activity of system xC-. We demonstrate that an environment of enhanced PSSG promotes GSH increases via a system xC--dependent mechanism. Absence of the deglutathionylase, glutaredoxin (GLRX), augmented SLC7A11 protein and led to significant increases of GSH content. S-glutathionylation of C23 or C204 of the deubiquitinase OTUB1 promoted interaction with the E2-conjugating enzyme UBCH5A, leading to diminished ubiquitination and proteasomal degradation of SLC7A11 and augmentation of GSH, effects that were reversed by GLRX. These findings demonstrate an intricate link between GLRX and GSH via S-glutathionylation of OTUB1 and system xC- and illuminate a previously unknown feed-forward regulatory mechanism whereby enhanced GSH protein oxidation augments cellular GSH.
Collapse
Affiliation(s)
- Reem Aboushousha
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jos van der Velden
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Nicholas Hamilton
- Department of Chemistry, University of Vermont, Burlington, VT 05405, USA
| | - Zhihua Peng
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Maximilian MacPherson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Sheryl White
- Department of Neurological Sciences, University of Vermont, Burlington, VT 05405, USA
| | - Emiel F. M. Wouters
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
- Ludwig Boltzmann Institute for Lung Research, Vienna, Austria
| | - Niki L. Reynaert
- Department of Respiratory Medicine, NUTRIM School of nutrition and translational research in metabolism, Maastricht University Medical Center, Maastricht, Netherlands
| | - David J. Seward
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jianing Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Vermont, Burlington, VT 05405, USA
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
23
|
Liu Y, Xie B, Chen Q. RAS signaling and immune cells: a sinister crosstalk in the tumor microenvironment. J Transl Med 2023; 21:595. [PMID: 37670322 PMCID: PMC10481548 DOI: 10.1186/s12967-023-04486-9] [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: 07/27/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023] Open
Abstract
The rat sarcoma virus (RAS) gene is the most commonly mutated oncogene in cancer, with about 19% of cancer patients carrying RAS mutations. Studies on the interaction between RAS mutation and tumor immune microenvironment (TIM) have been flourishing in recent years. More and more evidence has proved that RAS signals regulate immune cells' recruitment, activation, and differentiation while assisting tumor cells to evade immune surveillance. This review concluded the direct and indirect treatment strategies for RAS mutations. In addition, we updated the underlying mechanisms by which RAS signaling modulated immune infiltration and immune escape. Finally, we discussed advances in RAS-targeted immunotherapies, including cancer vaccines and adoptive cell therapies, with a particular focus on combination strategies with personalized therapy and great potential to achieve lasting clinical benefits.
Collapse
Affiliation(s)
- Yongting Liu
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Bin Xie
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
24
|
Castillo DR, Jeon WJ, Park D, Pham B, Yang C, Joung B, Moon JH, Lee J, Chong EG, Park K, Reeves ME, Duerksen-Hughes P, Mirshahidi HR, Mirshahidi S. Comprehensive Review: Unveiling the Pro-Oncogenic Roles of IL-1ß and PD-1/PD-L1 in NSCLC Development and Targeting Their Pathways for Clinical Management. Int J Mol Sci 2023; 24:11547. [PMID: 37511306 PMCID: PMC10380530 DOI: 10.3390/ijms241411547] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
In the past decade, targeted therapies for solid tumors, including non-small cell lung cancer (NSCLC), have advanced significantly, offering tailored treatment options for patients. However, individuals without targetable mutations pose a clinical challenge, as they may not respond to standard treatments like immune-checkpoint inhibitors (ICIs) and novel targeted therapies. While the mechanism of action of ICIs seems promising, the lack of a robust response limits their widespread use. Although the expression levels of programmed death ligand 1 (PD-L1) on tumor cells are used to predict ICI response, identifying new biomarkers, particularly those associated with the tumor microenvironment (TME), is crucial to address this unmet need. Recently, inflammatory cytokines such as interleukin-1 beta (IL-1β) have emerged as a key area of focus and hold significant potential implications for future clinical practice. Combinatorial approaches of IL-1β inhibitors and ICIs may provide a potential therapeutic modality for NSCLC patients without targetable mutations. Recent advancements in our understanding of the intricate relationship between inflammation and oncogenesis, particularly involving the IL-1β/PD-1/PD-L1 pathway, have shed light on their application in lung cancer development and clinical outcomes of patients. Targeting these pathways in cancers like NSCLC holds immense potential to revolutionize cancer treatment, particularly for patients lacking targetable genetic mutations. However, despite these promising prospects, there remain certain aspects of this pathway that require further investigation, particularly regarding treatment resistance. Therefore, the objective of this review is to delve into the role of IL-1β in NSCLC, its participation in inflammatory pathways, and its intricate crosstalk with the PD-1/PD-L1 pathway. Additionally, we aim to explore the potential of IL-1β as a therapeutic target for NSCLC treatment.
Collapse
Affiliation(s)
- Dani Ran Castillo
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Won Jin Jeon
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Daniel Park
- Department of Internal Medicine, University of San Francisco-Fresno, Fresno, CA 93701, USA;
| | - Bryan Pham
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Chieh Yang
- Department of Internal Medicine, School of Medicine, University of California Riverside, Riverside, CA 92521, USA;
| | - Bowon Joung
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Jin Hyun Moon
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.J.J.); (B.P.); (B.J.); (J.H.M.)
| | - Jae Lee
- School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Esther G. Chong
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Kiwon Park
- Department of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Mark E. Reeves
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Penelope Duerksen-Hughes
- Division of Biochemistry, Department of Medicine & Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Hamid R. Mirshahidi
- Division of Hematology and Oncology, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA; (D.R.C.); (E.G.C.); (M.E.R.); (H.R.M.)
| | - Saied Mirshahidi
- Biospecimen Laboratory, Loma Linda University Cancer Center, Loma Linda, CA 92354, USA
- Division of Microbiology and Molecular Genetics, Department of Medicine & Basic Sciences, Loma Linda University, Loma Linda 92350, CA, USA
| |
Collapse
|
25
|
Pandey R, Nema R, Vishwakarma S, Singh AP, Mohan S, Patel P, Halder S, Halder A, Singh R, Agarwal R, Gupta V, Kumar A. Single Nucleotide Polymorphisms in Cytokine Genes are Associated with the Susceptibility to Oral Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2023; 24:2353-2360. [PMID: 37505766 PMCID: PMC10676475 DOI: 10.31557/apjcp.2023.24.7.2353] [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: 02/11/2023] [Accepted: 07/07/2023] [Indexed: 07/29/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of cancer among men in the Indian subcontinent. Cytokines regulate inflammation and angiogenesis in a variety of cancers. Genetic variability in the cytokine genes can potentially influence the predisposition to oral carcinogenesis. The aim of the current study was to investigate the associations of SNPs in cytokine genes with the susceptibility of oral squamous cell carcinoma. In the present study, we have analyzed the allelic frequency of 32 single nucleotide polymorphisms (SNPs) using MassArray-based iPLEX assay in 16 cytokine genes in 166 OSCC patients and 151 healthy subjects from central India. Out of 32 SNPs analyzed, five SNPs were significantly associated with the risk of OSCC. AA and GG genotypes of IL-1β +3953 were associated with an increased and decreased risk of OSCC, respectively. In several genetic models, GG genotype and G allele in IL-12A 3'UTR G>A were found to be associated with an increased risk of OSCC. Similarly, the GG genotype of IL-12B +1188 T>G was associated with increased susceptibility to OSCC. We conclude that SNPs in the genes coding for IL-1β, IL-12A and IL-12B are associated with increased genetic susceptibility to OSCC in the central Indian population.
Collapse
Affiliation(s)
- Ritu Pandey
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Rajeev Nema
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Supriya Vishwakarma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Ajay Pal Singh
- All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
- Present Address: Department of Internal Medicine, All India Institute of Medical Sciences (AIIMS) Rishikesh, Rishikesh 249203, India.
| | - Sruthy Mohan
- All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Priti Patel
- All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Subhojit Halder
- All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Anupam Halder
- All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Renu Singh
- Jawaharlal Nehru Cancer Hospital and Research Center, Idgah Hills, Bhopal, India.
| | - Rahul Agarwal
- Jawaharlal Nehru Cancer Hospital and Research Center, Idgah Hills, Bhopal, India.
| | - Vikas Gupta
- Department of ENT and Head and Neck Surgery, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, India.
| |
Collapse
|
26
|
Wu S, Sun Z, Guo Z, Li P, Mao Q, Tang Y, Chen H, Peng H, Wang S, Cao Y. The effectiveness of blood-activating and stasis-transforming traditional Chinese medicines (BAST) in lung cancer progression-a comprehensive review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116565. [PMID: 37172918 DOI: 10.1016/j.jep.2023.116565] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Blood-activating and stasis-transforming traditional Chinese medicines (BAST) are a class of herbs that have the effect of dilating blood vessels and dispersing stagnation. Modern pharmaceutical research has demonstrated that they are capable of improving hemodynamics and micro-flow, resist thrombosis and promote blood flow. BAST contain numerous active ingredients, which can theoretically regulate multiple targets at the same time and have a wide range of pharmacological effects in the treatment of diseases including human cancers. Clinically, BAST have minimal side effects and can be used in combination with Western medicine to improve patients' quality of life, lessen adverse effects and minimize the risk of recurrence and metastasis of cancers. AIM OF THE REVIEW We aimed to summarize the research progression of BAST on lung cancer in the past five years and present a prospect for the future. Particularly, this review further analyzes the effects and molecular mechanisms that BAST inhibit the invasion and metastasis of lung cancer. MATERIALS AND METHODS Relevant studies about BSAT were collected from PubMed and Web of science. RESULTS Lung cancer is one of the malignant tumors with the highest mortality rate. Most patients with lung cancer are diagnosed at an advanced stage and are highly susceptible to metastasis. Recent studies have shown that BAST, a class of traditional Chinese medicine (TCM) with the function of opening veins and dispersing blood stasis, significantly improve hemodynamics and microcirculation, prevent thrombosis and promote blood flow, and thereby inhibiting the invasion and metastasis of lung cancer. In the current review, we analyzed 51 active ingredients extracted from BAST. It was found that BAST and their active ingredients contribute to the prevention of invasion and metastasis of lung cancer through multiple mechanisms, such as regulation of EMT process, specific signaling pathway and metastasis-related genes, tumor blood vessel formation, immune microenvironment and inflammatory response of tumors. CONCLUSIONS BSAT and its active ingredients have showed promising anticancer activity and significantly inhibit the invasion and metastasis of lung cancer. A growing number of studies have realized their potential clinical significance in the therapy of lung cancer, which will provide substantial evidences for the development of new TCM for lung cancer therapy.
Collapse
Affiliation(s)
- Siqi Wu
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zhe Sun
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Zehuai Guo
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Peiqin Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Qianqian Mao
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yang Tang
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Hongyu Chen
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Huiting Peng
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Sisi Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Yang Cao
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
27
|
Xie Y, Wang M, Xia H, Sun H, Yuan Y, Jia J, Chen L. Development and validation of a CECT-based radiomics model for predicting IL1B expression and prognosis of head and neck squamous cell carcinoma. Front Oncol 2023; 13:1121485. [PMID: 36969073 PMCID: PMC10036854 DOI: 10.3389/fonc.2023.1121485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionIt is necessary to explore a noninvasive method to stratify head and neck squamous cell carcinoma (HNSCC)’s prognosis and to seek new indicators for individualized precision treatment. As a vital inflammatory cytokine, IL1B might drive a new tumor subtype that could be reflected in overall survival (OS) and predicted using the radiomics method.MethodsA total of 139 patients with RNA-Seq data from The Cancer Genome Atlas (TCGA) and matched CECT data from The Cancer Image Archive (TCIA) were included in the analysis. The prognostic value of IL1B expression in patients with HNSCC was analyzed using Kaplan-Meier analysis, Cox regression analysis and subgroup analysis. Furthermore, the molecular function of IL1B on HNSCC was explored using function enrichment and immunocytes infiltration analyses. Radiomic features were extracted with PyRadiomics and processed using max-relevance minredundancy, recursive feature elimination, and gradient boosting machine algorithm to construct aradiomics model for predicting IL1B expression. The area under the receiver operating characteristic curve (AUC), calibration curve, precision recall (PR) curve, and decision curve analysis (DCA) curve were used to examine the performance of the model.ResultsIncreased IL1B expression in patients with HNSCC indicated a poor prognosis (hazard ratio [HR] = 1.56, P = 0.003) and was harmful in patients who underwent radiotherapy (HR = 1.87, P = 0.007) or chemotherapy (HR = 2.514, P < 0.001). Shape_Sphericity, glszm_SmallAreaEmphasis, and firstorder_Kurtosis were included in the radiomics model (AUC: training cohort, 0.861; validation cohort, 0.703). The calibration curves, PR curves and DCA showed good diagnostic effect of the model. The rad-score was close related to IL1B (P = 4.490*10-9), and shared the same corelated trend to EMT-related genes with IL1B. A higher rad-score was associated with worse overall survival (P = 0.041).DiscussionThe CECT-based radiomics model provides preoperative IL1B expression predictionand offers non-invasive instructions for the prognosis and individualized treatment of patients withHNSCC.
Collapse
Affiliation(s)
- Yang Xie
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Min Wang
- Hubei-MOST and KLOBM, Department of Oral Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haibin Xia
- Hubei-MOST and KLOBM, Department of Oral Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huifang Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Yuan
- Department of Oral Radiology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Liangwen Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- *Correspondence: Liangwen Chen,
| |
Collapse
|
28
|
Karimi N, Moghaddam SJ. KRAS-Mutant Lung Cancer: Targeting Molecular and Immunologic Pathways, Therapeutic Advantages and Restrictions. Cells 2023; 12:749. [PMID: 36899885 PMCID: PMC10001046 DOI: 10.3390/cells12050749] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
RAS mutations are among the most common oncogenic mutations in human cancers. Among RAS mutations, KRAS has the highest frequency and is present in almost 30% of non-small-cell lung cancer (NSCLC) patients. Lung cancer is the number one cause of mortality among cancers as a consequence of outrageous aggressiveness and late diagnosis. High mortality rates have been the reason behind numerous investigations and clinical trials to discover proper therapeutic agents targeting KRAS. These approaches include the following: direct KRAS targeting; synthetic lethality partner inhibitors; targeting of KRAS membrane association and associated metabolic rewiring; autophagy inhibitors; downstream inhibitors; and immunotherapies and other immune-modalities such as modulating inflammatory signaling transcription factors (e.g., STAT3). The majority of these have unfortunately encountered limited therapeutic outcomes due to multiple restrictive mechanisms including the presence of co-mutations. In this review we plan to summarize the past and most recent therapies under investigation, along with their therapeutic success rate and potential restrictions. This will provide useful information to improve the design of novel agents for treatment of this deadly disease.
Collapse
Affiliation(s)
- Nastaran Karimi
- Faculty of Medicine, Marmara University, Istanbul 34899, Turkey
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
29
|
Wang D, Li W, Albasha N, Griffin L, Chang H, Amaya L, Ganguly S, Zeng L, Keum B, González-Navajas JM, Levin M, AkhavanAghdam Z, Snyder H, Schwartz D, Tao A, Boosherhri LM, Hoffman HM, Rose M, Estrada MV, Varki N, Herdman S, Corr M, Webster NJG, Raz E, Bertin S. Long-term exposure to house dust mites accelerates lung cancer development in mice. J Exp Clin Cancer Res 2023; 42:26. [PMID: 36670473 PMCID: PMC9863279 DOI: 10.1186/s13046-022-02587-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/26/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Individuals with certain chronic inflammatory lung diseases have a higher risk of developing lung cancer (LC). However, the underlying mechanisms remain largely unknown. Here, we hypothesized that chronic exposure to house dust mites (HDM), a common indoor aeroallergen associated with the development of asthma, accelerates LC development through the induction of chronic lung inflammation (CLI). METHODS: The effects of HDM and heat-inactivated HDM (HI-HDM) extracts were evaluated in two preclinical mouse models of LC (a chemically-induced model using the carcinogen urethane and a genetically-driven model with oncogenic KrasG12D activation in lung epithelial cells) and on murine macrophages in vitro. Pharmacological blockade or genetic deletion of the Nod-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome, caspase-1, interleukin-1β (IL-1β), and C-C motif chemokine ligand 2 (CCL2) or treatment with an inhaled corticosteroid (ICS) was used to uncover the pro-tumorigenic effect of HDM. RESULTS: Chronic intranasal (i.n) instillation of HDM accelerated LC development in the two mouse models. Mechanistically, HDM caused a particular subtype of CLI, in which the NLRP3/IL-1β signaling pathway is chronically activated in macrophages, and made the lung microenvironment conducive to tumor development. The tumor-promoting effect of HDM was significantly decreased by heat treatment of the HDM extract and was inhibited by NLRP3, IL-1β, and CCL2 neutralization, or ICS treatment. CONCLUSIONS Collectively, these data indicate that long-term exposure to HDM can accelerate lung tumorigenesis in susceptible hosts (e.g., mice and potentially humans exposed to lung carcinogens or genetically predisposed to develop LC).
Collapse
Affiliation(s)
- Dongjie Wang
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Li
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Center for Immunology, Inflammation and Immune-Mediated Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Natalie Albasha
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Lindsey Griffin
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Han Chang
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Lauren Amaya
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Sneha Ganguly
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Liping Zeng
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Center for Immunology, Inflammation and Immune-Mediated Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bora Keum
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - José M González-Navajas
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Hospital General Universitario de Alicante, Alicante, Spain
- Alicante Institute of Health and Biomedical Research (ISABIAL), Alicante, Spain
| | | | | | | | | | - Ailin Tao
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Center for Immunology, Inflammation and Immune-Mediated Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Laela M Boosherhri
- Division of Pediatric Allergy, Immunology, and Rheumatology, Rady Children's Hospital of San Diego, University of California San Diego, La Jolla, CA, USA
| | - Hal M Hoffman
- Division of Pediatric Allergy, Immunology, and Rheumatology, Rady Children's Hospital of San Diego, University of California San Diego, La Jolla, CA, USA
| | - Michael Rose
- Tissue Technology Shared Resource, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Monica Valeria Estrada
- Tissue Technology Shared Resource, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Nissi Varki
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA
| | - Scott Herdman
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA
| | - Nicholas J G Webster
- Division of Endocrinology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, USA
- Medical Research Service, Veteran Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Eyal Raz
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA.
| | - Samuel Bertin
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0663, USA.
| |
Collapse
|
30
|
Sun Y, Li Q, Kirkland JL. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. LIFE MEDICINE 2022; 1:103-119. [PMID: 36699942 PMCID: PMC9869767 DOI: 10.1093/lifemedi/lnac030] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Aging is a natural but relentless process of physiological decline, leading to physical frailty, reduced ability to respond to physical stresses (resilience) and, ultimately, organismal death. Cellular senescence, a self-defensive mechanism activated in response to intrinsic stimuli and/or exogenous stress, is one of the central hallmarks of aging. Senescent cells cease to proliferate, while remaining metabolically active and secreting numerous extracellular factors, a feature known as the senescence-associated secretory phenotype. Senescence is physiologically important for embryonic development, tissue repair, and wound healing, and prevents carcinogenesis. However, chronic accumulation of persisting senescent cells contributes to a host of pathologies including age-related morbidities. By paracrine and endocrine mechanisms, senescent cells can induce inflammation locally and systemically, thereby causing tissue dysfunction, and organ degeneration. Agents including those targeting damaging components of the senescence-associated secretory phenotype or inducing apoptosis of senescent cells exhibit remarkable benefits in both preclinical models and early clinical trials for geriatric conditions. Here we summarize features of senescent cells and outline strategies holding the potential to be developed as clinical interventions. In the long run, there is an increasing demand for safe, effective, and clinically translatable senotherapeutics to address healthcare needs in current settings of global aging.
Collapse
Affiliation(s)
- Yu Sun
- Correspondence: (Y.S.), (Q.L.), (J.L.K.)
| | | | | |
Collapse
|