1
|
Wei S, Han C, Mo S, Huang H, Luo X. Advancements in programmed cell death research in antitumor therapy: a comprehensive overview. Apoptosis 2024:10.1007/s10495-024-02038-0. [PMID: 39487314 DOI: 10.1007/s10495-024-02038-0] [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] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Cell death is a normal physiological process within cells that involves multiple pathways, such as normal DNA damage, cell cycle arrest, and programmed cell death (PCD). Cell death has been a hot spot of research in tumor-related fields, especially programmed cell death, which is a key form of cell death and is classified into different types according to the mechanism of occurrence, such as apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and disulfidptosis. Given the important role of PCD in maintaining tissue homeostasis and inhibiting tumorigenesis and development, more and more basic and clinical studies are devoted to revealing its potential application in anti-tumor strategies. The purpose of this review is to systematically review the regulatory mechanisms of PCD and to summarize the latest research progress of anti-tumor treatment strategies based on PCD.
Collapse
Affiliation(s)
- Shuxin Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hailian Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoling Luo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China.
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
2
|
Pazhouhesh Far N, Hajiheidari Varnousafaderani M, Faghihkhorasani F, Etemad S, Abdulwahid AHRR, Bakhtiarinia N, Mousaei A, Dortaj E, Karimi S, Ebrahimi N, Aref AR. Breaking the barriers: Overcoming cancer resistance by targeting the NLRP3 inflammasome. Br J Pharmacol 2024. [PMID: 39394867 DOI: 10.1111/bph.17352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 10/14/2024] Open
Abstract
Inflammation has a pivotal role in the initiation and progression of various cancers, contributing to crucial processes such as metastasis, angiogenesis, cell proliferation and invasion. Moreover, the release of cytokines mediated by inflammation within the tumour microenvironment (TME) has a crucial role in orchestrating these events. The activation of inflammatory caspases, facilitated by the recruitment of caspase-1, is initiated by the activation of pattern recognition receptors on the immune cell membrane. This activation results in the production of proinflammatory cytokines, including IL-1β and IL-18, and participates in diverse biological processes with significant implications. The NOD-Like Receptor Protein 3 (NLRP3) inflammasome holds a central role in innate immunity and regulates inflammation through releasing IL-1β and IL-18. Moreover, it interacts with various cellular compartments. Recently, the mechanisms underlying NLRP3 inflammasome activation have garnered considerable attention. Disruption in NLRP3 inflammasome activation has been associated with a spectrum of inflammatory diseases, encompassing diabetes, enteritis, neurodegenerative diseases, obesity and tumours. The NLRP3 impact on tumorigenesis varies across different cancer types, with contrasting roles observed. For example, colorectal cancer associated with colitis can be suppressed by NLRP3, whereas gastric and skin cancers may be promoted by its activity. This review provides comprehensive insights into the structure, biological characteristics and mechanisms of the NLRP3 inflammasome, with a specific focus on the relationship between NLRP3 and tumour-related immune responses, and TME. Furthermore, the review explores potential strategies for targeting cancers via NLRP3 inflammasome modulation. This encompasses innovative approaches, including NLRP3-based nanoparticles, gene-targeted therapy and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | | | | | - Sareh Etemad
- Department of Pathology, Faculty of Anatomical Pathology, Ghaem Hospital, University of Medicine, Mashhad, Iran
| | | | | | - Afsaneh Mousaei
- Department of Biology, College of Science, Qaemshahr Branch, Islamic Azad University, Qaem Shahr, Iran
| | - Elahe Dortaj
- Department of Ergonomics, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soroush Karimi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
Li LR, Chen L, Sun ZJ. Igniting hope: Harnessing NLRP3 inflammasome-GSDMD-mediated pyroptosis for cancer immunotherapy. Life Sci 2024; 354:122951. [PMID: 39127315 DOI: 10.1016/j.lfs.2024.122951] [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: 05/10/2024] [Revised: 07/19/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
In the contemporary landscape of oncology, immunotherapy, represented by immune checkpoint blockade (ICB) therapy, stands out as a beacon of innovation in cancer treatment. Despite its promise, the therapy's progression is hindered by suboptimal clinical response rates. Addressing this challenge, the modulation of the NLRP3 inflammasome-GSDMD-mediated pyroptosis pathway holds promise as a means to augment the efficacy of immunotherapy. In the pathway, the NLRP3 inflammasome serves as a pivotal molecular sensor that responds to inflammatory stimuli within the organism. Its activation leads to the release of cytokines interleukin 1β and interleukin 18 through the cleavage of GSDMD, thereby forming membrane pores and potentially resulting in pyroptosis. This cascade of processes exerts a profound impact on tumor development and progression, with its function and expression exhibiting variability across different tumor types and developmental stages. Consequently, understanding the specific roles of the NLRP3 inflammasome and GSDMD-mediated pyroptosis in diverse tumors is imperative for comprehending tumorigenesis and crafting precise therapeutic strategies. This review aims to elucidate the structure and activation mechanisms of the NLRP3 inflammasome, as well as the induction mechanisms of GSDMD-mediated pyroptosis. Additionally, we provide a comprehensive overview of the involvement of this pathway in various cancer types and its applications in tumor immunotherapy, nanotherapy, and other fields. Emphasis is placed on the feasibility of leveraging this approach to enhance ICB therapy within the field of immunotherapy. Furthermore, we discuss the potential applications of this pathway in other immunotherapy methods, such as chimeric antigen receptor T-cell (CAR-T) therapy and tumor vaccines.
Collapse
Affiliation(s)
- Ling-Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Lei Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Coll RC, Schroder K. Inflammasome components as new therapeutic targets in inflammatory disease. Nat Rev Immunol 2024:10.1038/s41577-024-01075-9. [PMID: 39251813 DOI: 10.1038/s41577-024-01075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/11/2024]
Abstract
Inflammation drives pathology in many human diseases for which there are no disease-modifying drugs. Inflammasomes are signalling platforms that can induce pathological inflammation and tissue damage, having potential as an exciting new class of drug targets. Small-molecule inhibitors of the NLRP3 inflammasome that are now in clinical trials have demonstrated proof of concept that inflammasomes are druggable, and so drug development programmes are now focusing on other key inflammasome molecules. In this Review, we describe the potential of inflammasome components as candidate drug targets and the novel inflammasome inhibitors that are being developed. We discuss how the signalling biology of inflammasomes offers mechanistic insights for therapeutic targeting. We also discuss the major scientific and technical challenges associated with drugging these molecules during preclinical development and clinical trials.
Collapse
Affiliation(s)
- Rebecca C Coll
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
5
|
Woo J, Zhai T, Yang F, Xu H, Healey ML, Yates DP, Beste MT, Steensma DP. Effect of Clonal Hematopoiesis Mutations and Canakinumab Treatment on Incidence of Solid Tumors in the CANTOS Randomized Clinical Trial. Cancer Prev Res (Phila) 2024; 17:429-436. [PMID: 38701435 DOI: 10.1158/1940-6207.capr-23-0342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/01/2024] [Accepted: 05/01/2024] [Indexed: 05/05/2024]
Abstract
Clonal hematopoiesis (CH) is more common in older persons and has been associated with an increased risk of hematological cancers and cardiovascular diseases. The most common CH mutations occur in the DNMT3A and TET2 genes and result in increased proinflammatory signaling. The Canakinumab Anti-inflammatory Thrombosis Outcome Study (NCT01327846) evaluated the neutralizing anti-IL1β antibody canakinumab in 10,061 randomized patients with a history of myocardial infarction and persistent inflammation; DNA samples were available from 3,923 patients for targeted genomic sequencing. We examined the incidence of non-hematological malignancy by treatment assignment and CH mutations and estimated the cumulative incidence of malignancy events during trial follow-up. Patients with TET2 mutations treated with canakinumab had the lowest incidence of non-hematological malignancy across cancer types. The cumulative incidence of at least one reported malignancy was lower for patients with TET2 mutations treated with canakinumab versus those treated with placebo. These findings support a potential role for canakinumab in cancer prevention and provide evidence of IL1β blockade cooperating with CH mutations to modify the disease course. Prevention Relevance: We reveal that administering canakinumab is associated with a decrease in non-hematological malignancies among patients with clonal hematopoiesis (CH) mutations. These findings underscore canakinumab's potential in preventing cancer and provide proof of IL1β blockade collaborating with CH mutations to enhance its clinical benefits. See related Spotlight, p. 399.
Collapse
Affiliation(s)
- Janghee Woo
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Tingting Zhai
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Fang Yang
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Huilei Xu
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Margaret L Healey
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Denise P Yates
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Michael T Beste
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - David P Steensma
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| |
Collapse
|
6
|
Oberstein PE, Dias Costa A, Kawaler EA, Cardot-Ruffino V, Rahma OE, Beri N, Singh H, Abrams TA, Biller LH, Cleary JM, Enzinger P, Huffman BM, McCleary NJ, Perez KJ, Rubinson DA, Schlechter BL, Surana R, Yurgelun MB, Wang SJ, Remland J, Brais LK, Bollenrucher N, Chang E, Ali LR, Lenehan PJ, Dolgalev I, Werba G, Lima C, Keheler CE, Sullivan KM, Dougan M, Hajdu C, Dajee M, Pelletier MR, Nazeer S, Squires M, Bar-Sagi D, Wolpin BM, Nowak JA, Simeone DM, Dougan SK. Blockade of IL1β and PD1 with Combination Chemotherapy Reduces Systemic Myeloid Suppression in Metastatic Pancreatic Cancer with Heterogeneous Effects in the Tumor. Cancer Immunol Res 2024; 12:1221-1235. [PMID: 38990554 PMCID: PMC11369625 DOI: 10.1158/2326-6066.cir-23-1073] [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: 02/07/2024] [Revised: 05/15/2024] [Accepted: 07/10/2024] [Indexed: 07/12/2024]
Abstract
Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1β blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1β (IL1β), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1β blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.
Collapse
Affiliation(s)
- Paul E. Oberstein
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Emily A. Kawaler
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Victoire Cardot-Ruffino
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| | - Osama E. Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Nina Beri
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Harshabad Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Thomas A. Abrams
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Leah H. Biller
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - James M. Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Peter Enzinger
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Brandon M. Huffman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Nadine J. McCleary
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Kimberly J. Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Douglas A. Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Benjamin L. Schlechter
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Rishi Surana
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Matthew B. Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - S. Jennifer Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Joshua Remland
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Lauren K. Brais
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - Naima Bollenrucher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Eugena Chang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Lestat R. Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Patrick J. Lenehan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| | - Igor Dolgalev
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Gregor Werba
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Cibelle Lima
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
| | - C. Elizabeth Keheler
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Keri M. Sullivan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Michael Dougan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Cristina Hajdu
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Pathology, NYU Langone Health, New York, New York.
| | - Maya Dajee
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts.
| | - Marc R. Pelletier
- Novartis Institute for Biomedical Research, Cambridge, Massachusetts.
| | | | | | - Dafna Bar-Sagi
- Department of Medicine, NYU Langone Health, New York, New York.
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Jonathan A. Nowak
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Diane M. Simeone
- Perlmutter Cancer Center, NYU Langone Health, New York, New York.
- Department of Surgery, NYU Langone Health, New York, New York.
| | - Stephanie K. Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Immunology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
7
|
Dos Passos RR, Santos CV, Priviero F, Briones AM, Tostes RC, Webb RC, Bomfim GF. Immunomodulatory Activity of Cytokines in Hypertension: A Vascular Perspective. Hypertension 2024; 81:1411-1423. [PMID: 38686582 PMCID: PMC11168883 DOI: 10.1161/hypertensionaha.124.21712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Cytokines play a crucial role in the structure and function of blood vessels in hypertension. Hypertension damages blood vessels by mechanisms linked to shear forces, activation of the renin-angiotensin-aldosterone and sympathetic nervous systems, oxidative stress, and a proinflammatory milieu that lead to the generation of neoantigens and damage-associated molecular patterns, ultimately triggering the release of numerous cytokines. Damage-associated molecular patterns are recognized by PRRs (pattern recognition receptors) and activate inflammatory mechanisms in endothelial cells, smooth muscle cells, perivascular nerves, and perivascular adipose tissue. Activated vascular cells also release cytokines and express factors that attract macrophages, dendritic cells, and lymphocytes to the blood vessels. Activated and differentiated T cells into Th1, Th17, and Th22 in secondary lymphoid organs migrate to the vessels, releasing specific cytokines that further contribute to vascular dysfunction and remodeling. This chronic inflammation alters the profile of endothelial and smooth muscle cells, making them dysfunctional. Here, we provide an overview of how cytokines contribute to hypertension by impacting the vasculature. Furthermore, we explore clinical perspectives about the modulation of cytokines as a potential therapeutic intervention to specifically target hypertension-linked vascular dysfunction.
Collapse
Affiliation(s)
- Rinaldo R Dos Passos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
| | - Cintia V Santos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - Fernanda Priviero
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Ana M Briones
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Spain (A.M.B.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.M.B.)
- CIBER Cardiovascular, Madrid, Spain (A.M.B.)
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - R Clinton Webb
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Gisele F Bomfim
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- NUPADS - Health Education and Research Center, Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil (G.F.B.)
| |
Collapse
|
8
|
Auti A, Tathode M, Marino MM, Vitiello A, Ballini A, Miele F, Mazzone V, Ambrosino A, Boccellino M. Nature's weapons: Bioactive compounds as anti-cancer agents. AIMS Public Health 2024; 11:747-772. [PMID: 39416904 PMCID: PMC11474324 DOI: 10.3934/publichealth.2024038] [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: 02/29/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 10/19/2024] Open
Abstract
Cancer represents a major global health burden, prompting continuous research for effective therapeutic strategies. Natural compounds derived from plants have emerged as potential strategies for preventing cancer and treatment because of their inherent pharmacological properties. This comprehensive review aimed to evaluate the therapeutic potential of five key natural compounds: apigenin, quercetin, piperine, curcumin, and resveratrol in cancer prevention and therapy. By examining their molecular mechanisms and preclinical evidence, this review seeks to elucidate their role as potential adjuvants or stand-alone therapies in cancer management. The exploration of natural compounds as cancer therapeutics offers several advantages, including low toxicity, wide availability, and compatibility with conventional chemotherapeutic agents. We highlighted the current understanding of their anticancer mechanisms and clinical applications for advancing personalized cancer care to improve patient outcomes. We discussed the empirical findings from in vitro, in vivo, and clinical studies reporting biological activity and therapeutic efficacy in antioxidant, immunomodulatory, anti-carcinogenic, and chemo-sensitizing modes. Innovative delivery systems and personalized treatment approaches may further enhance their bioavailability and therapeutic utility in a synergistic approach with chemo- and radiotherapeutic disease management. This review underscores the importance of natural compounds in cancer prevention and treatment, promoting a multidisciplinary approach to the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Amogh Auti
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Madhura Tathode
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Maria Michela Marino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Antonio Vitiello
- Ministry of Health, Directorate-General for Health Prevention, 00144 Rome, Italy
| | - Andrea Ballini
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122, Foggia, Italy
| | - Francesco Miele
- General Surgery Unit, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Valeria Mazzone
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Alessia Ambrosino
- Department of Experimental Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Mariarosaria Boccellino
- Department of Precision Medicine, Università della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy
| |
Collapse
|
9
|
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
|
10
|
Whately KM, Sengottuvel N, Edatt L, Srivastava S, Woods AT, Tsai YS, Porrello A, Zimmerman MP, Chack AC, Jefferys SR, Yacovone G, Kim DJ, Dudley AC, Amelio AL, Pecot CV. Spon1+ inflammatory monocytes promote collagen remodeling and lung cancer metastasis through lipoprotein receptor 8 signaling. JCI Insight 2024; 9:e168792. [PMID: 38716730 PMCID: PMC11141919 DOI: 10.1172/jci.insight.168792] [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: 01/12/2023] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world, and non-small cell lung cancer (NSCLC) is the most common subset. We previously found that infiltration of tumor inflammatory monocytes (TIMs) into lung squamous carcinoma (LUSC) tumors is associated with increased metastases and poor survival. To further understand how TIMs promote metastases, we compared RNA-Seq profiles of TIMs from several LUSC metastatic models with inflammatory monocytes (IMs) of non-tumor-bearing controls. We identified Spon1 as upregulated in TIMs and found that Spon1 expression in LUSC tumors corresponded with poor survival and enrichment of collagen extracellular matrix signatures. We observed SPON1+ TIMs mediate their effects directly through LRP8 on NSCLC cells, which resulted in TGF-β1 activation and robust production of fibrillar collagens. Using several orthogonal approaches, we demonstrated that SPON1+ TIMs were sufficient to promote NSCLC metastases. Additionally, we found that Spon1 loss in the host, or Lrp8 loss in cancer cells, resulted in a significant decrease of both high-density collagen matrices and metastases. Finally, we confirmed the relevance of the SPON1/LRP8/TGF-β1 axis with collagen production and survival in patients with NSCLC. Taken together, our study describes how SPON1+ TIMs promote collagen remodeling and NSCLC metastases through an LRP8/TGF-β1 signaling axis.
Collapse
Affiliation(s)
| | - Nisitha Sengottuvel
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Lincy Edatt
- UNC Lineberger Comprehensive Cancer Center and
| | - Sonal Srivastava
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Allison T. Woods
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Yihsuan S. Tsai
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Matthew P. Zimmerman
- UNC Lineberger Comprehensive Cancer Center and
- Department of Cell Biology and Physiology and
| | - Aaron C. Chack
- UNC Lineberger Comprehensive Cancer Center and
- Department of Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | - Dae Joong Kim
- Department of Microbiology, Immunology, and Cancer Biology and
| | - Andrew C. Dudley
- Department of Microbiology, Immunology, and Cancer Biology and
- UVA Comprehensive Cancer Center, The University of Virginia, Charlottesville, Virginia, USA
| | - Antonio L. Amelio
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Head and Neck-Endocrine Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Chad V. Pecot
- UNC Lineberger Comprehensive Cancer Center and
- Division of Oncology and
- RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Tang M, Yin Y, Wang W, Gong K, Dong J, Gao X, Li J, Fang L, Ma J, Hong Y, Li Z, Bi T, Zhang W, Liu W. Exploring the multifaceted effects of Interleukin-1 in lung cancer: From tumor development to immune modulation. Life Sci 2024; 342:122539. [PMID: 38423172 DOI: 10.1016/j.lfs.2024.122539] [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: 11/27/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/02/2024]
Abstract
Lung cancer, acknowledged as one of the most fatal cancers globally, faces limited treatment options on an international scale. The success of clinical treatment is impeded by challenges such as late diagnosis, restricted treatment alternatives, relapse, and the emergence of drug resistance. This predicament has led to a saturation point in lung cancer treatment, prompting a rapid shift in focus towards the tumor microenvironment (TME) as a pivotal area in cancer research. Within the TME, Interleukin-1 (IL-1) is abundantly present, originating from immune cells, tissue stromal cells, and tumor cells. IL-1's induction of pro-inflammatory mediators and chemokines establishes an inflammatory milieu influencing tumor occurrence, development, and the interaction between tumors and the host immune system. Notably, IL-1 expression in the TME exhibits characteristics such as staging, tissue specificity, and functional pluripotency. This comprehensive review aims to delve into the impact of IL-1 on lung cancer, encompassing aspects of occurrence, invasion, metastasis, immunosuppression, and immune surveillance. The ultimate goal is to propose a novel treatment approach, considering the intricate dynamics of IL-1 within the TME.
Collapse
Affiliation(s)
- Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yipeng Yin
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Wei Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, Jinan, Shandong 250021, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Kejian Gong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Junxue Dong
- Laboratory of Infection Oncology, Institute of Clinical Molecular Biology, Universitätsklinikum Schleswig-Holstein (UKSH), Christian Albrechts University of Kiel, Kiel, Germany
| | - Xinliang Gao
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Linan Fang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jianzun Ma
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Yang Hong
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhiqin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Taiyu Bi
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Wenyu Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
12
|
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
|
13
|
Kelly AG, Wang W, Rothenberger E, Yang J, Gilligan MM, Kipper FC, Attaya A, Gartung A, Hwang SH, Gillespie MJ, Bayer RL, Quinlivan KM, Torres KL, Huang S, Mitsiades N, Yang H, Hammock BD, Panigrahy D. Enhancing cancer immunotherapy via inhibition of soluble epoxide hydrolase. Proc Natl Acad Sci U S A 2024; 121:e2314085121. [PMID: 38330013 PMCID: PMC10873624 DOI: 10.1073/pnas.2314085121] [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: 08/16/2023] [Accepted: 12/22/2023] [Indexed: 02/10/2024] Open
Abstract
Cancer therapy, including immunotherapy, is inherently limited by chronic inflammation-induced tumorigenesis and toxicity within the tumor microenvironment. Thus, stimulating the resolution of inflammation may enhance immunotherapy and improve the toxicity of immune checkpoint inhibition (ICI). As epoxy-fatty acids (EpFAs) are degraded by the enzyme soluble epoxide hydrolase (sEH), the inhibition of sEH increases endogenous EpFA levels to promote the resolution of cancer-associated inflammation. Here, we demonstrate that systemic treatment with ICI induces sEH expression in multiple murine cancer models. Dietary omega-3 polyunsaturated fatty acid supplementation and pharmacologic sEH inhibition, both alone and in combination, significantly enhance anti-tumor activity of ICI in these models. Notably, pharmacological abrogation of the sEH pathway alone or in combination with ICI counter-regulates an ICI-induced pro-inflammatory and pro-tumorigenic cytokine storm. Thus, modulating endogenous EpFA levels through dietary supplementation or sEH inhibition may represent a unique strategy to enhance the anti-tumor activity of paradigm cancer therapies.
Collapse
Affiliation(s)
- Abigail G. Kelly
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Weicang Wang
- Department of Entomology and Nematology, University of California, Davis,CA95616
- University of California Davis Comprehensive Cancer Center, Sacramento, CA95817
- Department of Food Science, Purdue University, West Lafayette, IN47907
| | - Eva Rothenberger
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jun Yang
- Department of Entomology and Nematology, University of California, Davis,CA95616
- University of California Davis Comprehensive Cancer Center, Sacramento, CA95817
| | - Molly M. Gilligan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Franciele C. Kipper
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Ahmed Attaya
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Allison Gartung
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Sung Hee Hwang
- Department of Entomology and Nematology, University of California, Davis,CA95616
- University of California Davis Comprehensive Cancer Center, Sacramento, CA95817
| | - Michael J. Gillespie
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Rachel L. Bayer
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Katherine M. Quinlivan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Kimberly L. Torres
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Sui Huang
- Institute of Systems Biology, Seattle, WA98109
| | - Nicholas Mitsiades
- University of California Davis Comprehensive Cancer Center, Sacramento, CA95817
- Department of Internal Medicine, University of CaliforniaDavis,CA95817
| | - Haixia Yang
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Food Nutrition and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing100083, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis,CA95616
- University of California Davis Comprehensive Cancer Center, Sacramento, CA95817
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
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
|
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
|
17
|
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
|
18
|
Hommel U, Hurth K, Rondeau JM, Vulpetti A, Ostermeier D, Boettcher A, Brady JP, Hediger M, Lehmann S, Koch E, Blechschmidt A, Yamamoto R, Tundo Dottorello V, Haenni-Holzinger S, Kaiser C, Lehr P, Lingel A, Mureddu L, Schleberger C, Blank J, Ramage P, Freuler F, Eder J, Bornancin F. Discovery of a selective and biologically active low-molecular weight antagonist of human interleukin-1β. Nat Commun 2023; 14:5497. [PMID: 37679328 PMCID: PMC10484922 DOI: 10.1038/s41467-023-41190-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Human interleukin-1β (hIL-1β) is a pro-inflammatory cytokine involved in many diseases. While hIL-1β directed antibodies have shown clinical benefit, an orally available low-molecular weight antagonist is still elusive, limiting the applications of hIL-1β-directed therapies. Here we describe the discovery of a low-molecular weight hIL-1β antagonist that blocks the interaction with the IL-1R1 receptor. Starting from a low affinity fragment-based screening hit 1, structure-based optimization resulted in a compound (S)-2 that binds and antagonizes hIL-1β with single-digit micromolar activity in biophysical, biochemical, and cellular assays. X-ray analysis reveals an allosteric mode of action that involves a hitherto unknown binding site in hIL-1β encompassing two loops involved in hIL-1R1/hIL-1β interactions. We show that residues of this binding site are part of a conformationally excited state of the mature cytokine. The compound antagonizes hIL-1β function in cells, including primary human fibroblasts, demonstrating the relevance of this discovery for future development of hIL-1β directed therapeutics.
Collapse
Affiliation(s)
- Ulrich Hommel
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland.
| | - Konstanze Hurth
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland.
| | - Jean-Michel Rondeau
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Anna Vulpetti
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Daniela Ostermeier
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Jacob Peter Brady
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Michael Hediger
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Sylvie Lehmann
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Elke Koch
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Anke Blechschmidt
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Rina Yamamoto
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | | | | | - Christian Kaiser
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Philipp Lehr
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Andreas Lingel
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Luca Mureddu
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, LE1 7RH, UK
| | - Christian Schleberger
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Jutta Blank
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Paul Ramage
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Felix Freuler
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Joerg Eder
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland
| | - Frédéric Bornancin
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4002, Basel, Switzerland.
| |
Collapse
|
19
|
Liu J, Osman AEG, Bolton K, Godley LA. Germline predisposition to clonal hematopoiesis. Leuk Res 2023; 132:107344. [PMID: 37421681 DOI: 10.1016/j.leukres.2023.107344] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/10/2023]
Abstract
We now recognize that with aging, hematopoietic stem and progenitor cells (HSPCs) acquire mutations that confer a fitness advantage and clonally expand in a process now termed clonal hematopoiesis (CH). Because CH predisposes to a variety of health problems, including cancers, cardiovascular diseases, and inflammatory conditions, there is intense interest in the inherited alleles associated with the development of CH. DNA variants near TERT, SMC4, KPNA4, IL12A, CD164, and ATM confer the strongest associations. In this review, we discuss our current state of knowledge regarding germline predisposition to CH.
Collapse
Affiliation(s)
- Jie Liu
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Afaf E G Osman
- Division of Hematology and Hematologic Malignancies, University of Utah, Salt Lake City, UT, USA
| | - Kelly Bolton
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Lucy A Godley
- Division of Hematology/Oncology, Department of Medicine, and the Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
20
|
Kawashima N, Bezzerri V, Corey SJ. The Molecular and Genetic Mechanisms of Inherited Bone Marrow Failure Syndromes: The Role of Inflammatory Cytokines in Their Pathogenesis. Biomolecules 2023; 13:1249. [PMID: 37627314 PMCID: PMC10452082 DOI: 10.3390/biom13081249] [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: 07/18/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Inherited bone marrow failure syndromes (IBMFSs) include Fanconi anemia, Diamond-Blackfan anemia, Shwachman-Diamond syndrome, dyskeratosis congenita, severe congenital neutropenia, and other rare entities such as GATA2 deficiency and SAMD9/9L mutations. The IBMFS monogenic disorders were first recognized by their phenotype. Exome sequencing has validated their classification, with clusters of gene mutations affecting DNA damage response (Fanconi anemia), ribosome structure (Diamond-Blackfan anemia), ribosome assembly (Shwachman-Diamond syndrome), or telomere maintenance/stability (dyskeratosis congenita). The pathogenetic mechanisms of IBMFSs remain to be characterized fully, but an overarching hypothesis states that different stresses elicit TP53-dependent growth arrest and apoptosis of hematopoietic stem, progenitor, and precursor cells. Here, we review the IBMFSs and propose a role for pro-inflammatory cytokines, such as TGF-β, IL-1β, and IFN-α, in mediating the cytopenias. We suggest a pathogenic role for cytokines in the transformation to myeloid neoplasia and hypothesize a role for anti-inflammatory therapies.
Collapse
Affiliation(s)
- Nozomu Kawashima
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy;
| | - Seth J. Corey
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, OH 44195, USA;
| |
Collapse
|
21
|
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
|
22
|
Diwanji R, O'Brien NA, Choi JE, Nguyen B, Laszewski T, Grauel AL, Yan Z, Xu X, Wu J, Ruddy DA, Piquet M, Pelletier MR, Savchenko A, Charette L, Rodrik-Outmezguine V, Baum J, Millholland JM, Wong CC, Martin AM, Dranoff G, Pruteanu-Malinici I, Cremasco V, Sabatos-Peyton C, Jayaraman P. Targeting the IL1β Pathway for Cancer Immunotherapy Remodels the Tumor Microenvironment and Enhances Antitumor Immune Responses. Cancer Immunol Res 2023; 11:777-791. [PMID: 37040466 DOI: 10.1158/2326-6066.cir-22-0290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 12/14/2022] [Accepted: 04/07/2023] [Indexed: 04/13/2023]
Abstract
High levels of IL1β can result in chronic inflammation, which in turn can promote tumor growth and metastasis. Inhibition of IL1β could therefore be a promising therapeutic option in the treatment of cancer. Here, the effects of IL1β blockade induced by the mAbs canakinumab and gevokizumab were evaluated alone or in combination with docetaxel, anti-programmed cell death protein 1 (anti-PD-1), anti-VEGFα, and anti-TGFβ treatment in syngeneic and humanized mouse models of cancers of different origin. Canakinumab and gevokizumab did not show notable efficacy as single-agent therapies; however, IL1β blockade enhanced the effectiveness of docetaxel and anti-PD-1. Accompanying these effects, blockade of IL1β alone or in combination induced significant remodeling of the tumor microenvironment (TME), with decreased numbers of immune suppressive cells and increased tumor infiltration by dendritic cells (DC) and effector T cells. Further investigation revealed that cancer-associated fibroblasts (CAF) were the cell type most affected by treatment with canakinumab or gevokizumab in terms of change in gene expression. IL1β inhibition drove phenotypic changes in CAF populations, particularly those with the ability to influence immune cell recruitment. These results suggest that the observed remodeling of the TME following IL1β blockade may stem from changes in CAF populations. Overall, the results presented here support the potential use of IL1β inhibition in cancer treatment. Further exploration in ongoing clinical studies will help identify the best combination partners for different cancer types, cancer stages, and lines of treatment.
Collapse
Affiliation(s)
- Rohan Diwanji
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Neil A O'Brien
- Division of Hematology/Oncology, Department of Medicine, Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, California
| | - Jiyoung E Choi
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Beverly Nguyen
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Tyler Laszewski
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Angelo L Grauel
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Zheng Yan
- Oncology Translational Research, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xin Xu
- Oncology Data Sciences, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jincheng Wu
- Oncology Data Sciences, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David A Ruddy
- Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Michelle Piquet
- Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Marc R Pelletier
- Oncology Translational Research, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | | - Jason Baum
- Precision Medicine, Novartis Pharmaceuticals, Cambridge, Massachusetts
| | | | - Connie C Wong
- Precision Medicine, Novartis Pharmaceuticals, Cambridge, Massachusetts
| | - Anne-Marie Martin
- Precision Medicine, Novartis Pharmaceuticals, Cambridge, Massachusetts
| | - Glenn Dranoff
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Viviana Cremasco
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Pushpa Jayaraman
- Immuno Oncology, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| |
Collapse
|
23
|
Han Y, Zhang YY, Pan YQ, Zheng XJ, Liao K, Mo HY, Sheng H, Wu QN, Liu ZX, Zeng ZL, Yang W, Yuan SQ, Huang P, Ju HQ, Xu RH. IL-1β-associated NNT acetylation orchestrates iron-sulfur cluster maintenance and cancer immunotherapy resistance. Mol Cell 2023:S1097-2765(23)00335-0. [PMID: 37244254 DOI: 10.1016/j.molcel.2023.05.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 05/29/2023]
Abstract
Interleukin-1β (IL-1β) is a key protein in inflammation and contributes to tumor progression. However, the role of IL-1β in cancer is ambiguous or even contradictory. Here, we found that upon IL-1β stimulation, nicotinamide nucleotide transhydrogenase (NNT) in cancer cells is acetylated at lysine (K) 1042 (NNT K1042ac) and thereby induces the mitochondrial translocation of p300/CBP-associated factor (PCAF). This acetylation enhances NNT activity by increasing the binding affinity of NNT for NADP+ and therefore boosts NADPH production, which subsequently sustains sufficient iron-sulfur cluster maintenance and protects tumor cells from ferroptosis. Abrogating NNT K1042ac dramatically attenuates IL-1β-promoted tumor immune evasion and synergizes with PD-1 blockade. In addition, NNT K1042ac is associated with IL-1β expression and the prognosis of human gastric cancer. Our findings demonstrate a mechanism of IL-1β-promoted tumor immune evasion, implicating the therapeutic potential of disrupting the link between IL-1β and tumor cells by inhibiting NNT acetylation.
Collapse
Affiliation(s)
- Yi Han
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China; Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510060, P. R. China
| | - Yan-Yu Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Yi-Qian Pan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Xiao-Jun Zheng
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510060, P. R. China
| | - Kun Liao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Hai-Yu Mo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Hui Sheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Qi-Nian Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Ze-Xian Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Zhao-Lei Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Wei Yang
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510060, P. R. China
| | - Shu-Qiang Yuan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China; Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P. R. China
| | - Peng Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China
| | - Huai-Qiang Ju
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, P. R. China.
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, P. R. China; Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, P. R. China.
| |
Collapse
|
24
|
Wen J, Xuan B, Liu Y, Wang L, He L, Meng X, Zhou T, Wang Y. NLRP3 inflammasome-induced pyroptosis in digestive system tumors. Front Immunol 2023; 14:1074606. [PMID: 37081882 PMCID: PMC10110858 DOI: 10.3389/fimmu.2023.1074606] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Programmed cell death (PCD) refers to cell death in a manner that depends on specific genes encoding signals or activities. PCD includes apoptosis, pyroptosis, autophagy and necrosis (programmed necrosis). Among these mechanisms, pyroptosis is mediated by the gasdermin family and is accompanied by inflammatory and immune responses. When pathogens or other danger signals are detected, cytokine action and inflammasomes (cytoplasmic multiprotein complexes) lead to pyroptosis. The relationship between pyroptosis and cancer is complex and the effect of pyroptosis on cancer varies in different tissue and genetic backgrounds. On the one hand, pyroptosis can inhibit tumorigenesis and progression; on the other hand, pyroptosis, as a pro-inflammatory death, can promote tumor growth by creating a microenvironment suitable for tumor cell growth. Indeed, the NLRP3 inflammasome is known to mediate pyroptosis in digestive system tumors, such as gastric cancer, pancreatic ductal adenocarcinoma, gallbladder cancer, oral squamous cell carcinoma, esophageal squamous cell carcinoma, in which a pyroptosis-induced cellular inflammatory response inhibits tumor development. The same process occurs in hepatocellular carcinoma and some colorectal cancers. The current review summarizes mechanisms and pathways of pyroptosis, outlining the involvement of NLRP3 inflammasome-mediated pyroptosis in digestive system tumors.
Collapse
Affiliation(s)
- Jiexia Wen
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Bin Xuan
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Yang Liu
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Liwei Wang
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Li He
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Xiangcai Meng
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Tao Zhou
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Yimin Wang
- Department of Central Laboratory, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
- Department of General Surgery, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| |
Collapse
|
25
|
Spella M, Ntaliarda G, Skiadas G, Lamort AS, Vreka M, Marazioti A, Lilis I, Bouloukou E, Giotopoulou GA, Pepe MAA, Weiss SAI, Petrera A, Hauck SM, Koch I, Lindner M, Hatz RA, Behr J, Arendt KAM, Giopanou I, Brunn D, Savai R, Jenne DE, de Château M, Yull FE, Blackwell TS, Stathopoulos GT. Non-Oncogene Addiction of KRAS-Mutant Cancers to IL-1β via Versican and Mononuclear IKKβ. Cancers (Basel) 2023; 15:1866. [PMID: 36980752 PMCID: PMC10047096 DOI: 10.3390/cancers15061866] [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: 02/16/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Kirsten rat sarcoma virus (KRAS)-mutant cancers are frequent, metastatic, lethal, and largely undruggable. While interleukin (IL)-1β and nuclear factor (NF)-κB inhibition hold promise against cancer, untargeted treatments are not effective. Here, we show that human KRAS-mutant cancers are addicted to IL-1β via inflammatory versican signaling to macrophage inhibitor of NF-κB kinase (IKK) β. Human pan-cancer and experimental NF-κB reporter, transcriptome, and proteome screens reveal that KRAS-mutant tumors trigger macrophage IKKβ activation and IL-1β release via secretory versican. Tumor-specific versican silencing and macrophage-restricted IKKβ deletion prevents myeloid NF-κB activation and metastasis. Versican and IKKβ are mutually addicted and/or overexpressed in human cancers and possess diagnostic and prognostic power. Non-oncogene KRAS/IL-1β addiction is abolished by IL-1β and TLR1/2 inhibition, indicating cardinal and actionable roles for versican and IKKβ in metastasis.
Collapse
Affiliation(s)
- Magda Spella
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Giannoula Ntaliarda
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgios Skiadas
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Anne-Sophie Lamort
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Malamati Vreka
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Antonia Marazioti
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioannis Lilis
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Eleni Bouloukou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Georgia A. Giotopoulou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Mario A. A. Pepe
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Stefanie A. I. Weiss
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science-Core Facility Proteomics, Helmholtz Center Munich–German Research Center for Environmental Health, 80939 Munich, Germany
| | - Ina Koch
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Michael Lindner
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Rudolph A. Hatz
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich and Asklepios Medical Center, 82131 Gauting, Germany
| | - Juergen Behr
- Department of Internal Medicine V, Ludwig-Maximilian-University of Munich, 81377 Munich, Germany
| | - Kristina A. M. Arendt
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - Ioanna Giopanou
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
- Department of Internal Medicine and Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Dieter E. Jenne
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Max-Planck-Institute of Neurobiology, 82152 Planegg, Germany
| | | | - Fiona E. Yull
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Timothy S. Blackwell
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Georgios T. Stathopoulos
- Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
- Comprehensive Pneumology Center and Institute for Lung Biology and Disease, Helmholtz Center Munich-German Research Center for Environmental Health, 81377 Munich, Germany
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
26
|
Zhang Z, Li X, Wang Y, Wei Y, Wei X. Involvement of inflammasomes in tumor microenvironment and tumor therapies. J Hematol Oncol 2023; 16:24. [PMID: 36932407 PMCID: PMC10022228 DOI: 10.1186/s13045-023-01407-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/08/2023] [Indexed: 03/19/2023] Open
Abstract
Inflammasomes are macromolecular platforms formed in response to damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns, whose formation would cause maturation of interleukin-1 (IL-1) family members and gasdermin D (GSDMD), leading to IL-1 secretion and pyroptosis respectively. Several kinds of inflammasomes detecting different types of dangers have been found. The activation of inflammasomes is regulated at both transcription and posttranscription levels, which is crucial in protecting the host from infections and sterile insults. Present findings have illustrated that inflammasomes are involved in not only infection but also the pathology of tumors implying an important link between inflammation and tumor development. Generally, inflammasomes participate in tumorigenesis, cell death, metastasis, immune evasion, chemotherapy, target therapy, and radiotherapy. Inflammasome components are upregulated in some tumors, and inflammasomes can be activated in cancer cells and other stromal cells by DAMPs, chemotherapy agents, and radiation. In some cases, inflammasomes inhibit tumor progression by initiating GSDMD-mediated pyroptosis in cancer cells and stimulating IL-1 signal-mediated anti-tumor immunity. However, IL-1 signal recruits immunosuppressive cell subsets in other cases. We discuss the conflicting results and propose some possible explanations. Additionally, we also summarize interventions targeting inflammasome pathways in both preclinical and clinical stages. Interventions targeting inflammasomes are promising for immunotherapy and combination therapy.
Collapse
Affiliation(s)
- Ziqi Zhang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xue Li
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yang Wang
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Yuquan Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of China
| | - Xiawei Wei
- grid.13291.380000 0001 0807 1581Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan People’s Republic of 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
|
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
|
29
|
Abstract
Historically, cancer research and therapy have focused on malignant cells and their tumor microenvironment. However, the vascular, lymphatic and nervous systems establish long-range communication between the tumor and the host. This communication is mediated by metabolites generated by the host or the gut microbiota, as well by systemic neuroendocrine, pro-inflammatory and immune circuitries-all of which dictate the trajectory of malignant disease through molecularly defined biological mechanisms. Moreover, aging, co-morbidities and co-medications have a major impact on the development, progression and therapeutic response of patients with cancer. In this Perspective, we advocate for a whole-body 'ecological' exploration of malignant disease. We surmise that accumulating knowledge on the intricate relationship between the host and the tumor will shape rational strategies for systemic, bodywide interventions that will eventually improve tumor control, as well as quality of life, in patients with cancer.
Collapse
|
30
|
Fang X, Yu S, Jiang Y, Xiang Y, Lu K. Circulating tumor DNA detection in MRD assessment and diagnosis and treatment of non-small cell lung cancer. Front Oncol 2022; 12:1027664. [PMID: 36387176 PMCID: PMC9646858 DOI: 10.3389/fonc.2022.1027664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
Abstract
Circulating tumor DNA (ctDNA) has contributed immensely to the management of hematologic malignancy and is now considered a valuable detection tool for solid tumors. ctDNA can reflect the real-time tumor burden and be utilized for analyzing specific cancer mutations via liquid biopsy which is a non-invasive procedure that can be used with a relatively high frequency. Thus, many clinicians use ctDNA to assess minimal residual disease (MRD) and it serves as a prognostic and predictive biomarker for cancer therapy, especially for non-small cell lung cancer (NSCLC). Advanced methods have been developed to detect ctDNA, and recent clinical trials have shown the rationality and feasibility of ctDNA for identifying mutations and guiding treatments in NSCLC. Here, we have reviewed recently developed ctDNA detection methods and the importance of sequence analyses of ctDNA in NSCLC.
Collapse
Affiliation(s)
| | | | | | | | - Kaihua Lu
- Department of Oncology, The First Affiliated Hosptial of Nanjing Medicial University, Nanjing, China
| |
Collapse
|
31
|
Di Filippo M, Hennig P, Karakaya T, Slaufova M, Beer HD. NLRP1 in Cutaneous SCCs: An Example of the Complex Roles of Inflammasomes in Cancer Development. Int J Mol Sci 2022; 23:12308. [PMID: 36293159 PMCID: PMC9603439 DOI: 10.3390/ijms232012308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
Abstract
Protein complexes termed inflammasomes ensure tissue protection from pathogenic and sterile stressors by induction of inflammation. This is mediated by different caspase-1-induced downstream pathways, including activation of the pro-inflammatory cytokines proIL-1β and -18, induction of a lytic type of cell death, and regulation of the release of other pro-inflammatory molecules. Aberrant inflammasome activation underlies the pathology of numerous (auto)inflammatory diseases. Furthermore, inflammasomes support or suppress tumor development in a complex cell-type- and stage-dependent manner. In human keratinocytes and skin, NLRP1 is the central inflammasome sensor activated by cellular perturbation induced, for example, by UVB radiation. UVB represents the main inducer of skin cancer, which is the most common type of malignancy in humans. Recent evidence demonstrates that activation of NLRP1 in human skin supports the development of cutaneous squamous cell carcinomas (cSCCs) by inducing skin inflammation. In contrast, the NLRP1 inflammasome pathway is restrained in established cSCCs, suggesting that, at this stage, the protein complex has a tumor suppressor role. A better understanding of the complex functions of NLRP1 in the development of cSCCs and in general of inflammasomes in cancer might pave the way for novel strategies for cancer prevention and therapy. These strategies might include stage-specific modulation of inflammasome activation or its downstream pathways by mono- or combination therapy.
Collapse
Affiliation(s)
- Michela Di Filippo
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Paulina Hennig
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Tugay Karakaya
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Marta Slaufova
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, 8091 Zurich, Switzerland
- Faculty of Medicine, University of Zurich, 8032 Zurich, Switzerland
| |
Collapse
|
32
|
Luciano M, Krenn PW, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia. Front Immunol 2022; 13:1000996. [PMID: 36248849 PMCID: PMC9554002 DOI: 10.3389/fimmu.2022.1000996] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous malignancy of the blood and bone marrow, characterized by clonal expansion of myeloid stem and progenitor cells and rapid disease progression. Chemotherapy has been the first-line treatment for AML for more than 30 years. Application of recent high-throughput next-generation sequencing technologies has revealed significant molecular heterogeneity to AML, which in turn has motivated efforts to develop new, targeted therapies. However, due to the high complexity of this disease, including multiple driver mutations and the coexistence of multiple competing tumorigenic clones, the successful incorporation of these new agents into clinical practice remains challenging. These continuing difficulties call for the identification of innovative therapeutic approaches that are effective for a larger cohort of AML patients. Recent studies suggest that chronic immune stimulation and aberrant cytokine signaling act as triggers for AML initiation and progression, facets of the disease which might be exploited as promising targets in AML treatment. However, despite the greater appreciation of cytokine profiles in AML, the exact functions of cytokines in AML pathogenesis are not fully understood. Therefore, unravelling the molecular basis of the complex cytokine networks in AML is a prerequisite to develop new therapeutic alternatives based on targeting cytokines and their receptors.
Collapse
Affiliation(s)
- Michela Luciano
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Peter W. Krenn
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
33
|
Millet A, Pescarmona R, Belot A, Machon C, Jamilloux Y, Guitton J. Quantification of canakinumab in human plasma by liquid chromatography-high resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1211:123475. [PMID: 36179539 DOI: 10.1016/j.jchromb.2022.123475] [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: 05/07/2022] [Revised: 08/09/2022] [Accepted: 09/17/2022] [Indexed: 11/27/2022]
Abstract
Canakinumab is a fully-human monoclonal immunoglobulin gamma 1 kappa. This interleukin-1β blocker is used for the treatment of autoinflammatory diseases. Various studies have demonstrated the value of therapeutic drug monitoring of monoclonal antibodies in the management of inflammatory diseases. The purpose of this study was to develop a method to quantify canakinumab plasmatic concentration using liquid chromatography-high-resolution (Orbitrap®) mass spectrometry. The quantification was based on a bottom-up approach with the analysis of one surrogate peptide after an immunopurification of IgG followed by tryptic proteolysis. Rituximab and cetuximab, both IgG1, were tested as internal standards. Chromatographic separation was performed on a bioZenTM Peptide PS-C18 column. Mass detection was conducted in positive ionization mode with Parallel Reaction Monitoring at a resolution of 70,000. The method was fully validated in terms of linearity, sensitivity, selectivity, accuracy and matrix effect. Standards ranged from 2.5 to 75 µg/mL. Intra- and inter-day coefficients of variation ranged from 3.7 to 14.7 %, and accuracy from 97.4 to 104.1 %. This method allowed the determination of canakinumab plasmatic concentrations from eight treated patients. This method is efficient and suitable for routine use in therapeutic drug monitoring or pharmacokinetic studies.
Collapse
Affiliation(s)
- Aurélien Millet
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France
| | - Rémi Pescarmona
- Immunology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France
| | - Alexandre Belot
- Pediatric Nephrology, Rheumatology, Dermatology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France; Lyon Immunopathology FEderation (LIFE), Lyon, France
| | - Christelle Machon
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France; Analytical Chemistry Laboratory, Faculty of Pharmacy ISPBL, University Lyon 1, F-69373 Lyon, France
| | - Yvan Jamilloux
- Lyon Immunopathology FEderation (LIFE), Lyon, France; Internal Medicine, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Guitton
- Biochemistry and Pharmacology-Toxicology Laboratory, Lyon-Sud Hospital, Hospices Civils de Lyon, F-69495 Pierre Bénite, France; Toxicology Laboratory, Faculty of Pharmacy ISPBL, University of Lyon 1, F-69373 Lyon, France.
| |
Collapse
|
34
|
Mansouri S, Heylmann D, Stiewe T, Kracht M, Savai R. Cancer genome and tumor microenvironment: Reciprocal crosstalk shapes lung cancer plasticity. eLife 2022; 11:79895. [PMID: 36074553 PMCID: PMC9457687 DOI: 10.7554/elife.79895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
Lung cancer classification and treatment has been revolutionized by improving our understanding of driver mutations and the introduction of tumor microenvironment (TME)-associated immune checkpoint inhibitors. Despite the significant improvement of lung cancer patient survival in response to either oncogene-targeted therapy or anticancer immunotherapy, many patients show initial or acquired resistance to these new therapies. Recent advances in genome sequencing reveal that specific driver mutations favor the development of an immunosuppressive TME phenotype, which may result in unfavorable outcomes in lung cancer patients receiving immunotherapies. Clinical studies with follow-up after immunotherapy, assessing oncogenic driver mutations and the TME immune profile, not only reveal the underlying potential molecular mechanisms in the resistant lung cancer patients but also hold the key to better treatment choices and the future of personalized medicine. In this review, we discuss the crosstalk between cancer cell genomic features and the TME to reveal the impact of genetic alterations on the TME phenotype. We also provide insights into the regulatory role of cellular TME components in defining the genetic landscape of cancer cells during tumor development.
Collapse
Affiliation(s)
- Siavash Mansouri
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Daniel Heylmann
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Thorsten Stiewe
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Institute of Molecular Oncology, Marburg, Germany.,Member of the German Center for Lung Research (DZL), Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.,Member of the German Center for Lung Research (DZL), Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.,Member of the Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Member of the German Center for Lung Research (DZL), Giessen, Germany.,Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.,Member of the Cardio-Pulmonary Institute (CPI), Frankfurt, Germany.,Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
35
|
Olivera I, Sanz-Pamplona R, Bolaños E, Rodriguez I, Etxeberria I, Cirella A, Egea J, Garasa S, Migueliz I, Eguren-Santamaria I, Sanmamed MF, Glez-Vaz J, Azpilikueta A, Alvarez M, Ochoa MC, Malacrida B, Propper D, de Andrea CE, Berraondo P, Balkwill FR, Teijeira Á, Melero I. A Therapeutically Actionable Protumoral Axis of Cytokines Involving IL-8, TNFα, and IL-1β. Cancer Discov 2022; 12:2140-2157. [PMID: 35771565 DOI: 10.1158/2159-8290.cd-21-1115] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 05/20/2022] [Accepted: 06/26/2022] [Indexed: 11/16/2022]
Abstract
Interleukin-8 (CXCL8) produced in the tumor microenvironment correlates with poor response to checkpoint inhibitors and is known to chemoattract and activate immunosuppressive myeloid leukocytes. In human cancer, IL8 mRNA levels correlate with IL1B and TNF transcripts. Both cytokines induced IL-8 functional expression from a broad variety of human cancer cell lines, primary colon carcinoma organoids, and fresh human tumor explants. Although IL8 is absent from the mouse genome, a similar murine axis in which TNFα and IL-1β upregulate CXCL1 and CXCL2 in tumor cells was revealed. Furthermore, intratumoral injection of TNFα and IL-1β induced IL-8 release from human malignant cells xenografted in immunodeficient mice. In all these cases, the clinically used TNFα blockers infliximab and etanercept or the IL-1β inhibitor anakinra was able to interfere with this pathogenic cytokine loop. Finally, in paired plasma samples of patients with cancer undergoing TNFα blockade with infliximab in a clinical trial, reductions of circulating IL-8 were substantiated. SIGNIFICANCE IL-8 attracts immunosuppressive protumor myeloid cells to the tumor microenvironment, and IL-8 levels correlate with poor response to checkpoint inhibitors. TNFα and IL-1β are identified as major inducers of IL-8 expression on malignant cells across cancer types and models in a manner that is druggable with clinically available neutralizing agents. This article is highlighted in the In This Issue feature, p. 2007.
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
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Oncobell Program, Catalan Institute of Cancer (ICO), Bellvitge Biomedical Research Institute (IDIBELL), CIBERESP, Hospitalet de Llobregat, Barcelona, Spain and ARAID Researcher, Aragon Health Research institute (IIS Aragon), Zaragoza, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Inmaculada Rodriguez
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Iñaki Etxeberria
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Josune Egea
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Itziar Migueliz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona 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
| | - Miguel F Sanmamed
- 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
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona Spain
| | - Maite Alvarez
- 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
| | - María C Ochoa
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Beatrice Malacrida
- Center for tumour microenvironment, Barts Cancer Institute, Queen Mary University of London, London, United Kindgom
| | - David Propper
- Center for tumour microenvironment, Barts Cancer Institute, Queen Mary University of London, London, United Kindgom
| | - Carlos E de Andrea
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Pathology, 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
| | - Frances R Balkwill
- Center for tumour microenvironment, Barts Cancer Institute, Queen Mary University of London, London, United Kindgom
| | - Álvaro 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
| | - 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
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
36
|
Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov 2022; 21:799-820. [PMID: 35974096 PMCID: PMC9380983 DOI: 10.1038/s41573-022-00520-5] [Citation(s) in RCA: 659] [Impact Index Per Article: 329.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
Tumour-associated macrophages are an essential component of the tumour microenvironment and have a role in the orchestration of angiogenesis, extracellular matrix remodelling, cancer cell proliferation, metastasis and immunosuppression, as well as in resistance to chemotherapeutic agents and checkpoint blockade immunotherapy. Conversely, when appropriately activated, macrophages can mediate phagocytosis of cancer cells and cytotoxic tumour killing, and engage in effective bidirectional interactions with components of the innate and adaptive immune system. Therefore, they have emerged as therapeutic targets in cancer therapy. Macrophage-targeting strategies include inhibitors of cytokines and chemokines involved in the recruitment and polarization of tumour-promoting myeloid cells as well as activators of their antitumorigenic and immunostimulating functions. Early clinical trials suggest that targeting negative regulators (checkpoints) of myeloid cell function indeed has antitumor potential. Finally, given the continuous recruitment of myelomonocytic cells into tumour tissues, macrophages are candidates for cell therapy with the development of chimeric antigen receptor effector cells. Macrophage-centred therapeutic strategies have the potential to complement, and synergize with, currently available tools in the oncology armamentarium. Macrophages can promote tumorigenesis and enhance the antitumour response. This Review discusses the molecular mechanisms underlying the reprogramming of macrophages in the tumour microenvironment and provides an overview of macrophage-targeted therapies for the treatment of cancer.
Collapse
Affiliation(s)
- Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy. .,IRCCS- Humanitas Research Hospital, Milan, Italy. .,The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Paola Allavena
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| | - Federica Marchesi
- IRCCS- Humanitas Research Hospital, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.,IRCCS- Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
37
|
Sanmamed MF, Berraondo P, Rodriguez-Ruiz ME, Melero I. Charting roadmaps towards novel and safe synergistic immunotherapy combinations. NATURE CANCER 2022; 3:665-680. [PMID: 35764745 DOI: 10.1038/s43018-022-00401-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Checkpoint inhibitor-based cancer immunotherapy is often combined in the clinic with other immunotherapy strategies, targeted therapies, chemotherapy or standard-of-care treatments to achieve superior therapeutic efficacy. The large number of immunotherapy combinations that are currently undergoing clinical testing necessitate the establishment of faithful criteria to prioritize optimal combinations with evidence of synergy, to determine their safety and optimal sequence of administration and to identify biomarkers of therapy resistance and response. In this review, we focus on recent developments in immunotherapy combinations and reflect on how combinations should be optimized to maximize the impact of immunotherapy in clinical oncology.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| |
Collapse
|
38
|
Pretre V, Papadopoulos D, Regard J, Pelletier M, Woo J. Interleukin-1 (IL-1) and the inflammasome in cancer. Cytokine 2022; 153:155850. [PMID: 35279620 DOI: 10.1016/j.cyto.2022.155850] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/13/2022] [Accepted: 03/03/2022] [Indexed: 12/14/2022]
Abstract
Numerous preclinical and clinical studies have demonstrated the significant contribution of inflammation to the development and progression of various types of cancer. Inflammation in the tumor microenvironment mediates complex interactions between innate immunity, adaptive immunity, microbiomes and stroma, and ultimately alters the overall fitness of tumor cells at multiple stages of carcinogenesis. Malignancies are known to arise in areas of chronic inflammation and inflammation in the tumor microenvironment (often called tumor-promoting inflammation) is believed to allow cancer cells to evade immunosurveillance while promoting genetic instability, survival and progression. Among the strongest data suggesting a causal role for inflammation in cancer come from the recent CANTOS trial which demonstrated that interleukin-1β (IL-1β) inhibition with canakinumab leads to a significant, dose-dependent decrease in incident lung cancer. This observation has launched a series of additional clinical studies to understand the role of IL-1β and the inflammasome in cancer, and the clinical utility of IL-1β inhibition in different stages of lung cancer. In this article we will review recent data implicating IL-1β signaling and its upstream regulator NLRP3 in both solid tumor and hematologic malignancies. We will discuss the key preclinical observations and the current clinical landscape, and describe the pharmacologic tools which will be used to evaluate the effects of blocking tumor-promoting inflammation clinically.
Collapse
|
39
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
40
|
Abstract
In a recent publication in Nature Immunology, Bruchard et al. report that type 3 innate lymphoid cells, activated and recruited by cisplatin-induced chemokine ligand 20 (CCL20) and interleukin-1 beta (IL-1β), promote CD4 and CD8 T cell infiltration into murine tumors. This turns "cold" tumors "hot", thereby enhancing responsiveness to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Kingston H G Mills
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
41
|
Guerville F, Bourdel-Marchasson I, Déchanet-Merville J, Pellegrin I, Soubeyran P, Appay V, Lemoine M. Does Inflammation Contribute to Cancer Incidence and Mortality during Aging? A Conceptual Review. Cancers (Basel) 2022; 14:1622. [PMID: 35406394 PMCID: PMC8996949 DOI: 10.3390/cancers14071622] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/11/2022] [Accepted: 03/19/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is associated with chronic low-grade inflammation, cancer incidence and mortality. As inflammation contributes to cancer initiation and progression, one could hypothesize that age-associated chronic low-grade inflammation contributes to the increase in cancer incidence and/or mortality observed during aging. Here, we review the evidence supporting this hypothesis: (1) epidemiological associations between biomarkers of systemic inflammation and cancer incidence and mortality in older people, (2) therapeutic clues suggesting that targeting inflammation could reduce cancer incidence and mortality and (3) experimental evidence from animal models highlighting inflammation as a link between various mechanisms of aging and cancer initiation and progression. Despite a large body of literature linking aging, inflammation and cancer, convincing evidence for the clear implication of specific inflammatory pathways explaining cancer incidence or mortality during aging is still lacking. Further dedicated research is needed to fill these gaps in evidence and pave the way for the development of applications in clinical care.
Collapse
Affiliation(s)
- Florent Guerville
- ImmunoConcEpT, CNRS UMR5164, INSERM ERL1303, Université de Bordeaux, F-33076 Bordeaux, France; (J.D.-M.); (I.P.); (V.A.); (M.L.)
- Clinical Gerontology Department, Bordeaux University Hospital, F-33000 Bordeaux, France;
| | - Isabelle Bourdel-Marchasson
- Clinical Gerontology Department, Bordeaux University Hospital, F-33000 Bordeaux, France;
- CRMSB, CNRS UMR 5536, Université de Bordeaux, F-33000 Bordeaux, France
| | - Julie Déchanet-Merville
- ImmunoConcEpT, CNRS UMR5164, INSERM ERL1303, Université de Bordeaux, F-33076 Bordeaux, France; (J.D.-M.); (I.P.); (V.A.); (M.L.)
| | - Isabelle Pellegrin
- ImmunoConcEpT, CNRS UMR5164, INSERM ERL1303, Université de Bordeaux, F-33076 Bordeaux, France; (J.D.-M.); (I.P.); (V.A.); (M.L.)
- Laboratory of Immunology and Immunogenetics, Bordeaux University Hospital, F-33000 Bordeaux, France
| | - Pierre Soubeyran
- Department of Medical Oncology, Institut Bergonie, F-33076 Bordeaux, France;
| | - Victor Appay
- ImmunoConcEpT, CNRS UMR5164, INSERM ERL1303, Université de Bordeaux, F-33076 Bordeaux, France; (J.D.-M.); (I.P.); (V.A.); (M.L.)
| | - Maël Lemoine
- ImmunoConcEpT, CNRS UMR5164, INSERM ERL1303, Université de Bordeaux, F-33076 Bordeaux, France; (J.D.-M.); (I.P.); (V.A.); (M.L.)
| |
Collapse
|
42
|
Role of FSCN1 in the Tumor Microenvironment of Lung Squamous Cell Carcinoma. Immunobiology 2022; 227:152206. [DOI: 10.1016/j.imbio.2022.152206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 11/22/2022]
|
43
|
Wang C, Ma H, Wu W, Lu X. Drug Discovery in Spinal Cord Injury With Ankylosing Spondylitis Identified by Text Mining and Biomedical Databases. Front Genet 2022; 13:799970. [PMID: 35281834 PMCID: PMC8914062 DOI: 10.3389/fgene.2022.799970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/19/2022] [Indexed: 11/15/2022] Open
Abstract
Spinal cord injury (SCI) and ankylosing spondylitis (AS) are common inflammatory diseases in spine surgery. However, it is a project where the relationship between the two diseases is ambiguous and the efficiency of drug discovery is limited. Therefore, the study aimed to investigate new drug therapies for SCI and AS. First, text mining was used to obtain the interacting genes related to SCI and AS, and then, the functional analysis was conducted. Protein–protein interaction (PPI) networks were constructed by STRING online and Cytoscape software to identify hub genes. Last, hub genes and potential drugs were performed after undergoing drug–gene interaction analysis, and MicroRNA and transcription factors regulatory networks were also analyzed. Two hundred five genes common to “SCI” and “AS” identified by text mining were enriched in inflammatory responses. PPI network analysis showed that 30 genes constructed two significant modules. Ultimately, nine (SST, VWF, IL1B, IL6, CXCR4, VEGFA, SERPINE1, FN1, and PROS1) out of 30 genes could be targetable by a total of 13 drugs. In conclusion, the novel core genes contribute to a novel insight for latent functional mechanisms and present potential prognostic indicators and therapeutic targets in SCI and AS.
Collapse
|
44
|
The Immune Underpinnings of Barrett's-Associated Adenocarcinogenesis: a Retrial of Nefarious Immunologic Co-Conspirators. Cell Mol Gastroenterol Hepatol 2022; 13:1297-1315. [PMID: 35123116 PMCID: PMC8933845 DOI: 10.1016/j.jcmgh.2022.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022]
Abstract
There is no doubt that chronic gastroesophageal reflux disease increases the risk of esophageal adenocarcinoma (EAC) by several fold (odds ratio, 6.4; 95% CI, 4.6-9.1), and some relationships between reflux disease-mediated inflammation and oncogenic processes have been explored; however, the precise interconnections between the immune response and genomic instabilities underlying these pathologic processes only now are emerging. Furthermore, the precise cell of origin of the precancerous stages associated with EAC development, Barrett's esophagus, be it cardia resident or embryonic remnant, may shape our interpretation of the likely immune drivers. This review integrates the current collective knowledge of the immunology underlying EAC development and outlines a framework connecting proinflammatory pathways, such as those mediated by interleukin 1β, tumor necrosis factor α, leukemia inhibitory factor, interleukin 6, signal transduction and activator of transcription 3, nuclear factor-κB, cyclooxygenase-2, and transforming growth factor β, with oncogenic pathways in the gastroesophageal reflux disease-Barrett's esophagus-EAC cancer sequence. Further defining these immune and molecular railroads may show a map of the routes taken by gastroesophageal cells on their journey toward EAC tumor phylogeny. The selective pressures applied by this immune-induced journey likely impact the phenotype and genotype of the resulting oncogenic destination and further exploration of lesser-defined immune drivers may be useful in future individualized therapies or enhanced selective application of recent immune-driven therapeutics.
Collapse
|
45
|
Zhang J, Veeramachaneni N. Targeting interleukin-1β and inflammation in lung cancer. Biomark Res 2022; 10:5. [PMID: 35086565 PMCID: PMC8796434 DOI: 10.1186/s40364-021-00341-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammation is a process that protects organs against various potentially harmful stimuli and enables repair. Dysregulated inflammation, however, damages tissues and leads to disease, including cancer. Cancer-related inflammation is characterized by cytokine production, leukocyte infiltration, angiogenesis, and tissue remodeling-all critical processes in modulating the tumor microenvironment (TME). The TME is known to play a key role in tumor progression, and targeting its immune component to achieve a better anti-tumor response is the basis of immunotherapy. Despite the critical role cytokines play in the TME and tumor progression, there is currently only one therapy approved by the FDA that directly involves cytokine signaling: human recombinant interleukin-2 protein, aldesleukin. The recent Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) trial evaluated the use of anti-interleukin-1β therapy in atherosclerotic disease; however, it also revealed interleukin-1β (IL-1β) blockade with canakinumab led to a significantly lower incidence of lung cancer. This has opened a promising new avenue for lung cancer therapy, and strategies using anti-IL-1β therapy alone or in combination with chemotherapy and/or immune checkpoint blockade are currently being evaluated in several clinical trials.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Nirmal Veeramachaneni
- Department of Cardiovascular and Thoracic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160 , USA
| |
Collapse
|
46
|
A Comparative and Comprehensive Review of Antibody Applications in the Treatment of Lung Disease. Life (Basel) 2022; 12:life12010130. [PMID: 35054524 PMCID: PMC8778790 DOI: 10.3390/life12010130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Antibodies are a type of protein produced by active B cells in response to antigen stimulation. A series of monoclonal antibodies and neutralizing antibodies have been invented and put into clinical use because of their high therapeutic effect and bright developing insight. Patients with cancer, infectious diseases, and autoimmune diseases can all benefit from antibody therapy. However, the targeting aspects and potential mechanisms for treating these diseases differ. In the treatment of patients with infectious diseases such as COVID-19, neutralizing antibodies have been proposed as reliable vaccines against COVID-19, which target the ACE2 protein by preventing virus entry into somatic cells. Monoclonal antibodies can target immune checkpoints (e.g., PD-L1 and CTLA-4), tyrosine kinase and subsequent signaling pathways (e.g., VEGF), and cytokines in cancer patients (e.g. IL-6 and IL-1β). It is debatable whether there is any connection between the use of antibodies in these diseases. It would be fantastic to discover the related points and explain the burden for the limitation of cross-use of these techniques. In this review, we provided a comprehensive overview of the use of antibodies in the treatment of infectious disease and cancer patients. There are also discussions of their mechanisms and history. In addition, we discussed our future outlook on the use of antibodies.
Collapse
|
47
|
Suzuki J, Tsuboi M, Ishii G. Cancer-associated fibroblasts and the tumor microenvironment in non-small cell lung cancer. Expert Rev Anticancer Ther 2022; 22:169-182. [PMID: 34904919 DOI: 10.1080/14737140.2022.2019018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) has a markedly poor prognosis as it progresses, and the prognosis is still unsatisfactory even with modern treatments. Cancer is composed of not only cancer cells, but also stroma consisting of various cell types. Cancer-associated fibroblasts (CAFs) are a major component of the stroma and the associated tumor microenvironment (TME). Particularly, CAFs are a critical component in elucidating the biological mechanisms of cancer progression and new therapeutic targets. This article outlines the TME formed by CAFs in NSCLC. AREAS COVERED Focusing on the TME in NSCLC, we discuss the mechanisms by which CAFs are involved in cancer progression, drug resistance, and the development of therapies targeting CAFs. EXPERT OPINION In the TME, CAFs profoundly contribute to tumor progression by interacting with cancer cells through direct contact or paracrine cytokine signaling. CAFs also interact with various other stromal components to establish a tumor-promoting immunosuppressive microenvironment and remodel the extracellular matrix. Furthermore, these effects are closely associated with drug resistance. Further elucidation of the stromal microenvironment, including CAFs, could prove to be crucial in the treatment of NSCLC.
Collapse
Affiliation(s)
- Jun Suzuki
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
48
|
Dosch AR, Singh S, Nagathihalli NS, Datta J, Merchant NB. Interleukin-1 signaling in solid organ malignancies. Biochim Biophys Acta Rev Cancer 2021; 1877:188670. [PMID: 34923027 DOI: 10.1016/j.bbcan.2021.188670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
As inflammation plays a critical role in the development and progression of cancer, therapeutic targeting of cytokine pathways involved in both tumorigenesis and dictating response to clinical treatments are of significant interest. Recent evidence has highlighted the importance of the pro-inflammatory cytokine interleukin-1 (IL-1) as a key mediator of tumor growth, metastatic disease spread, immunosuppression, and drug resistance in cancer. IL-1 promotes tumorigenesis through diverse mechanisms, including the activation of oncogenic signaling pathways directly in tumor cells and via orchestrating crosstalk between the cellular constituents of the tumor microenvironment (TME), thereby driving cancer growth. This review will provide an overview of IL-1 signaling and physiology and summarize the disparate mechanisms involving IL-1 in tumorigenesis and cancer progression. Additionally, clinical studies targeting IL-1 signaling in the management of solid organ tumors will be summarized herein.
Collapse
Affiliation(s)
- Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Samara Singh
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America.
| |
Collapse
|
49
|
Neganova M, Liu J, Aleksandrova Y, Klochkov S, Fan R. Therapeutic Influence on Important Targets Associated with Chronic Inflammation and Oxidative Stress in Cancer Treatment. Cancers (Basel) 2021; 13:6062. [PMID: 34885171 PMCID: PMC8657135 DOI: 10.3390/cancers13236062] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/28/2021] [Accepted: 11/28/2021] [Indexed: 01/17/2023] Open
Abstract
Chronic inflammation and oxidative stress are the interconnected pathological processes, which lead to cancer initiation and progression. The growing level of oxidative and inflammatory damage was shown to increase cancer severity and contribute to tumor spread. The overproduction of reactive oxygen species (ROS), which is associated with the reduced capacity of the endogenous cell defense mechanisms and/or metabolic imbalance, is the main contributor to oxidative stress. An abnormal level of ROS was defined as a predisposing factor for the cell transformation that could trigger pro-oncogenic signaling pathways, induce changes in gene expression, and facilitate accumulation of mutations, DNA damage, and genomic instability. Additionally, the activation of transcription factors caused by a prolonged oxidative stress, including NF-κB, p53, HIF1α, etc., leads to the expression of several genes responsible for inflammation. The resulting hyperactivation of inflammatory mediators, including TNFα, TGF-β, interleukins, and prostaglandins can contribute to the development of neoplasia. Pro-inflammatory cytokines were shown to trigger adaptive reactions and the acquisition of resistance by tumor cells to apoptosis, while promoting proliferation, invasion, and angiogenesis. Moreover, the chronic inflammatory response leads to the excessive production of free radicals, which further aggravate the initiated reactions. This review summarizes the recent data and progress in the discovery of mechanisms that associate oxidative stress and chronic inflammation with cancer onset and metastasis. In addition, the review provides insights for the development of therapeutic approaches and the discovery of natural substances that will be able to simultaneously inhibit several key oncological and inflammation-related targets.
Collapse
Affiliation(s)
- Margarita Neganova
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Sergey Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China; (M.N.); (J.L.)
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
50
|
Melero I, Teijeira A, Aranda F, Berraondo P. Firefighters for the Wrong Type of Inflammation in Tumors. Cancer Discov 2021; 11:2372-2374. [PMID: 34598950 DOI: 10.1158/2159-8290.cd-21-1004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this issue of Cancer Discovery, Pelly and colleagues show that inhibition of prostaglandin E2 synthesis or its activity on EP2 and EP4 receptors synergizes with anti-PD-1 immunotherapy and triggers a potent intratumoral IFNγ response in mouse models and in fresh surgical human tumor explants. This therapeutic strategy is in line with other interventions that aim at fostering immunotherapy by means of quenching protumor inflammation.See related article by Pelly et al., p. 2602.
Collapse
Affiliation(s)
- Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain. .,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|