1
|
Xia F, Sha Y, Jin Y, Yang J, Chen C, Gong B, Liu Y, Zhao Q. Autophagy inhibition amplifies anti-tumor immunity effect of dinutuximab beta on neuroblastoma via the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Int Immunopharmacol 2025; 158:114862. [PMID: 40378433 DOI: 10.1016/j.intimp.2025.114862] [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: 06/24/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Dinutuximab beta has shown limited efficacy in treating high-risk neuroblastoma (NB). Combining autophagy inhibitors with immune checkpoint inhibitors (ICIs) has proven effective in many malignancies. However, the anti-tumor effects of autophagy inhibition in conjunction with anti-GD2 immunotherapy remain unknown. In this study, dinutuximab beta induces anti-proliferation and anti-EMT activity in NB cells. Dinutuximab beta also triggers autophagy in NB cells, and inhibition of the VEGFR pathway with anlotinib amplifies dinutuximab beta-induced autophagy. In addition, dinutuximab beta induces the synthesis of the chemokine CXCL9 and the infiltration of CD8+ T cells. Mechanistically, dinutuximab beta inhibits the VEGFR/AKT/mTOR and ROS/NF-κB pathways. Furthermore, autophagy inhibition by CQ enhances CXCL9 expression and anti-tumor T cell responses of single anti-GD2 therapy in vitro and in vivo. Collectively, this study suggests autophagy inhibitors may be a promising strategy for enhancing therapeutic efficacy in NB in conjunction with anti-GD2 immunotherapy.
Collapse
Affiliation(s)
- Fantong Xia
- Radiation Oncology Center, Chongqing University Cancer Hospital, College of Medicine, Chongqing University, Chongqing, China
| | - Yongliang Sha
- Department of General Surgery, Xuzhou Central Hospital, Xuzhou, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yun Liu
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
2
|
Ye J, Qin SS, Hughson AL, Hannon G, Salama NA, Vrooman TG, Lesch ML, Lesser S, Eckl SL, Jewell R, Benoodt L, Mills BN, Johnston CJ, Lord E, Belt BA, Calvi LM, Linehan D, Luheshi N, Eyles J, Gerber SA. Blocking LIF and PD-L1 enhances the antitumor efficacy of SBRT in murine PDAC models. J Immunother Cancer 2025; 13:e010820. [PMID: 40341024 PMCID: PMC12067785 DOI: 10.1136/jitc-2024-010820] [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: 10/13/2024] [Accepted: 04/21/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Recent preclinical and clinical data suggest that leukemia inhibitory factor (LIF) is a potential target for various tumor types including pancreatic ductal adenocarcinoma as LIF is involved in multiple protumor processes including cancer stem cell maintenance, epithelial-mesenchymal transition (EMT), immunosuppression, and chemo/radioresistance. Anti-LIF antibody therapy has demonstrated safety and tolerability but limited efficacy in phase 1 clinical trial in advanced solid tumors. This prompted us to explore combination therapies, suggesting that LIF blockade, when combined with standard-of-care chemotherapy, radiotherapy, and/or immunotherapy, could present a promising therapeutic strategy. METHODS We evaluated the impact of combining systemic inhibition of LIF/programmed death-ligand 1 (PD-L1) with localized stereotactic body radiotherapy (SBRT) on tumor progression across multiple murine orthotopic pancreatic tumor models and examined systemic antitumor immunity using a hepatic rechallenge model. The antitumor immune response was characterized throughflow cytometry and Luminex assays. To identify differentially expressed genes and signaling pathways following treatment, we performed bulk RNA sequencing on pancreatic tumors. Additionally, single-cell RNA sequencing was conducted to further examine changes in tumor-infiltrating immune cells and their signaling pathways. RESULTS We showed that simultaneous inhibition of LIF and PD-L1 significantly amplified the antitumor efficacy of SBRT, resulting in extended survival. The triple therapy (SBRT+anti-LIF+anti-PD-L1) generated an immunostimulatory tumor microenvironment, characterized by a proinflammatory shift in the cytokine/chemokine profile, increased infiltration of effector CD8+ T cells, and upregulated activation or maturation signals in tumor-infiltrating CD8+ T cells and macrophages. The beneficial effects of triple therapy were mostly abrogated by depletion of CD8+ T cells. In addition, triple therapy downregulated pathways related to tumor stemness, proliferation, and metabolism, and reduced EMT. Importantly, the combination of local SBRT treatment with systemic LIF and PD-L1 blockade resulted in long-term systemic antitumor memory.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Shuyang S Qin
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Angela L Hughson
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Noah A Salama
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tara G Vrooman
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Maggie L Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Sidney Lesser
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Sarah L Eckl
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Rachel Jewell
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lauren Benoodt
- Genomic Research Center, University of Rochester Medical Center, Rochester, New York, USA
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Edith Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Laura M Calvi
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Medicine Hematology/Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - David Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Nadia Luheshi
- The Discovery Center, Cambridge Biomedical Campus, Cambridge, UK
| | - Jim Eyles
- Oncology R&D, Research and Early Development, AstraZeneca R&D, Cambridge, Cambridgeshire, UK
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
3
|
Chao YL, Zhou KI, Forbes KK, Porrello A, Gentile GM, Zhu Y, Chack AC, John Mary DJS, Liu H, Cockman E, Edatt L, Goda GA, Zhao JJ, Abou Assi H, Wiedner HJ, Tsai Y, Wilkinson L, Van Swearingen AED, Carey LA, Giudice J, Dominguez D, Holley CL, Pecot CV. Snord67 promotes breast cancer metastasis by guiding U6 modification and modulating the splicing landscape. Nat Commun 2025; 16:4118. [PMID: 40316533 PMCID: PMC12048515 DOI: 10.1038/s41467-025-59406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Previously considered "housekeeping" genes, small nucleolar RNAs (snoRNAs) are increasingly understood to have wide-ranging functions in cancer, yet their role in metastasis has been less well studied. Here, we identify the snoRNA Snord67 as a regulator of lymph node (LN) metastasis in breast cancer. Snord67 expression is enriched in LN metastases in an immune-competent mouse model of female breast cancer. In an orthotopic breast cancer model, loss of Snord67 decreases LN metastasis. In a model of lymphatic metastasis, Snord67 loss decreases LN tumor growth and distant metastases. In breast cancer cell lines, Snord67 knockout results in loss of targeted 2'-O-methylation on U6 small nuclear RNA, as well as widespread changes in splicing. Together, these results demonstrate that Snord67 regulates splicing and promotes the growth of LN metastases and subsequent spread to distant metastases. SnoRNA-guided modifications of the spliceosome and regulation of splicing may represent a potentially targetable pathway in cancer.
Collapse
Affiliation(s)
- Yvonne L Chao
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
- VA Pittsburgh Health System, Pittsburgh, PA, USA
| | - Katherine I Zhou
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kwame K Forbes
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alessandro Porrello
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gabrielle M Gentile
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yinzhou Zhu
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Aaron C Chack
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dixcy J S John Mary
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haizhou Liu
- Division of Hematology & Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Cockman
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Lincy Edatt
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Grant A Goda
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Justin J Zhao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hala Abou Assi
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Hannah J Wiedner
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yihsuan Tsai
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Lily Wilkinson
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jimena Giudice
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology (GMB), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Dominguez
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Christopher L Holley
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Chad V Pecot
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.
- Division of Hematology & Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Saeed MU, Choudhury A, Ansari J, Mohammad T, Hussain A, Fatima U, Alajmi MF, Hassan MI. Discovering potential therapeutic targets in glioblastoma multiforme using a multi-omics approach. Pathol Res Pract 2025; 269:155942. [PMID: 40168776 DOI: 10.1016/j.prp.2025.155942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/08/2025] [Accepted: 03/09/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor associated with high fatality rates, poor prognosis, and limited treatment options. In this study, we utilized RNA-Seq gene count data from GBM patients, sourced from the Gene Expression Omnibus (GEO) database, to conduct an in-depth analysis of gene expression patterns. METHODS Our investigation involved stratifying samples into two distinct sets, Group I and Group II, comparing normal, low-grade, and GBM tumor samples, respectively. Subsequently, we performed differential expression analysis and enrichment analysis to uncover significant gene signatures. To elucidate the protein-protein interactions associated with GBM, we used the STRING plugin within Cytoscape for comprehensive network visualization and analysis. RESULTS By applying Maximal clique centrality (MCC) scores, we identified a set of 10 hub genes in each group. These hub genes were subjected to survival analysis, highlighting their prognostic relevance. In Group I, comprising BUB1, DLGAP5, BUB1B, CDK1, TOP2A, CDC20, KIF20A, ASPM, BIRC5, and CCNB2, these genes emerged as potential biomarkers associated with the transition to low-grade tumors. In Group II, genes such as LIF, LBP, CSF3, IL6, CCL2, SAA1, CCL20, MMP9, CXCL10, and MMP1 were found to be involved in the transformation to adult glioblastoma. Kaplan-Meier's overall survival analysis of these hub genes revealed that modifications, particularly the upregulation of these candidate genes, were associated with reduced survival in GBM patients. CONCLUSIONS The findings established the significance of genomic alterations and differential gene expression in GBM, presenting opportunities for prognostic and targeted therapeutic interventions. This study provides valuable insights into potential avenues for enhancing the clinical management of GBM.
Collapse
Affiliation(s)
- Mohammad Umar Saeed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Urooj Fatima
- UAMS, Donald W. Reynolds Institute on Aging, Department of Geriatrics, Little Rock, AR 72205, USA
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
5
|
Nagase Y, Kodama M, Aimono E, Nakamura K, Takamatsu R, Abe K, Yoshimura T, Chiyoda T, Yamagami W, Nishihara H. CXCL9 and CXCL13 shape endometrial cancer immune-activated microenvironment via tertiary lymphoid structure formation. Cancer Sci 2025; 116:1193-1202. [PMID: 39960836 PMCID: PMC12044659 DOI: 10.1111/cas.16371] [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: 06/18/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has been successfully applied to various cancers; however, not all patients respond to ICI therapy. Tumors with an immune-activated environment are highly responsive to ICIs. To identify the cells and molecules essential to the formation of an immune-activated cancer microenvironment, we focused on the tertiary lymphoid structure (TLS) and performed histological and genomic analyses using endometrial cancer material. In the high immunogenic group, numerous TLSs were observed, and CXCL9 and CXCL13 expression was markedly increased. CXCL9-positive antigen-presenting and CXCL13-positive follicular dendritic cells were distributed in the T- and B-cell zones of TLSs, respectively. A group of molecules whose expression was upregulated along with CXCL9 and CXCL13 expression was strongly associated with cellular immunity. These results suggest that CXCL9-expressing antigen-presenting cells and CXCL13-expressing follicular dendritic cells coordinately shape the immune-activated microenvironment through TLS formation. The current findings will contribute to a better understanding of the mechanisms underlying the activated cancer immune microenvironment, thereby advancing the field of precision cancer medicine.
Collapse
Affiliation(s)
| | - Makoto Kodama
- Department of PathologyTokyo Yamate Medical CenterTokyoJapan
- Department of Human Pathology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Eriko Aimono
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Reika Takamatsu
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| | - Keiko Abe
- Department of PathologyTokyo Yamate Medical CenterTokyoJapan
| | - Takuma Yoshimura
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Wataru Yamagami
- Department of Obstetrics and GynecologyKeio University School of MedicineTokyoJapan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer CenterKeio University School of MedicineTokyoJapan
| |
Collapse
|
6
|
Fan Z, Yi Z, Li S, He J. Parabacteroides distasonis promotes CXCL9 secretion of tumor-associated macrophages and enhances CD8 +T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in non-small cell lung cancer mice. BMC Biotechnol 2025; 25:30. [PMID: 40241108 PMCID: PMC12004837 DOI: 10.1186/s12896-025-00963-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Parabacteroides distasonis (P. distasonis) could regulate inflammatory markers, promote intestinal barrier integrity, and block tumor formation in colon. However, the regulatory effect of P. distasonis on non-small cell lung cancer (NSCLC) remains unknown. This study aimed to investigate the regulatory effect of P. distasonis on NSCLC and its impact on tumor immunity. METHODS We first established a mouse model of Lewis lung cancer, and administered P. distasonis and intrabitoneal injection of anti-mouse PD-1 monoclonal antibody to assess the impact of P. distasonis on tumor immunity, and mouse intestinal barrier. Then, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secretion mediated by tumor-associated macrophages (TAM). We used the TLR1/2 complex inhibitor CPT22 to evaluate its effect on macrophage activation. Finally, we explored the effect of P. distasonis on CD8+T cells and CXCL9 secreted by TAM in vivo. RESULTS In vivo, P. distasonis enhanced anti-tumor effects of anti-PD-1 in NSCLC mice, improved intestinal barrier integrity, recruited macrophages, and promoted M1 polarization. In vitro, CD86 and iNOS levels in BMDM were elevated and CD206 and Arg1 levels were suppressed in membrane fraction of P. distasonis (PdMb) group in comparison to Control group. With additional CPT22 pre-treatment, the levels of CD86 and iNOS in BMDM were reduced, and the levels of CD206 and Arg1 were increased. Compared to PBS group, P. distasonis group exhibited higher proportion of CD8+T cells in tumor tissues, along with increased positive proportion of GZMB and IFN-γ in CD8+T cells. Additionally, in comparison to Control group, PdMb group showed an elevated proportion of GZMB+T and IFN-γ+T cells within CD8+T cells, and secretion of IFN-γ, TNF-α, perforin, and GZMB in CD8+T cell supernatant increased. Moreover, the proportion of CXCL9+F4/80+ macrophages in tumor tissues was higher in P. distasonis group compared to PBS group. In comparison to Control group, CXCL9 protein level in BMDM and CXCL9 secretion level in BMDM supernatant were increased in PdMb group. Finally, P. distasonis enhanced CD8+T cell activity by secreting CXCL9 from macrophages in vivo. CONCLUSIONS P. distasonis promoted CXCL9 secretion of TAM and enhanced CD8+T cell activity to trigger anti-tumor immunity against anti-PD-1 treatment in NSCLC mice.
Collapse
Affiliation(s)
- Zhijun Fan
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Zheng Yi
- Department of Cardiothoracic Surgery, The People's Hospital of Liuyang, Changsha, China
| | - Sheng Li
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China
| | - Junjun He
- Department of Gastrointestinal Surgery, The Central Hospital of Shaoyang, Shaoyang, China.
| |
Collapse
|
7
|
Wang J, Liang Y, Meng Y, Chen J, Fang L, Yang H, Li P. Assessment of lncRNA biomarkers based on NETs for prognosis and therapeutic response in ovarian cancer. Sci Rep 2025; 15:13042. [PMID: 40234525 PMCID: PMC12000398 DOI: 10.1038/s41598-025-97548-5] [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: 11/09/2024] [Accepted: 04/04/2025] [Indexed: 04/17/2025] Open
Abstract
Ovarian cancer (OC) usually progresses rapidly and is associated with high mortality, while a reliable clinical factor for OC patients to predict prognosis is currently lacking. Recently, the pathogenic role of neutrophils releasing neutrophil extracellular traps (NETs) in various cancers including OC has gradually been recognized. The study objective was to determine whether NETs-related biomarkers can be used to accurately predict the prognosis and guide clinical decision-making in OC. In this study, we utilized univariate and multivariate Cox regression to identify key prognostic features and developed a model with six NETs-related lncRNAs, selected via LASSO regression. The model's predictive capability was assessed through Kaplan-Meier, ROC, and Cox analyses. To understand the model's mechanisms, we conducted GO term analysis, KEGG pathway enrichment, and GSEA. We also analyzed gene mutation status, tumor mutation load, survival rates, and model correlation. Additionally, we compared immune functions, immune checkpoint expression, and chemotherapy sensitivity between risk groups. Besides, we validated the model's predictive value using test data and tissues acquired from our institution. Finally, we performed in vitro and in vivo experiments to confirm the expression of model lncRNAs and the cellular level function of GAS5. We developed a model using six NETs-associated lncRNAs: GAS5, GBP1P1, LINC00702, LINC01933, LINC02362, and ZNF687-AS1. The model's predictive performance, evaluated via ROC curve, was compared with traditional clinicopathological features. GO process analysis highlighted molecular functions related to antigen binding and immune system biological processes. Variations were observed in transcription regulators affecting immune response, inflammation, cytotoxicity, and regulation. We also predicted IC50 values for chemotherapeutic drugs (bexarotene, bicalutamide, embelin, GDC0941, and thapsigargin) in high- and low-risk groups, finding higher IC50 values in low-risk patients. The risk model's robustness was validated using OC cells, tissues, and clinical datas.
Collapse
Affiliation(s)
- Jingmeng Wang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Yusen Liang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Yimei Meng
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Jialin Chen
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China
| | - Lei Fang
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huike Yang
- Laboratory of Department of Anatomy, Harbin Medical University, Harbin, China.
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Harbin Medical University, Ministry of Education, Harbin, China.
| | - Peiling Li
- Department of Gynecology and Obstetrics, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
8
|
Rengarajan M, Normand R, Tran H, Nieman LT, Arnold B, Calcaterra M, Xu KH, Richieri P, Rodriguez EE, Slowikowski K, Song Y, Tirard A, Stephen AE, Sadow PM, Parangi S, Daniels GH, Luster AD, Villani AC. Immune-parenchymal multicellular niches are shared across distinct thyroid autoimmune diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.30.646176. [PMID: 40236152 PMCID: PMC11996388 DOI: 10.1101/2025.03.30.646176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Thyroid hormone, produced in the thyroid gland, regulates metabolism, development, and cardiac function. The thyroid is susceptible to autoimmune attack by both cellular and humoral immunity exemplified by Hashimoto's thyroiditis (HT) and Graves' Disease (GD), respectively. In HT, immune-mediated destruction impairs thyroid hormone production, while in GD, stimulating autoantibodies promote over-production. Here, we generated a multi-modal atlas of 604,076 human thyroid and blood cells from HT, GD, and control patients. We found that, despite markedly different clinical presentations and distinct antigenic triggers, HT and GD exhibit convergent cellular dynamics resulting in a shared continuum of immune infiltration. Along this continuum, a key feature is a thyrocyte niche containing CD8 + T cells that may segregate pathogenic T cells from regions with preserved thyroid hormone production. These findings of a shared disease continuum characterized by spatially defined immune niches provide a new framework for understanding tissue homeostasis in human autoimmune disease.
Collapse
|
9
|
Wen N, Lu Y, Zhuo Y, Fu B, Wang H, He Y, Wu H, Wang Z, Tan W, Qiu L. Enhancing T-Cell Infiltration and Immunity in Solid Tumors via DNA Nanolinker-Mediated Monocyte Hitchhiking. J Am Chem Soc 2025; 147:9800-9809. [PMID: 40042588 DOI: 10.1021/jacs.4c18455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cytotoxic CD8+ T cells are one of the most powerful effectors in the antitumor immune response. However, their insufficient tumor infiltration severely limits the clinical success of immunotherapy in solid tumors. In this work, by using amphiphilic aptamer-incorporated DNA tetrahedra (aptTDN) as the intercellular nanolinker, we developed a monocyte-hitchhiked T-cell delivery strategy to actively promote the intratumoral infiltration of effector CD8+ T cells. Our results demonstrated that membrane-anchoring of aptTDN enabled the specific and stable ligation of T cells with Ly6c+ monocytes, without compromising the migratory behavior of monocytes and the antitumor activity of T cells. By leveraging the intrinsic tumor-homing capability of monocytes, the ligated T cells efficiently accumulated within tumor-associated vasculature and then deeply infiltrated the tumor bed. Additionally, the enhanced intratumoral presence of adoptively transferred effector CD8+ T cells facilitated the establishment of an immunosupportive microenvironment, that further recruited endogenous T cells and ultimately bolstered antitumor immunity. Moreover, our monocyte-hitchhiked T-cell tumor infiltration system could significantly improve the efficacy of immune checkpoint blockade therapy. Collectively, by utilizing chemically synthetic nanolinkers to modulate cellular interactions and develop a delivery system of therapeutic cells, our work presents a new paradigm for the advancement of immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Nachuan Wen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yao Lu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuting Zhuo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Bo Fu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yao He
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhimin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
10
|
Weidle UH, Birzele F. Prostate Cancer: De-regulated Circular RNAs With Efficacy in Preclinical In Vivo Models. Cancer Genomics Proteomics 2025; 22:136-165. [PMID: 39993805 PMCID: PMC11880926 DOI: 10.21873/cgp.20494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 11/28/2025] [Accepted: 12/03/2024] [Indexed: 02/26/2025] Open
Abstract
Therapy resistance, including castration-resistance and metastasis, remains a major hurdle in the treatment of prostate cancer. In order to identify novel therapeutic targets and treatment modalities for prostate cancer, we conducted a comprehensive literature search on PubMed to identify de-regulated circular RNAs that influence treatment efficacy in preclinical prostate cancer-related in vivo models. Our analysis identified 49 circular RNAs associated with various processes, including treatment resistance, transmembrane and secreted proteins, transcription factors, signaling cascades, human antigen R, nuclear receptor binding, ubiquitination, metabolism, epigenetics and other target categories. The identified targets and circular RNAs can be further scrutinized through target validation approaches. Down-regulated circular RNAs are candidates for reconstitution therapy, while up-regulated RNAs can be inhibited using small interfering RNA (siRNA), antisense oligonucleotides (ASO) or clustered regularly interspaced short palindromic repeats/CRISPR associated (CRISPR-CAS)-related approaches.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
11
|
Ye J, Qin SS, Hughson AL, Hannon G, Vrooman TG, Lesch ML, Eckl SL, Benoodt L, Mills BN, Lord EM, Belt BA, Linehan DC, Luheshi N, Eyles J, Gerber SA. Blockade of LIF and PD-L1 Enhances Chemotherapy in Preclinical PDAC Models. Cancers (Basel) 2025; 17:204. [PMID: 39857986 PMCID: PMC11763610 DOI: 10.3390/cancers17020204] [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: 11/20/2024] [Revised: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Pancreatic ductal adenocarcinoma (PDAC), expecting to be the second leading cause of cancer deaths by 2030, resists immune checkpoint therapies due to its immunosuppressive tumor microenvironment (TME). Leukemia inhibitory factor (LIF) is a key target in PDAC, promoting stemness, epithelial-mesenchymal transition (EMT), and therapy resistance. Phase 1 clinical trials showed anti-LIF therapy is safe but with limited efficacy, suggesting better outcomes when combined with chemotherapy, radiotherapy, or immunotherapy. Methods: We assessed the combination of chemotherapy (gemcitabine/nab-paclitaxel) and dual blockade of LIF and PD-L1 on tumor growth and survival in orthotopic and spontaneous PDAC models. Flow cytometry and scRNA-seq were utilized to monitor the antitumor immune response. The role of key immune cells was further confirmed by depleting these immune cells, including CD4, CD8, or inflammatory monocytes. Results: Sequential treatment with chemotherapy (gemcitabine/nab-paclitaxel) and dual blockade of LIF and PD-L1 significantly improved antitumor efficacy compared to monotherapy or dual combinations of these therapies. This chemo/anti-LIF/anti-PD-L1 approach reduced EMT in tumor cells and enhanced the antitumor immune response, primarily through CD8 T cells, as depleting CD8 cells largely abrogated the effect of treatment. This combination therapy also shifted macrophages and dendritic cells towards an antitumor phenotype. Conclusions: The combination of chemotherapy, anti-LIF, and anti-PD-L1 not only targeted tumor cells but also augmented the anti-tumor immune response. These findings strongly support advancing chemo/anti-LIF/anti-PD-L1 combination therapy to clinical trials in PDAC.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Shuyang S. Qin
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Angela L. Hughson
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Tara G. Vrooman
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sarah L. Eckl
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lauren Benoodt
- Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Bradley N. Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Edith M. Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brian A. Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - David C. Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
| | - Nadia Luheshi
- Oncology R&D, AstraZeneca, Aaron Klug Building, Granta Park, Cambridge CB2 0AA, UK; (N.L.); (J.E.)
| | - Jim Eyles
- Oncology R&D, AstraZeneca, Aaron Klug Building, Granta Park, Cambridge CB2 0AA, UK; (N.L.); (J.E.)
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (S.S.Q.); (A.L.H.); (G.H.); (T.G.V.); (M.L.L.); (S.L.E.); (B.N.M.); (B.A.B.); (D.C.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14627, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14626, USA
| |
Collapse
|
12
|
Yao J, Cui Z, Zhang F, Li H, Tian L. Biomaterials enhancing localized cancer therapy activated anti-tumor immunity: a review. J Mater Chem B 2024; 13:117-136. [PMID: 39544081 DOI: 10.1039/d4tb01995d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Localized cancer therapies such as radiotherapy, phototherapy, and chemotherapy are precise cancer treatment strategies aimed at minimizing systemic side effects. However, cancer metastasis remains the primary cause of mortality among cancer patients in clinical settings, and localized cancer treatments have limited efficacy against metastatic cancer. Therefore, researchers are exploring strategies that combine localized therapy with immunotherapy to activate robust anti-tumor immune responses, thereby eradicating metastatic cancer. Biomaterials, as novel materials, exhibit great potential in biomedical applications and have achieved great progress in clinic translation. This review introduces biomaterials and their applications in research focused on enhancing localized cancer treatment activated anti-tumor immunity. Additionally, the current challenges and future directions of biomaterials are also discussed, providing insights and references for related research.
Collapse
Affiliation(s)
- Jipeng Yao
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Zhencun Cui
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Feifei Zhang
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Haidong Li
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Longlong Tian
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| |
Collapse
|
13
|
Cui YH, Wei J, Fan H, Li W, Zhao L, Wilkinson E, Peterson J, Xie L, Zou Z, Yang S, Applebaum MA, Kline J, Chen J, He C, He YY. Targeting DTX2/UFD1-mediated FTO degradation to regulate antitumor immunity. Proc Natl Acad Sci U S A 2024; 121:e2407910121. [PMID: 39661064 DOI: 10.1073/pnas.2407910121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Here, we show that vitamin E succinate (VES) acts as a degrader for the m6A RNA demethylase fat mass and obesity-associated protein (FTO), thus suppressing tumor growth and resistance to immunotherapy. FTO is ubiquitinated by its E3 ligase DTX2, followed by UFD1 recruitment and subsequent degradation in the proteasome. VES binds to FTO and DTX2, leading to enhanced FTO-DTX2 interaction, FTO ubiquitination, and degradation in FTO-dependent tumor cells. VES suppressed tumor growth and enhanced antitumor immunity and response to immunotherapy in vivo in mouse models. Genetic FTO knockdown or VES treatment increased m6A methylation in the LIF (Leukemia Inhibitory Factor) gene and decreased LIF mRNA decay, and thus sensitized melanoma cells to T cell-mediated cytotoxicity. Taken together, our findings reveal the underlying molecular mechanism for FTO protein degradation and identify a dietary degrader for FTO that inhibits tumor growth and overcomes immunotherapy resistance.
Collapse
Affiliation(s)
- Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Jiangbo Wei
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Hao Fan
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Wenlong Li
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Lijie Zhao
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637
| | - Jack Peterson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
- The College, University of Chicago, Chicago, IL 60637
| | - Lishi Xie
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Zhongyu Zou
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| | - Mark A Applebaum
- Department of Pediatrics, Section of Hematology/Oncology, University of Chicago, Chicago, IL 60637
| | - Justin Kline
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Jing Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637
| | - Chuan He
- Departments of Chemistry, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637
- Department of Biochemistry and Molecular Biology Institute for Biophysical Dynamics University of Chicago, Chicago, IL 60637
- HHMI, University of Chicago, Chicago, IL 60637
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
14
|
Grigore IA, Rajagopal A, Chow JTS, Stone TJ, Salmena L. Discovery of miRNA-mRNA regulatory networks in glioblastoma reveals novel insights into tumor microenvironment remodeling. Sci Rep 2024; 14:27493. [PMID: 39528571 PMCID: PMC11555236 DOI: 10.1038/s41598-024-78337-y] [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: 06/17/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Adult glioblastoma (GBM) is a highly aggressive primary brain tumor, accounting for nearly half of all malignant brain tumors, with a median survival rate of only 8 months. Treatment for GBM is largely ineffective due to the highly invasive nature and complex tumor composition of this malignancy. MicroRNAs (miRNA) are short, non-coding RNAs that regulate gene expression by binding to messenger RNAs (mRNA). While specific miRNA have been associated with GBM, their precise roles in tumor development and progression remain unclear. In this study, the analysis of miRNA expression data from 743 adult GBM cases and 59 normal brain samples identified 94 downregulated miRNA and 115 upregulated miRNA. Many of these miRNA were previously linked to GBM pathology, confirming the robustness of our approach, while we also identified novel miRNA that may act as potential regulators in GBM. By integrating miRNA predictions with gene expression data, we were able to associate downregulated miRNA with tumor microenvironment factors, including extracellular matrix remodeling and signaling pathways involved in tumor initiation, while upregulated miRNA were found to be associated with essential neuronal processes. This analysis highlights the significance of miRNA in GBM and serves as a foundation for further investigation.
Collapse
Affiliation(s)
- Iulia A Grigore
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Athulram Rajagopal
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Jonathan Tak-Sum Chow
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Thomas J Stone
- Developmental Biology and Cancer Research and Teaching Department, UCL GOS Institute of Child Health, University College London, London, UK
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Kabiljo J, Theophil A, Homola J, Renner AF, Stürzenbecher N, Ammon D, Zirnbauer R, Stang S, Tran L, Laengle J, Kulu A, Chen A, Fabits M, Atanasova VS, Pusch O, Weninger W, Walczak H, Herndler Brandstetter D, Egger G, Dolznig H, Kusienicka A, Farlik M, Bergmann M. Cancer-associated fibroblasts shape early myeloid cell response to chemotherapy-induced immunogenic signals in next generation tumor organoid cultures. J Immunother Cancer 2024; 12:e009494. [PMID: 39500527 PMCID: PMC11535717 DOI: 10.1136/jitc-2024-009494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Patient-derived colorectal cancer (CRC) organoids (PDOs) solely consisting of malignant cells led to major advances in the understanding of cancer treatments. Yet, a major limitation is the absence of cells from the tumor microenvironment, thereby prohibiting potential investigation of treatment responses on immune and structural cells. Currently there are sparse reports describing the interaction of PDOs, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs) in complex primary co-culture assay systems. METHODS Primary PDOs and patient matched CAF cultures were generated from surgical resections. Co-culture systems of PDOs, CAFs and monocytic myeloid cells were set up to recapitulate features seen in patient tumors. Single-cell transcriptomics and flow cytometry was used to show effects of culture systems on TAM populations in the co-culture assays under chemotherapeutic and oncolytic viral treatment. RESULTS In contrast to co-cultures of tumor cells and monocytes, CAF/monocyte co-cultures and CAF/monocyte/tumor cell triple cultures resulted in a partial differentiation into macrophages and a phenotypic switch, characterized by the expression of major immunosuppressive markers comparable to TAMs in CRC. Oxaliplatin and 5-fluorouracil, the standard-of-care chemotherapy for CRC, induced polarization of macrophages to a pro-inflammatory phenotype comparable to the immunogenic effects of treatment with an oncolytic virus. Monitoring phagocytosis as a functional proxy to macrophage activation and subsequent onset of an immune response, revealed that chemotherapy-induced cell death, but not virus-mediated cell death, is necessary to induce phagocytosis of CRC cells. Moreover, CAFs enhanced the phagocytic activity in chemotherapy treated CRC triple cultures. CONCLUSIONS Primary CAF-containing triple cultures successfully model TAM-like phenotypes ex vivo and allow the assessment of their functional and phenotypic changes in response to treatments following a precision medicine approach.
Collapse
Affiliation(s)
- Julijan Kabiljo
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Theophil
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jakob Homola
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Annalena F Renner
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nathalie Stürzenbecher
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Daphni Ammon
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Rebecca Zirnbauer
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Simone Stang
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Johannes Laengle
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Askin Kulu
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Anna Chen
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Markus Fabits
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Velina S Atanasova
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Oliver Pusch
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Henning Walczak
- Institute for Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Dietmar Herndler Brandstetter
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Gerda Egger
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Helmut Dolznig
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Anna Kusienicka
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Michael Bergmann
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Pietrobono S, Bertolini M, De Vita V, Sabbadini F, Fazzini F, Frusteri C, Scarlato E, Mangiameli D, Quinzii A, Casalino S, Zecchetto C, Merz V, Melisi D. CCL3 predicts exceptional response to TGFβ inhibition in basal-like pancreatic cancer enriched in LIF-producing macrophages. NPJ Precis Oncol 2024; 8:246. [PMID: 39478186 PMCID: PMC11525688 DOI: 10.1038/s41698-024-00742-3] [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: 04/24/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
The TGFβ receptor inhibitor galunisertib showed promising efficacy in patients with pancreatic ductal adenocarcinoma (PDAC) in the phase 2 H9H-MC-JBAJ study. Identifying biomarkers for this treatment remains essential. Baseline plasma levels of chemokine CCL3 were integrated with clinical outcomes in PDAC patients treated with galunisertib plus gemcitabine (n = 104) or placebo plus gemcitabine (n = 52). High CCL3 was a poor prognostic factor in the placebo group (mOS 3.6 vs. 10.1 months; p < 0.01) but a positive predictor for galunisertib (mOS 9.2 vs. 3.6 months; p < 0.01). Mechanistically, tumor-derived CCL3 activates Tgfβ signaling in macrophages, inducing their M2 phenotype and Lif secretion, sustaining a mesenchymal/basal-like ecotype. TGFβ inhibition redirects macrophage polarization to M1, reducing Lif and shifting PDAC cells to a more epithelial/classical phenotype, improving gemcitabine sensitivity. This study supports exploring TGFβ-targeting agents in PDAC with a mesenchymal/basal-like ecotype driven by high CCL3 levels.
Collapse
Affiliation(s)
- Silvia Pietrobono
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Monica Bertolini
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Veronica De Vita
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Fabio Sabbadini
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Federica Fazzini
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Cristina Frusteri
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Enza Scarlato
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Domenico Mangiameli
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Alberto Quinzii
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Simona Casalino
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Camilla Zecchetto
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Valeria Merz
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy
| | - Davide Melisi
- Department of Medicine, Digestive Molecular Clinical Oncology Research Unit, University of Verona, Verona, Italy.
- Investigational Cancer Therapeutics Clinical Unit, Azienda Ospedaliera Universitaria Integrata, Verona, Italy.
| |
Collapse
|
17
|
Li J, Wang Y, Wang Z, Wei Y, Diao P, Wu Y, Wang D, Jiang H, Wang Y, Cheng J. Super-Enhancer Driven LIF/LIFR-STAT3-SOX2 Regulatory Feedback Loop Promotes Cancer Stemness in Head and Neck Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404476. [PMID: 39206755 PMCID: PMC11516160 DOI: 10.1002/advs.202404476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Super-enhancers (SEs) have been recognized as key epigenetic regulators underlying cancer stemness and malignant traits by aberrant transcriptional control and promising therapeutic targets against human cancers. However, the SE landscape and their roles during head and neck squamous cell carcinoma (HNSCC) development especially in cancer stem cells (CSCs) maintenance remain underexplored yet. Here, we identify leukemia inhibitory factor (LIF)-SE as a representative oncogenic SE to activate LIF transcription in HNSCC. LIF secreted from cancer cells and cancer-associated fibroblasts promotes cancer stemness by driving SOX2 transcription in an autocrine/paracrine manner, respectively. Mechanistically, enhancer elements E1, 2, 4 within LIF-SE recruit SOX2/SMAD3/BRD4/EP300 to facilitate LIF transcription; LIF activates downstream LIFR-STAT3 signaling to drive SOX2 transcription, thus forming a previously unknown regulatory feedback loop (LIF-SE-LIF/LIFR-STAT3-SOX2) to maintain LIF overexpression and CSCs stemness. Clinically, increased LIF abundance in clinical samples correlate with malignant clinicopathological features and patient prognosis; higher LIF concentrations in presurgical plasma dramatically diminish following cancer eradication. Therapeutically, pharmacological targeting LIF-SE-LIF/LIFR-STAT3 significantly impairs tumor growth and reduces CSC subpopulations in xenograft and PDX models. Our findings reveal a hitherto uncharacterized LIF-SE-mediated auto-regulatory loop in regulating HNSCC stemness and highlight LIF as a novel noninvasive biomarker and potential therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Jin Li
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yuhan Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Ziyu Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yuxiang Wei
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Pengfei Diao
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yaping Wu
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Dongmiao Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
| | - Hongbing Jiang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| | - Yanling Wang
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
| | - Jie Cheng
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Nanjing Medical UniversityJiangsu210029China
- Jiangsu Key Laboratory of Oral DiseaseNanjing Medical UniversityJiangsu210029China
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjing Medical UniversityJiangsu210029China
| |
Collapse
|
18
|
Shahraz A, Penney M, Candido J, Opoku‐Ansah G, Neubauer M, Eyles J, Ojo O, Liu N, Luheshi NM, Phipps A, Vishwanathan K. A mechanistic PK/PD model of AZD0171 (anti-LIF) to support Phase II dose selection. CPT Pharmacometrics Syst Pharmacol 2024; 13:1670-1681. [PMID: 39041713 PMCID: PMC11494920 DOI: 10.1002/psp4.13204] [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: 02/17/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/24/2024] Open
Abstract
AZD0171 (INN: Falbikitug) is being developed as a humanized monoclonal antibody (mAb), immunoglobulin G subclass 1 (IgG1), which binds specifically to the immunosuppressive human cytokine leukemia inhibitory factor (LIF) and inhibits downstream signaling by blocking recruitment of glycoprotein 130 (gp130) to the LIF receptor (LIFR) subunit (gp190) and the phosphorylation of signal transducer and activator of transcription 3 (STAT3) and is intended to treat adult participants with advanced solid tumors. LIF is a pleiotropic cytokine (and a member of the IL-6 family of cytokines) involved in many physiological and pathological processes and is highly expressed in a subset of solid tumors, including non-small cell lung cancer (NSCLC), colon, ovarian, prostate, and pancreatic cancer. The aim of this work was to develop a mechanistic PK/PD model to investigate the effect of AZD0171 on tumor LIF levels, predict the level of downstream signaling complex (LIF:LIFR:gp130) inhibition, and examine the dose-response relationship to support dose selection for a Phase II clinical study. Modeling results show that tumor LIF is inhibited in a dose-dependent manner with >90% inhibition for 95% of patients at the Phase II clinical dose of 1500 mg Q2W.
Collapse
Affiliation(s)
- Azar Shahraz
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaWalthamMassachusettsUSA
| | - Mark Penney
- Early Oncology DMPK, Oncology R&D, AstraZenecaCambridgeUK
| | | | | | | | - Jim Eyles
- Oncology R&D, AstraZenecaCambridgeUK
| | | | | | | | - Alex Phipps
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaCambridgeUK
| | - Karthick Vishwanathan
- Clinical Pharmacology & Quantitative PharmacologyBioPharmaceuticals R&D, AstraZenecaWalthamMassachusettsUSA
| |
Collapse
|
19
|
Giangreco G, Rullan A, Naito Y, Biswas D, Liu YH, Hooper S, Nenclares P, Bhide S, Chon U Cheang M, Chakravarty P, Hirata E, Swanton C, Melcher A, Harrington K, Sahai E. Cancer cell - Fibroblast crosstalk via HB-EGF, EGFR, and MAPK signaling promotes the expression of macrophage chemo-attractants in squamous cell carcinoma. iScience 2024; 27:110635. [PMID: 39262776 PMCID: PMC11387794 DOI: 10.1016/j.isci.2024.110635] [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: 07/07/2023] [Revised: 04/09/2024] [Accepted: 07/30/2024] [Indexed: 09/13/2024] Open
Abstract
Interactions between cells in the tumor microenvironment (TME) shape cancer progression and patient prognosis. To gain insights into how the TME influences cancer outcomes, we derive gene expression signatures indicative of signaling between stromal fibroblasts and cancer cells, and demonstrate their prognostic significance in multiple and independent squamous cell carcinoma cohorts. By leveraging information within the signatures, we discover that the HB-EGF/EGFR/MAPK axis represents a hub of tumor-stroma crosstalk, promoting the expression of CSF2 and LIF and favoring the recruitment of macrophages. Together, these analyses demonstrate the utility of our approach for interrogating the extent and consequences of TME crosstalk.
Collapse
Affiliation(s)
- Giovanni Giangreco
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Antonio Rullan
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Yutaka Naito
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dhruva Biswas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, 72 Huntley Street, London WC1E 6DD, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Bill Lyons Informatics Centre, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Yun-Hsin Liu
- Bioinformatics Platform, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Steven Hooper
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Pablo Nenclares
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Shreerang Bhide
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Maggie Chon U Cheang
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Probir Chakravarty
- Bioinformatics Platform, Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eishu Hirata
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Bill Lyons Informatics Centre, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Alan Melcher
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Kevin Harrington
- Department of Radiotherapy and Imaging, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
- Head and Neck Unit, The Royal Marsden Hospital, 203 Fulham Road, London SW3 6JJ, UK
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
20
|
Do CTP, Prochnau JY, Dominguez A, Wang P, Rao MK. The Road Ahead in Pancreatic Cancer: Emerging Trends and Therapeutic Prospects. Biomedicines 2024; 12:1979. [PMID: 39335494 PMCID: PMC11428787 DOI: 10.3390/biomedicines12091979] [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: 08/01/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
This review explores the challenges and emerging trends in pancreatic cancer therapy. In particular, we focus on the tumor microenvironment and the potential of immunotherapy for pancreatic cancer. Pancreatic ductal adenocarcinoma, characterized by its dense stromal architecture, presents unique challenges for effective treatment. Recent advancements have emphasized the role of the tumor microenvironment in therapeutic resistance and disease progression. We discuss novel strategies targeting the desmoplastic barrier and immunosuppressive cells to enhance immune cell infiltration and activation. Recent clinical trials, particularly those involving novel immunotherapeutic agents and tumor vaccines, are examined to understand their efficacy and limitations. Our analysis reveals that combining immunotherapy with chemotherapy, radiation therapy, or drugs targeting epigenetic processes shows promise, improving overall survival rates and response to treatment. For instance, trials utilizing checkpoint inhibitors in combination with standard chemotherapies have extended disease-free survival by up to 6 months compared to chemotherapy alone. Importantly, vaccines targeting specific tumor neoantigens have shown the potential to increase patient survival. However, these approaches also face significant challenges, including overcoming the immunosuppressive tumor microenvironment and enhancing the delivery and efficacy of therapeutic agents. By providing an overview of both the promising results and the obstacles encountered, this review aims to highlight ongoing efforts to refine immunotherapy approaches for better patient outcomes.
Collapse
Affiliation(s)
- Chris T P Do
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jack Y Prochnau
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Angel Dominguez
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
21
|
Seifati SM, Zare F, Bafghi SAM, Hadinedoushan H. Impact of anti leukemia inhibitory factor antibody on immune related gene expression in breast cancer Balb/c mouse model. Sci Rep 2024; 14:20403. [PMID: 39223212 PMCID: PMC11369080 DOI: 10.1038/s41598-024-71014-0] [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: 05/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Leukemia inhibitory factor (LIF) is involved in the progression of different cancers. In this study, we investigated the effect of anti-LIF antibodies on immune-related gene expression in the Balb/c mouse model of breast cancer. To immunize mice against LIF, recombinant LIF with Freund adjuvant was injected into the test group, whereas the control group received phosphate-buffered saline with adjuvant. Tumor induction (4T1 cell line) was performed by increasing the antibody titer. The expression of immune-related genes was evaluated by real-time PCR. The anti-LIF titer was significantly increased in the immunized group. The expression of genes related to the differentiation of T helper (Th)-1, Th-2, and Th-17 cells was significantly higher in the immunized group than in the control group. In addition, anti-LIF did not have a significant effect on the expression of genes related to the differentiation of regulatory T cells, and immune checkpoint-associated genes. Additionally, the test group had higher survival and lower tumor development rates. The results demonstrated that the anti-LIF antibody may potentially play a role in the differentiation of immune cells or immune responses. However, further studies utilizing advanced techniques are necessary to validate its function.
Collapse
Affiliation(s)
- Seyed Mohammad Seifati
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
| | | | - Hossein Hadinedoushan
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran.
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
22
|
Tsui KH, Liu CL, Yeh HL, Liu MK, Li CH, Chen WH, Jiang KC, Li HR, Thuy Dung PV, Hsiao M, Abou-Kheir W, Liu YN. WNT1-inducible signaling pathway protein 1 activation through C-X-C motif chemokine ligand 5/C-X-C chemokine receptor type 2/leukemia inhibitory factor/leukemia inhibitory factor receptor signaling promotes immunosuppression and neuroendocrine differentiation in prostate cancer. iScience 2024; 27:110562. [PMID: 39175775 PMCID: PMC11338985 DOI: 10.1016/j.isci.2024.110562] [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: 08/29/2023] [Revised: 02/16/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
The interaction between prostate cancer (PCa) cells and prostate stromal cells fosters an immunosuppressive tumor microenvironment (TME) that promotes tumor growth and immune evasion. However, the specific signaling pathways involved remain unclear. We identified a key mechanism involving the CXCL5/CXCR2 and LIF/LIFR pathways, which create a feedforward loop that enhances neuroendocrine differentiation (NED) in PCa cells and upregulates WNT1-inducible signaling pathway protein 1 (WISP1) in both cell types. WISP1 upregulation is essential for inducing immune checkpoints and immunosuppressive cytokines via LIF/LIFR signaling and STAT3 phosphorylation. This process leads to increased neuroendocrine markers, immune checkpoints, cell proliferation, and migration. Notably, WISP1 levels in patient sera correlate with PCa progression, suggesting its potential as a biomarker. Our findings elucidate the mechanisms by which reciprocal communication between PCa cells and stromal cells contributes to the formation of an immunosuppressive TME, driving the malignant progression of PCa and highlighting potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Ke Hung Tsui
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan
- Department of Urology, School of Medicine, College of Medical, Taipei Medical University, Taipei 110, Taiwan
| | - Chien-Liang Liu
- Department of Surgery, Division of Urology, Chi Mei Medical Center, Tainan 710, Taiwan
| | - Hsiu-Lien Yeh
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Kun Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Chien-Hsiu Li
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, Taipei 110, Taiwan
| | - Wei-Hao Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Kuo-Ching Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Han-Ru Li
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Phan Vu Thuy Dung
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Yen-Nien Liu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
23
|
Majid U, Bergsland CH, Sveen A, Bruun J, Eilertsen IA, Bækkevold ES, Nesbakken A, Yaqub S, Jahnsen FL, Lothe RA. The prognostic effect of tumor-associated macrophages in stage I-III colorectal cancer depends on T cell infiltration. Cell Oncol (Dordr) 2024; 47:1267-1276. [PMID: 38407700 PMCID: PMC11322253 DOI: 10.1007/s13402-024-00926-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are associated with unfavorable patient prognosis in many cancer types. However, TAMs are a heterogeneous cell population and subsets have been shown to activate tumor-infiltrating T cells and confer a good patient prognosis. Data on the prognostic value of TAMs in colorectal cancer are conflicting. We investigated the prognostic effect of TAMs in relation to tumor-infiltrating T cells in colorectal cancers. METHODS The TAM markers CD68 and CD163 were analyzed by multiplex fluorescence immunohistochemistry and digital image analysis on tissue microarrays of 1720 primary colorectal cancers. TAM density in the tumor stroma was scored in relation to T cell density (stromal CD3+ and epithelial CD8+ cells) and analyzed in Cox proportional hazards models of 5-year relapse-free survival. Multivariable survival models included clinicopathological factors, MSI status and BRAFV600E mutation status. RESULTS High TAM density was associated with a favorable 5-year relapse-free survival in a multivariable model of patients with stage I-III tumors (p = 0.004, hazard ratio 0.94, 95% confidence interval 0.90-0.98). However, the prognostic effect was dependent on tumoral T-cell density. High TAM density was associated with a good prognosis in patients who also had high T-cell levels in their tumors, while high TAM density was associated with poorer prognosis in patients with low T-cell levels (pinteraction = 0.0006). This prognostic heterogeneity was found for microsatellite stable tumors separately. CONCLUSIONS This study supported a phenotypic heterogeneity of TAMs in colorectal cancer, and showed that combined tumor immunophenotyping of multiple immune cell types improved the prediction of patient prognosis.
Collapse
Affiliation(s)
- Umair Majid
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Christian Holst Bergsland
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jarle Bruun
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ina Andrassy Eilertsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Espen S Bækkevold
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
- Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Arild Nesbakken
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sheraz Yaqub
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hepatobiliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Frode L Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
24
|
von Roemeling CA, Patel JA, Carpenter SL, Yegorov O, Yang C, Bhatia A, Doonan BP, Russell R, Trivedi VS, Klippel K, Ryu DH, Grippin A, Futch HS, Ran Y, Hoang-Minh LB, Weidert FL, Golde TE, Mitchell DA. Adeno-associated virus delivered CXCL9 sensitizes glioblastoma to anti-PD-1 immune checkpoint blockade. Nat Commun 2024; 15:5871. [PMID: 38997283 PMCID: PMC11245621 DOI: 10.1038/s41467-024-49989-1] [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: 11/07/2023] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability.
Collapse
Affiliation(s)
- Christina A von Roemeling
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA.
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| | - Jeet A Patel
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Savannah L Carpenter
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Oleg Yegorov
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Changlin Yang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Alisha Bhatia
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Bently P Doonan
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
- Department of Medicine, Hematology and Oncology, University of Florida, Gainesville, FL, USA
| | - Rylynn Russell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Vrunda S Trivedi
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Kelena Klippel
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Daniel H Ryu
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Adam Grippin
- Department of Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Hunter S Futch
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong Ran
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Lan B Hoang-Minh
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Frances L Weidert
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Todd E Golde
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA.
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
25
|
Houlder EL, Stam KA, Koopman JPR, König MH, Langenberg MCC, Hoogerwerf MA, Niewold P, Sonnet F, Janse JJ, Partal MC, Sijtsma JC, de Bes-Roeleveld LHM, Kruize YCM, Yazdanbakhsh M, Roestenberg M. Early symptom-associated inflammatory responses shift to type 2 responses in controlled human schistosome infection. Sci Immunol 2024; 9:eadl1965. [PMID: 38968336 DOI: 10.1126/sciimmunol.adl1965] [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: 10/13/2023] [Accepted: 06/07/2024] [Indexed: 07/07/2024]
Abstract
Schistosomiasis is an infection caused by contact with Schistosoma-contaminated water and affects more than 230 million people worldwide with varying morbidity. The roles of T helper 2 (TH2) cells and regulatory immune responses in chronic infection are well documented, but less is known about human immune responses during acute infection. Here, we comprehensively map immune responses during controlled human Schistosoma mansoni infection using male or female cercariae. Immune responses to male or female parasite single-sex infection were comparable. An early TH1-biased inflammatory response was observed at week 4 after infection, which was particularly apparent in individuals experiencing symptoms of acute schistosomiasis. By week 8 after infection, inflammatory responses were followed by an expansion of TH2 and regulatory cell subsets. This study demonstrates the shift from TH1 to both TH2 and regulatory responses, typical of chronic schistosomiasis, in the absence of egg production and provides immunological insight into the clinical manifestations of acute schistosomiasis.
Collapse
Affiliation(s)
- Emma L Houlder
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Koen A Stam
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jan Pieter R Koopman
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marion H König
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marijke C C Langenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Marie-Astrid Hoogerwerf
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Paula Niewold
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Friederike Sonnet
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jacqueline J Janse
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Miriam Casacuberta Partal
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Jeroen C Sijtsma
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Laura H M de Bes-Roeleveld
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Yvonne C M Kruize
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Maria Yazdanbakhsh
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333ZA Leiden, Netherlands
| |
Collapse
|
26
|
Fiorucci S, Marchianò S, Urbani G, Di Giorgio C, Distrutti E, Zampella A, Biagioli M. Immunology of bile acids regulated receptors. Prog Lipid Res 2024; 95:101291. [PMID: 39122016 DOI: 10.1016/j.plipres.2024.101291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Bile acids are steroids formed at the interface of host metabolism and intestinal microbiota. While primary bile acids are generated in the liver from cholesterol metabolism, secondary bile acids represent the products of microbial enzymes. Close to 100 different enzymatic modifications of bile acids structures occur in the human intestine and clinically guided metagenomic and metabolomic analyses have led to the identification of an extraordinary number of novel metabolites. These chemical mediators make an essential contribution to the composition and function of the postbiota, participating to the bidirectional communications of the intestinal microbiota with the host and contributing to the architecture of intestinal-liver and -brain and -endocrine axes. Bile acids exert their function by binding to a group of cell membrane and nuclear receptors collectively known as bile acid-regulated receptors (BARRs), expressed in monocytes, tissue-resident macrophages, CD4+ T effector cells, including Th17, T regulatory cells, dendritic cells and type 3 of intestinal lymphoid cells and NKT cells, highlighting their role in immune regulation. In this review we report on how bile acids and their metabolitesmodulate the immune system in inflammations and cancers and could be exploiting for developing novel therapeutic approaches in these disorders.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Silvia Marchianò
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | | | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
27
|
Lu B, Shi J, Cheng T, Wang C, Xu M, Sun P, Zhang X, Yang L, Li P, Wu H, Kuai X. Chemokine ligand 14 correlates with immune cell infiltration in the gastric cancer microenvironment in predicting unfavorable prognosis. Front Pharmacol 2024; 15:1397656. [PMID: 38887558 PMCID: PMC11180770 DOI: 10.3389/fphar.2024.1397656] [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: 03/08/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Gastric cancer (GC) is the world's third-leading cause of cancer-related mortality; the prognosis for GC patients remains poor in terms of a lack of reliable biomarkers for early diagnosis and immune therapy response prediction. Here, we aim to discover the connection between chemokine ligand 14 (CCL14) expression in the gastric tumor microenvironment (TME) and its clinical significance and investigate its correlation with immune cell infiltration. METHODS We assessed CCL14 mRNA expression and its interrelation with tumor-infiltrating immune cells (TILs) using bioinformatics analysis in gastric cancer. CCL14 protein expression, TILs, and immune checkpoints were detected by multiple immunohistochemistry analyses in gastric cancer tissue microarrays. Then, we conducted statistics analysis to determine the association between CCL14-related patient survival and immune cell infiltration (p < 0.05). RESULTS We found that the CCL14 protein was separately expressed in the carcinoma cells and TILs in stomach cancer tissues. The CCL14 protein was related to tumor differentiation and tumor depth and positively correlated with the presentation of LAG3 and PD-L1 in gastric cancer cells. In addition, the CCL14 protein in the TILs of gastric cancer tissues was related to Lauren's type cells, T cells (CD4+ and CD8+), and CD68+ macrophages in the TME. Kaplan-Meier survival and multivariate analyses showed that the CCL14 expression in gastric cancer cells was an independent prognostic factor. CONCLUSION Our study illustrated that CCL14 is a poor prognosis biomarker in gastric cancer, which may be associated with the potential for immunotherapy.
Collapse
Affiliation(s)
- Bing Lu
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
| | - Jiawen Shi
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
- Department of Pathology and Pathophysiology, School of Medicine, Nantong University, Nantong, China
| | - Tong Cheng
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
- Department of Oncology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Congshuo Wang
- Department of Gastroenterology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Manyu Xu
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
- Department of Oncology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Pingping Sun
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
| | - Xiaojing Zhang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
| | - Lei Yang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, China
| | - Peng Li
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Han Wu
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Xiaoling Kuai
- Department of Gastroenterology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| |
Collapse
|
28
|
Ebrahimi B, Viswanadhapalli S, Pratap UP, Rahul G, Yang X, Pitta Venkata P, Drel V, Santhamma B, Konda S, Li X, Sanchez ALR, Yan H, Sareddy GR, Xu Z, Singh BB, Valente PT, Chen Y, Lai Z, Rao M, Kost ER, Curiel T, Tekmal RR, Nair HB, Vadlamudi RK. Pharmacological inhibition of the LIF/LIFR autocrine loop reveals vulnerability of ovarian cancer cells to ferroptosis. NPJ Precis Oncol 2024; 8:118. [PMID: 38789520 PMCID: PMC11126619 DOI: 10.1038/s41698-024-00612-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Of all gynecologic cancers, epithelial-ovarian cancer (OCa) stands out with the highest mortality rates. Despite all efforts, 90% of individuals who receive standard surgical and cytotoxic therapy experience disease recurrence. The precise mechanism by which leukemia inhibitory factor (LIF) and its receptor (LIFR) contribute to the progression of OCa remains unknown. Analysis of cancer databases revealed that elevated expression of LIF or LIFR was associated with poor progression-free survival of OCa patients and a predictor of poor response to chemotherapy. Using multiple primary and established OCa cell lines or tissues that represent five subtypes of epithelial-OCa, we demonstrated that LIF/LIFR autocrine signaling is active in OCa. Moreover, treatment with LIFR inhibitor, EC359 significantly reduced OCa cell viability and cell survival with an IC50 ranging from 5-50 nM. Furthermore, EC359 diminished the stemness of OCa cells. Mechanistic studies using RNA-seq and rescue experiments unveiled that EC359 primarily induced ferroptosis by suppressing the glutathione antioxidant defense system. Using multiple in vitro, ex vivo and in vivo models including cell-based xenografts, patient-derived explants, organoids, and xenograft tumors, we demonstrated that EC359 dramatically reduced the growth and progression of OCa. Additionally, EC359 therapy considerably improved tumor immunogenicity by robust CD45+ leukocyte tumor infiltration and polarizing tumor-associated macrophages (TAMs) toward M1 phenotype while showing no impact on normal T-, B-, and other immune cells. Collectively, our findings indicate that the LIF/LIFR autocrine loop plays an essential role in OCa progression and that EC359 could be a promising therapeutic agent for OCa.
Collapse
Affiliation(s)
- Behnam Ebrahimi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Gopalam Rahul
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Xue Yang
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, PR China
| | - Prabhakar Pitta Venkata
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Viktor Drel
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | | | | | - Xiaonan Li
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | | | - Hui Yan
- Department of microbiology and immunology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Zhenming Xu
- Department of microbiology and immunology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Brij B Singh
- Department of Periodontics, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Philip T Valente
- Department of Pathology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Yidong Chen
- Department of Population Sciences, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Manjeet Rao
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Edward R Kost
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Tyler Curiel
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth, NH, 03755, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | | | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
- Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
29
|
Tikhonova MA, Shoeva OY, Tenditnik MV, Akopyan AA, Litvinova EA, Popova NA, Amstislavskaya TG, Khlestkina EK. Antitumor Effects of an Anthocyanin-Rich Grain Diet in a Mouse Model of Lewis Lung Carcinoma. Int J Mol Sci 2024; 25:5727. [PMID: 38891915 PMCID: PMC11171629 DOI: 10.3390/ijms25115727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Functional foods enriched with plant polyphenol anthocyanins attract particular attention due to their health-promoting properties, including antitumor activity. We evaluated the effects of a grain diet rich in anthocyanins in a mouse model of Lewis lung carcinoma. Mice of the C57BL/6 strain were fed with wheat of near-isogenic lines differing in the anthocyanin content for four months prior to tumor transplantation. Although a significant decrease in the size of the tumor and the number of metastases in the lungs was revealed in the groups with both types of grain diet, the highest percentage of animals without metastases and with attenuated cell proliferation in the primary tumor were observed in the mice with the anthocyanin-rich diet. Both grain diets reduced the body weight gain and spleen weight index. The antitumor effects of the grain diets were associated with the activation of different mechanisms: immune response of the allergic type with augmented interleukin(IL)-9 and eotaxin serum levels in mice fed with control grain vs. inhibition of the IL-6/LIF system accompanied by a decrease in the tumor-associated M2 macrophage marker arginase 1 gene mRNA levels and enhanced autophagy in the tumor evaluated by the mRNA levels of Beclin 1 gene. Thus, anthocyanin-rich wheat is suggested as a promising source of functional nutrition with confirmed in vivo antitumor activity.
Collapse
Affiliation(s)
- Maria A. Tikhonova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia; (O.Y.S.); (T.G.A.)
- Scientific Research Institute of Neurosciences and Medicine (SRINM), 630117 Novosibirsk, Russia
| | - Olesya Y. Shoeva
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia; (O.Y.S.); (T.G.A.)
| | - Michael V. Tenditnik
- Scientific Research Institute of Neurosciences and Medicine (SRINM), 630117 Novosibirsk, Russia
| | - Anna A. Akopyan
- Scientific Research Institute of Neurosciences and Medicine (SRINM), 630117 Novosibirsk, Russia
| | - Ekaterina A. Litvinova
- Scientific Research Institute of Neurosciences and Medicine (SRINM), 630117 Novosibirsk, Russia
| | - Nelly A. Popova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia; (O.Y.S.); (T.G.A.)
- Department of Neuroscience, V. Zelman Institute for Medicine and Psychology, Faculty of Life Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Tamara G. Amstislavskaya
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia; (O.Y.S.); (T.G.A.)
- Scientific Research Institute of Neurosciences and Medicine (SRINM), 630117 Novosibirsk, Russia
- Department of Neuroscience, V. Zelman Institute for Medicine and Psychology, Faculty of Life Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Elena K. Khlestkina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (IC&G SB RAS), 630090 Novosibirsk, Russia; (O.Y.S.); (T.G.A.)
- N.I. Vavilov All-Russian Research Institute of Plant Genetic Resources, 190000 St. Petersburg, Russia
| |
Collapse
|
30
|
Di Giorgio C, Morretta E, Lupia A, Bellini R, Massa C, Urbani G, Bordoni M, Marchianò S, Lachi G, Rapacciuolo P, Finamore C, Sepe V, Chiara Monti M, Moraca F, Natalizi N, Graziosi L, Distrutti E, Biagioli M, Catalanotti B, Donini A, Zampella A, Fiorucci S. Bile acids serve as endogenous antagonists of the Leukemia inhibitory factor (LIF) receptor in oncogenesis. Biochem Pharmacol 2024; 223:116134. [PMID: 38494064 DOI: 10.1016/j.bcp.2024.116134] [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: 12/04/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The leukemia inhibitory factor (LIF) is member of interleukin (IL)-6 family of cytokines involved immune regulation, morphogenesis and oncogenesis. In cancer tissues, LIF binds a heterodimeric receptor (LIFR), formed by a LIFRβ subunit and glycoprotein(gp)130, promoting epithelial mesenchymal transition and cell growth. Bile acids are cholesterol metabolites generated at the interface of host metabolism and the intestinal microbiota. Here we demonstrated that bile acids serve as endogenous antagonist to LIFR in oncogenesis. The tissue characterization of bile acids content in non-cancer and cancer biopsy pairs from gastric adenocarcinomas (GC) demonstrated that bile acids accumulate within cancer tissues, with glyco-deoxycholic acid (GDCA) functioning as negative regulator of LIFR expression. In patient-derived organoids (hPDOs) from GC patients, GDCA reverses LIF-induced stemness and proliferation. In summary, we have identified the secondary bile acids as the first endogenous antagonist to LIFR supporting a development of bile acid-based therapies in LIF-mediated oncogenesis.
Collapse
Affiliation(s)
| | - Elva Morretta
- University of Salerno, Department of Pharmacy, Salerno, Italy
| | - Antonio Lupia
- University of Cagliari, Department of Life and Environmental Sciences, Cagliari, Italy; Net4Science srl, University "Magna Græcia", Campus Salvatore Venuta, Viale Europa, Catanzaro 88100, Italy
| | - Rachele Bellini
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Carmen Massa
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Ginevra Urbani
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Martina Bordoni
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Silvia Marchianò
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Ginevra Lachi
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | | | - Claudia Finamore
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Valentina Sepe
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | | | - Federica Moraca
- Net4Science srl, University "Magna Græcia", Campus Salvatore Venuta, Viale Europa, Catanzaro 88100, Italy; University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | | | | | | | - Michele Biagioli
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Bruno Catalanotti
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Annibale Donini
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy
| | - Angela Zampella
- University of Naples Federico II, Department of Pharmacy, Naples, Italy
| | - Stefano Fiorucci
- University of Perugia, Department of Medicine and Surgery, Perugia, Italy.
| |
Collapse
|
31
|
Zhang B, Shi J, Shi X, Xu X, Gao L, Li S, Liu M, Gao M, Jin S, Zhou J, Fan D, Wang F, Ji Z, Bian Z, Song Y, Tian W, Zheng Y, Xu L, Li W. Development and evaluation of a human CD47/HER2 bispecific antibody for Trastuzumab-resistant breast cancer immunotherapy. Drug Resist Updat 2024; 74:101068. [PMID: 38402670 DOI: 10.1016/j.drup.2024.101068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/28/2024] [Accepted: 02/10/2024] [Indexed: 02/27/2024]
Abstract
The treatment for trastuzumab-resistant breast cancer (BC) remains a challenge in clinical settings. It was known that CD47 is preferentially upregulated in HER2+ BC cells, which is correlated with drug resistance to trastuzumab. Here, we developed a novel anti-CD47/HER2 bispecific antibody (BsAb) against trastuzumab-resistant BC, named IMM2902. IMM2902 demonstrated high binding affinity, blocking activity, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), and internalization degradation effects against both trastuzumab-sensitive and trastuzumab-resistant BC cells in vitro. The in vivo experimental data indicated that IMM2902 was more effective than their respective controls in inhibiting tumor growth in a trastuzumab-sensitive BT474 mouse model, a trastuzumab-resistant HCC1954 mouse model, two trastuzumab-resistant patient-derived xenograft (PDX) mouse models and a cord blood (CB)-humanized HCC1954 mouse model. Through spatial transcriptome assays, multiplex immunofluorescence (mIFC) and in vitro assays, our findings provided evidence that IMM2902 effectively stimulates macrophages to generate C-X-C motif chemokine ligand (CXCL) 9 and CXCL10, thereby facilitating the recruitment of T cells and NK cells to the tumor site. Moreover, IMM2902 demonstrated a high safety profile regarding anemia and non-specific cytokines release. Collectively, our results highlighted a novel therapeutic approach for the treatment of HER2+ BCs and this approach exhibits significant anti-tumor efficacy without causing off-target toxicity in trastuzumab-resistant BC cells.
Collapse
Affiliation(s)
- Binglei Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jianxiang Shi
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaojing Shi
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaolu Xu
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Le Gao
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Song Li
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc, Shanghai 201203, China
| | - Mengmeng Liu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mengya Gao
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China; Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shuiling Jin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Zhou
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Dandan Fan
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhenyu Ji
- Henan Institute of Medical and Pharmaceutical Sciences, Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc, Shanghai 201203, China
| | - Yichao Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450008, China.
| | - Linping Xu
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
32
|
Lim RJ, Salehi-Rad R, Tran LM, Oh MS, Dumitras C, Crosson WP, Li R, Patel TS, Man S, Yean CE, Abascal J, Huang Z, Ong SL, Krysan K, Dubinett SM, Liu B. CXCL9/10-engineered dendritic cells promote T cell activation and enhance immune checkpoint blockade for lung cancer. Cell Rep Med 2024; 5:101479. [PMID: 38518770 PMCID: PMC11031384 DOI: 10.1016/j.xcrm.2024.101479] [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: 08/11/2023] [Revised: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 03/24/2024]
Abstract
Immune checkpoint blockade (ICB) with PD-1/PD-L1 inhibition has revolutionized the treatment of non-small cell lung cancer (NSCLC). Durable responses, however, are observed only in a subpopulation of patients. Defective antigen presentation and an immunosuppressive tumor microenvironment (TME) can lead to deficient T cell recruitment and ICB resistance. We evaluate intratumoral (IT) vaccination with CXCL9- and CXCL10-engineered dendritic cells (CXCL9/10-DC) as a strategy to overcome resistance. IT CXCL9/10-DC leads to enhanced T cell infiltration and activation in the TME and tumor inhibition in murine NSCLC models. The antitumor efficacy of IT CXCL9/10-DC is dependent on CD4+ and CD8+ T cells, as well as CXCR3-dependent T cell trafficking from the lymph node. IT CXCL9/10-DC, in combination with ICB, overcomes resistance and establishes systemic tumor-specific immunity in murine models. These studies provide a mechanistic understanding of CXCL9/10-DC-mediated host immune activation and support clinical translation of IT CXCL9/10-DC to augment ICB efficacy in NSCLC.
Collapse
Affiliation(s)
- Raymond J Lim
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ramin Salehi-Rad
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Linh M Tran
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Michael S Oh
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Camelia Dumitras
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - William P Crosson
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rui Li
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tejas S Patel
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha Man
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cara E Yean
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jensen Abascal
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - ZiLing Huang
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Stephanie L Ong
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kostyantyn Krysan
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Bin Liu
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
33
|
Enbergs N, Halabi EA, Goubet A, Schleyer K, Fredrich IR, Kohler RH, Garris CS, Pittet MJ, Weissleder R. Pharmacological Polarization of Tumor-Associated Macrophages Toward a CXCL9 Antitumor Phenotype. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309026. [PMID: 38342608 PMCID: PMC11022742 DOI: 10.1002/advs.202309026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/16/2024] [Indexed: 02/13/2024]
Abstract
Tumor-associated macrophages (TAM) are a diverse population of myeloid cells that are often abundant and immunosuppressive in human cancers. CXCL9Hi TAM has recently been described to have an antitumor phenotype and is linked to immune checkpoint response. Despite the emerging understanding of the unique antitumor TAM phenotype, there is a lack of TAM-specific therapeutics to exploit this new biological understanding. Here, the discovery and characterization of multiple small-molecule enhancers of chemokine ligand 9 (CXCL9) and their targeted delivery in a TAM-avid systemic nanoformulation is reported. With this strategy, it is efficient encapsulation and release of multiple drug loads that can efficiently induce CXCL9 expression in macrophages, both in vitro and in vivo in a mouse tumor model. These observations provide a window into the molecular features that define TAM-specific states, an insight a novel therapeutic anticancer approach is used to discover.
Collapse
Affiliation(s)
- Noah Enbergs
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Elias A. Halabi
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Anne‐Gaëlle Goubet
- Department of Pathology and ImmunologyUniversity of GenevaGeneva1211Switzerland
- AGORA Cancer CenterSwiss Cancer Center LemanLausanne1011Switzerland
| | - Kelton Schleyer
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Ina R. Fredrich
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Rainer H. Kohler
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Christopher S. Garris
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Mikaël J. Pittet
- Department of Pathology and ImmunologyUniversity of GenevaGeneva1211Switzerland
- AGORA Cancer CenterSwiss Cancer Center LemanLausanne1011Switzerland
- Ludwig Institute for Cancer ResearchLausanne1005Switzerland
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
| |
Collapse
|
34
|
Jia X, Li Z, Zhou R, Feng W, Yi L, Zhang H, Chen B, Li Q, Huang S, Zhu X. Single cell and bulk RNA sequencing identifies tumor microenvironment subtypes and chemoresistance-related IGF1 + cancer-associated fibroblast in gastric cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167123. [PMID: 38484940 DOI: 10.1016/j.bbadis.2024.167123] [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: 08/02/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The tumor microenvironment (TME) significantly influences prognosis and drug resistance in various tumors, yet its heterogeneity and the mechanisms affecting therapeutic response remain unclear in gastric cancer (GC). METHODS The heterogenous TME were explored with single-cell RNA-sequencing (scRNA-seq) data of 50 primary GC samples. We then identified four GC TME subtypes with nonnegative matrix factorization (NMF) and constructed a pearson nearest-centroid classifier based on subtype-specific upregulated genes. Genomic features and clinical significance of four subtypes were comprehensively evaluated. We reclustered fibroblasts to identify cancer-associated fibroblast (CAF) subtype associated with poor clinical outcomes. RT-qPCR and double immunofluorescence staining were applied to validate the findings. Cellchat analysis elucidated potential molecular mechanisms of the CAF subtype in GC disease progression and chemotherapy resistance. FINDINGS The GC TME exhibited high heterogeneity, influencing chemo-sensitivity. Four TME-based subtypes predicting response to immunotherapy and chemotherapy were identified and validated in 1406 GC patients. Among which, ISG1 subtype displayed higher fibroblasts infiltration and heightened oncogenic pathways, and inferior response to chemotherapy with unfavorable prognosis. Microsatellite instability-high (MSI-H) GCs within four TME subtypes showed immunological heterogeneity. We then reported an IGF1-overexpressing CAF was associated with chemo-resistance and GC recurrence. Cell communication analysis revealed IGF1+ CAF may induce drug-resistant phenotypes in tumor cells through IGF1-α6β4 integrin ligand-receptor binding and activation of EMT biological process. INTERPRETATION We identified four TME-based subtypes with different clinical outcomes and IGF1+ CAFs contributing to poor clinical outcomes in GC, which might provide guidance for individualized treatment and facilitate the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Xiya Jia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ziteng Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Runye Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wanjing Feng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lixia Yi
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hena Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Bing Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shenglin Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
35
|
Sweet-Cordero E, Marini K, Champion E, Lee A, Young I, Leung S, Mathey-Andrews N, Jacks T, Jackson P, Cochran J. The CLCF1-CNTFR axis drives an immunosuppressive tumor microenvironment and blockade enhances the effects of established cancer therapies. RESEARCH SQUARE 2024:rs.3.rs-4046823. [PMID: 38562778 PMCID: PMC10984090 DOI: 10.21203/rs.3.rs-4046823/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Tumors comprise a complex ecosystem consisting of many cell types that communicate through secreted factors. Targeting these intercellular signaling networks remains an important challenge in cancer research. Cardiotrophin-like cytokine factor 1 (CLCF1) is an interleukin-6 (IL-6) family member secreted by cancer-associated fibroblasts (CAFs) that binds to ciliary neurotrophic factor receptor (CNTFR), promoting tumor growth in lung and liver cancer1,2. A high-affinity soluble receptor (eCNTFR-Fc) that sequesters CLCF1 has anti-oncogenic effects3. However, the role of CLCF1 in mediating cell-cell interactions in cancer has remained unclear. We demonstrate that eCNTFR-Fc has widespread effects on both tumor cells and the tumor microenvironment and can sensitize cancer cells to KRAS inhibitors or immune checkpoint blockade. After three weeks of treatment with eCNTFR-Fc, there is a shift from an immunosuppressive to an immunostimulatory macrophage phenotype as well as an increase in activated T, NKT, and NK cells. Combination of eCNTFR-Fc and αPD1 was significantly more effective than single-agent therapy in a syngeneic allograft model, and eCNTFR-Fc sensitizes tumor cells to αPD1 in a non-responsive GEM model of lung adenocarcinoma. These data suggest that combining eCNTFR-Fc with KRAS inhibition or with αPD1 is a novel therapeutic strategy for lung cancer and potentially other cancers in which these therapies have been used but to date with only modest effect. Overall, we demonstrate the potential of cancer therapies that target cytokines to alter the immune microenvironment.
Collapse
Affiliation(s)
| | - Kieren Marini
- Division of Oncology, Department of Pediatrics, University of California San Francisco
| | - Emma Champion
- Division of Oncology, Department of Pediatrics, University of California San Francisco
| | - Alex Lee
- University of California, San Francisco
| | - Isabelle Young
- Division of Oncology, Department of Pediatrics, University of California San Francisco
| | - Stanley Leung
- Division of Oncology, Department of Pediatrics, University of California San Francisco
| | | | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research
| | | | | |
Collapse
|
36
|
Di Ceglie I, Carnevale S, Rigatelli A, Grieco G, Molisso P, Jaillon S. Immune cell networking in solid tumors: focus on macrophages and neutrophils. Front Immunol 2024; 15:1341390. [PMID: 38426089 PMCID: PMC10903099 DOI: 10.3389/fimmu.2024.1341390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The tumor microenvironment is composed of tumor cells, stromal cells and leukocytes, including innate and adaptive immune cells, and represents an ecological niche that regulates tumor development and progression. In general, inflammatory cells are considered to contribute to tumor progression through various mechanisms, including the formation of an immunosuppressive microenvironment. Macrophages and neutrophils are important components of the tumor microenvironment and can act as a double-edged sword, promoting or inhibiting the development of the tumor. Targeting of the immune system is emerging as an important therapeutic strategy for cancer patients. However, the efficacy of the various immunotherapies available is still limited. Given the crucial importance of the crosstalk between macrophages and neutrophils and other immune cells in the formation of the anti-tumor immune response, targeting these interactions may represent a promising therapeutic approach against cancer. Here we will review the current knowledge of the role played by macrophages and neutrophils in cancer, focusing on their interaction with other immune cells.
Collapse
Affiliation(s)
| | | | | | - Giovanna Grieco
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Piera Molisso
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Sebastien Jaillon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
37
|
Tang L, Fu C, Liu H, Yin Y, Cao Y, Feng J, Zhang A, Wang W. Chemoimmunotherapeutic Nanogel for Pre- and Postsurgical Treatment of Malignant Melanoma by Reprogramming Tumor-Associated Macrophages. NANO LETTERS 2024; 24:1717-1728. [PMID: 38270376 DOI: 10.1021/acs.nanolett.3c04563] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Surgery is the primary method to treat malignant melanoma; however, the residual microtumors that cannot be resected completely often trigger tumor recurrence, causing tumor-related mortality following melanoma resection. Herein, we developed a feasible strategy based on the combinational chemoimmunotherapy by cross-linking carboxymethyl chitosan (CMCS)-originated polymetformin (PolyMetCMCS) with cystamine to prepare stimuli-responsive nanogel (PMNG) owing to the disulfide bond in cystamine that can be cleaved by the massive glutathione (GSH) in tumor sites. Then, chemotherapeutic agent doxorubicin (DOX) was loaded in PMNG, which was followed by a hyaluronic acid coating to improve the overall biocompatibility and targeting ability of the prepared nanogel (D@HPMNG). Notably, PMNG effectively reshaped the tumor immune microenvironment by reprogramming tumor-associated macrophage phenotypes and recruiting intratumoral CD8+ T cells owing to the inherited immunomodulatory capability of metformin. Consequently, D@HPMNG treatment remarkably suppressed melanoma growth and inhibited its recurrence after surgical resection, proposing a promising solution for overcoming lethal melanoma recurrence.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Cong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Hening Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yuqi Cao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Jingwen Feng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Aining Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Cosmetics, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
38
|
Luan X, Lei T, Fang J, Liu X, Fu H, Li Y, Chu W, Jiang P, Tong C, Qi H, Fu Y. Blockade of C5a receptor unleashes tumor-associated macrophage antitumor response and enhances CXCL9-dependent CD8 + T cell activity. Mol Ther 2024; 32:469-489. [PMID: 38098230 PMCID: PMC10861991 DOI: 10.1016/j.ymthe.2023.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 12/25/2023] Open
Abstract
Macrophages play a crucial role in shaping the immune state within the tumor microenvironment (TME) and are often influenced by tumors to hinder antitumor immunity. However, the underlying mechanisms are still elusive. Here, we observed abnormal expression of complement 5a receptor (C5aR) in human ovarian cancer (OC), and identified high levels of C5aR expression on tumor-associated macrophages (TAMs), which led to the polarization of TAMs toward an immunosuppressive phenotype. C5aR knockout or inhibitor treatment restored TAM antitumor response and attenuated tumor progression. Mechanistically, C5aR deficiency reprogrammed macrophages from a protumor state to an antitumor state, associating with the upregulation of immune response and stimulation pathways, which in turn resulted in the enhanced antitumor response of cytotoxic T cells in a manner dependent on chemokine (C-X-C motif) ligand 9 (CXCL9). The pharmacological inhibition of C5aR also improved the efficacy of immune checkpoint blockade therapy. In patients, C5aR expression associated with CXCL9 production and infiltration of CD8+ T cells, and a high C5aR level predicted poor clinical outcomes and worse benefits from anti-PD-1 therapy. Thus, our study sheds light on the mechanisms underlying the modulation of TAM antitumor immune response by the C5a-C5aR axis and highlights the potential of targeting C5aR for clinical applications.
Collapse
Affiliation(s)
- Xiaojin Luan
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting Lei
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jie Fang
- Department of Gynecology, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang 212001, Jiangsu, China
| | - Xue Liu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Huijia Fu
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yiran Li
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Chu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Peng Jiang
- Department of Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chao Tong
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Hongbo Qi
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China.
| | - Yong Fu
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
39
|
Lee J, Kim D, Kong J, Ha D, Kim I, Park M, Lee K, Im SH, Kim S. Cell-cell communication network-based interpretable machine learning predicts cancer patient response to immune checkpoint inhibitors. SCIENCE ADVANCES 2024; 10:eadj0785. [PMID: 38295179 PMCID: PMC10830106 DOI: 10.1126/sciadv.adj0785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. However, only some patients respond to ICIs, and current biomarkers for ICI efficacy have limited performance. Here, we devised an interpretable machine learning (ML) model trained using patient-specific cell-cell communication networks (CCNs) decoded from the patient's bulk tumor transcriptome. The model could (i) predict ICI efficacy for patients across four cancer types (median AUROC: 0.79) and (ii) identify key communication pathways with crucial players responsible for patient response or resistance to ICIs by analyzing more than 700 ICI-treated patient samples from 11 cohorts. The model prioritized chemotaxis communication of immune-related cells and growth factor communication of structural cells as the key biological processes underlying response and resistance to ICIs, respectively. We confirmed the key communication pathways and players at the single-cell level in patients with melanoma. Our network-based ML approach can be used to expand ICIs' clinical benefits in cancer patients.
Collapse
Affiliation(s)
- Juhun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Donghyo Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - JungHo Kong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Doyeon Ha
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Inhae Kim
- ImmunoBiome Inc., Pohang 166-20, Korea
| | - Minhyuk Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Kwanghwan Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
- ImmunoBiome Inc., Pohang 166-20, Korea
- Institute of Convergence Science, Yonsei University, Seoul 120-749, Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
- Institute of Convergence Science, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
40
|
Santana-Hernández S, Suarez-Olmos J, Servitja S, Berenguer-Molins P, Costa-Garcia M, Comerma L, Rea A, Perera-Bel J, Menendez S, Arpí O, Bermejo B, Martínez MT, Cejalvo JM, Comino-Méndez I, Pascual J, Alba E, López-Botet M, Rojo F, Rovira A, Albanell J, Muntasell A. NK cell-triggered CCL5/IFNγ-CXCL9/10 axis underlies the clinical efficacy of neoadjuvant anti-HER2 antibodies in breast cancer. J Exp Clin Cancer Res 2024; 43:10. [PMID: 38167224 PMCID: PMC10763072 DOI: 10.1186/s13046-023-02918-4] [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: 11/07/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The variability in responses to neoadjuvant treatment with anti-HER2 antibodies prompts to personalized clinical management and the development of innovative treatment strategies. Tumor-infiltrating Natural Killer (TI-NK) cells can predict the efficacy of HER2-targeted antibodies independently from clinicopathological factors in primary HER2-positive breast cancer patients. Understanding the mechanism/s underlying this association would contribute to optimizing patient stratification and provide the rationale for combinatorial approaches with immunotherapy. METHODS We sought to uncover processes enriched in NK cell-infiltrated tumors as compared to NK cell-desert tumors by microarray analysis. Findings were validated in clinical trial-derived transcriptomic data. In vitro and in vivo preclinical models were used for mechanistic studies. Findings were analysed in clinical samples (tumor and serum) from breast cancer patients. RESULTS NK cell-infiltrated tumors were enriched in CCL5/IFNG-CXCL9/10 transcripts. In multivariate logistic regression analysis, IFNG levels underlie the association between TI-NK cells and pathological complete response to neoadjuvant treatment with trastuzumab. Mechanistically, the production of IFN-ɣ by CD16+ NK cells triggered the secretion of CXCL9/10 from cancer cells. This effect was associated to tumor growth control and the conversion of CD16 into CD16-CD103+ NK cells in humanized in vivo models. In human breast tumors, the CD16 and CD103 markers identified lineage-related NK cell subpopulations capable of producing CCL5 and IFN-ɣ, which correlated with tissue-resident CD8+ T cells. Finally, an early increase in serum CCL5/CXCL9 levels identified patients with NK cell-rich tumors showing good responses to anti-HER2 antibody-based neoadjuvant treatment. CONCLUSIONS This study identifies specialized NK cell subsets as the source of IFN-ɣ influencing the clinical efficacy of anti-HER2 antibodies. It also reveals the potential of serum CCL5/CXCL9 as biomarkers for identifying patients with NK cell-rich tumors and favorable responses to anti-HER2 antibody-based neoadjuvant treatment.
Collapse
Affiliation(s)
| | | | - Sonia Servitja
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Oncology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
| | | | | | - Laura Comerma
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Anna Rea
- University Pompeu Fabra, Barcelona, Spain
| | - Julia Perera-Bel
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Silvia Menendez
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Oriol Arpí
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Begoña Bermejo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Department of Oncology, Hospital Clínico de Valencia, Valencia, Spain
| | | | | | - Iñaki Comino-Méndez
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Hospitales Universitarios Regional y Virgen de La Victoria, Málaga, Spain
- The Biomedical Research Institute of Málaga, Málaga, Spain
| | - Javier Pascual
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Hospitales Universitarios Regional y Virgen de La Victoria, Málaga, Spain
- The Biomedical Research Institute of Málaga, Málaga, Spain
| | - Emilio Alba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Hospitales Universitarios Regional y Virgen de La Victoria, Málaga, Spain
- The Biomedical Research Institute of Málaga, Málaga, Spain
| | - Miguel López-Botet
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- University Pompeu Fabra, Barcelona, Spain
| | - Federico Rojo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- Department of Pathology, IIS 'Fundación Jimenez Díaz University Hospital', Madrid, Spain
| | - Ana Rovira
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Oncology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
| | - Joan Albanell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Oncology Department, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain
- University Pompeu Fabra, Barcelona, Spain
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERonc), Madrid, Spain.
- Universitat Autònoma de Barcelona, Hospital del Mar Research Institute (IMIM), Doctor Aiguader, 88, 08003, Barcelona, Spain.
| |
Collapse
|
41
|
Li W, Xu T, Jin H, Li M, Jia Q. Emerging role of cancer-associated fibroblasts in esophageal squamous cell carcinoma. Pathol Res Pract 2024; 253:155002. [PMID: 38056131 DOI: 10.1016/j.prp.2023.155002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Esophageal carcinoma is the sixth leading cause of cancer death globally and the majority of global cases are esophageal squamous cell carcinoma (ESCC). Difficulty in diagnosis exists as more than 70% of ESCC patients are diagnosed at the intermediate or advanced stage. Cancer-associated fibroblasts (CAFs) have been considered one of the crucial components in the process of tumor growth, promoting communications between cancer cells and the tumor microenvironment (TME). CAFs grow alongside malignancies dynamically and interact with ESCC cells to promote their progression, proliferation, invasion, tumor escape, chemo- and radio-resistance, etc. It is believed that CAFs qualify as a promising direction for treatment. Analyzing CAFs' subtypes and functions will elucidate the involvement of CAFs in ESCC and aid in therapeutics. This review summarizes current information on CAFs in ESCC and focuses on the latest interaction between CAFs and ESCC cancer cell discoveries. The origin of CAFs and their communication with ESCC cells and TME are also demonstrated. On the foundation of a thorough analysis, we highlight the clinical prospects and CAFs-related therapies in ESCC in the future.
Collapse
Affiliation(s)
- Wenqing Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
42
|
Zhou H, Yao J, Zhong Z, Wei H, He Y, Li W, Hu K. Lactate-Induced CCL8 in Tumor-Associated Macrophages Accelerates the Progression of Colorectal Cancer through the CCL8/CCR5/mTORC1 Axis. Cancers (Basel) 2023; 15:5795. [PMID: 38136340 PMCID: PMC10741879 DOI: 10.3390/cancers15245795] [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: 10/29/2023] [Revised: 11/28/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in shaping the tumor microenvironment. Lactic acid (LA) has been identified as an influential factor in promoting immune escape and tumor progression. However, the mechanisms through which LA modulates TAMs in colorectal cancer (CRC) remain poorly understood. We used qRT-PCR to quantify the expression of LA-related genes (LDHA and LAMP2) in CRC tumor tissues and adjacent nontumor tissues (n = 64). The biological effects and mechanisms of LA on macrophages and tumors were evaluated via qRT-PCR, Western blot, RNA-seq, wound healing assay, colony formation assay in vitro, and allograft mouse tumor models in vivo. We found the expression of LDHA and LAMP2 was highly elevated in the tumor regions and positively associated with a poor clinical stage of CRC. A high concentration of LA was generated under hypoxia; it could promote tumor progression and metastasis with the involvement of macrophages. The inhibition of LA release impaired this protumor phenomenon. Mechanically, LA induced M2 macrophages through the AKT/ERK signaling pathway; subsequently, M2 macrophages secreted CCL8 and facilitated the proliferation and metastasis of CRC cells by activating the CCL8/CCR5/mTORC1 axis. This effect was inhibited by the antagonist or knockdown of CCR5. In conclusion, lactate-induced CCL8 in TAMs accelerated CRC proliferation and metastasis through the CCL8/CCR5/mTORC1 axis.
Collapse
Affiliation(s)
- Hui Zhou
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, China; (H.Z.); (Y.H.)
| | - Jiayi Yao
- Center of Excellence, The Seventh Affiliated Hospital of Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, China;
| | - Zhaozhong Zhong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou 510630, China;
| | - Hongfa Wei
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Jinping District, Shantou 515041, China;
| | - Yulong He
- Digestive Diseases Center, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628 Zhenyuan Road, Shenzhen 518107, China; (H.Z.); (Y.H.)
| | - Wenchao Li
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou 510630, China
| | - Kunpeng Hu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe Road, Tianhe District, Guangzhou 510630, China
| |
Collapse
|
43
|
Liu X, Zhang Q, Liang Y, Xiong S, Cai Y, Cao J, Xu Y, Xu X, Wu Y, Lu Q, Xu X, Luo B. Nanoparticles (NPs)-mediated Siglec15 silencing and macrophage repolarization for enhanced cancer immunotherapy. Acta Pharm Sin B 2023; 13:5048-5059. [PMID: 38045048 PMCID: PMC10692376 DOI: 10.1016/j.apsb.2023.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 12/05/2023] Open
Abstract
T cell infiltration and proliferation in tumor tissues are the main factors that significantly affect the therapeutic outcomes of cancer immunotherapy. Emerging evidence has shown that interferon-gamma (IFNγ) could enhance CXCL9 secretion from macrophages to recruit T cells, but Siglec15 expressed on TAMs can attenuate T cell proliferation. Therefore, targeted regulation of macrophage function could be a promising strategy to enhance cancer immunotherapy via concurrently promoting the infiltration and proliferation of T cells in tumor tissues. We herein developed reduction-responsive nanoparticles (NPs) made with poly (disulfide amide) (PDSA) and lipid-poly (ethylene glycol) (lipid-PEG) for systemic delivery of Siglec15 siRNA (siSiglec15) and IFNγ for enhanced cancer immunotherapy. After intravenous administration, these cargo-loaded could highly accumulate in the tumor tissues and be efficiently internalized by tumor-associated macrophages (TAMs). With the highly concentrated glutathione (GSH) in the cytoplasm to destroy the nanostructure, the loaded IFNγ and siSiglec15 could be rapidly released, which could respectively repolarize macrophage phenotype to enhance CXCL9 secretion for T cell infiltration and silence Siglec15 expression to promote T cell proliferation, leading to significant inhibition of hepatocellular carcinoma (HCC) growth when combining with the immune checkpoint inhibitor. The strategy developed herein could be used as an effective tool to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaodi Liu
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qi Zhang
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yixia Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shiyu Xiong
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yan Cai
- Department of Ultrasound, Central People's Hospital of Zhanjiang, Zhanjiang 524045, China
| | - Jincheng Cao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yanni Xu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaolin Xu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ye Wu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qiang Lu
- Department of Ultrasound, Laboratory of Ultrasound Imaging and Drug, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoding Xu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| |
Collapse
|
44
|
Xu M, Cheng Y, Meng R, Yang P, Chen J, Qiao Z, Wu J, Qian K, Li Y, Wang P, Zhou L, Wang T, Sheng D, Zhang Q. Enhancement of Microglia Functions by Developed Nano-Immuno-Synergist to Ameliorate Immunodeficiency for Malignant Glioma Treatment. Adv Healthc Mater 2023; 12:e2301861. [PMID: 37573475 DOI: 10.1002/adhm.202301861] [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: 06/12/2023] [Revised: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Resident microglia are key factors in mediating immunity against brain tumors, but the microglia in malignant glioma are functionally impaired. Little immunotherapy is explored to restore microglial function against glioma. Herein, oleanolic acid (OA) (microglia "restorer") and D PPA-1 peptide (immune checkpoint blockade) are integrated on a nano-immuno-synergist (D PAM@OA) to work coordinately. The self-assembled OA core is coated with macrophage membrane for efficient blood-brain barrier penetration and microglia targeting, on which D PPA-1 peptide is attached via acid-sensitive bonds for specific release in tumor microenvironment. With the enhanced accumulation of the dual drugs in their respective action sites, D PAM@OA effectively promotes the recruitment and activation of effector T cells by inhibiting aberrant activation of Signal transducer and activator of transcription (STAT-3) pathway in microglia, and assists activated effector T cells in killing tumor cells by blocking elevated immune checkpoint proteins in malignant glioma. Eventually, as adjuvant therapy, the rationally designed nano-immuno-synergist hinders malignant glioma progression and recurrence with or without temozolomide. The work demonstrates the feasibility of a nano-formulation for microglia-based immunotherapy, which may provide a new direction for the treatment of brain tumors.
Collapse
Affiliation(s)
- Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jian Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Zhen Qiao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Pengzhen Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Dongyu Sheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
45
|
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
|
46
|
Gao Y, Ju Y, Ren X, Zhang L, Yin X. Enhanced infection efficiency and cytotoxicity mediated by vpx-containing lentivirus in chimeric antigen receptor macrophage (CAR-M). Heliyon 2023; 9:e21886. [PMID: 38058430 PMCID: PMC10696197 DOI: 10.1016/j.heliyon.2023.e21886] [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: 02/07/2023] [Revised: 09/26/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
Genetically modified macrophage infusion has been proven to be a novel treatment for cancer. One of the most important processes in macrophage-based therapy is the efficient transfer of genes. HIV-1-derived lentiviruses were widely used as delivery vectors in chimeric antigen receptor T and NK cell construction. While macrophages are relatively refractory to this lentiviral vector transduction as a result of the myeloid-specific restriction factor SAMHD1, which inhibited the virion cycle through exhausting the dNTPs pool and degradating RNAs. An efficient macrophage transduction strategy has been developed via packaging the HIV-2 accessory protein Vpx into the virion. Vpx counteracts SAMHD1 through CRL4 (DCAF1) E3 ubiquitin ligase mediated SAMHD1 degradation, yet the influence by the introduction of Vpx on macrophage has not been fully evaluated. Here, we constructed the chimeric lentiviral vector HIV-1-Vpx and systematically analyzed the infection efficiency of this vector in time-dependent manner. Our results showed that the simplified chimeric virus exhibited dramatically enhanced infection in human macrophages compared to normal lentivirus. Moreover, transcriptome sequencing was performed to evaluate the cellular status after chimeric virus infection. The sequencing results indicated that Vpx introduction promoted macrophage remodeling towards a proinflammatory phenotype, without affecting classic M1/M2 cell surface markers. Our results suggest that the Vpx-containing lentivirus could be used as an ideal tool for the generation of genetically engineered macrophages with high gene transfer efficiency and poised proinflammatory gene sets, especially for solid tumor treatment.
Collapse
Affiliation(s)
- Yun Gao
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Yue Ju
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China
| | - Xiaomeng Ren
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Luo Zhang
- Research Center of Bioengineering, the Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiushan Yin
- Applied Biology Laboratory, College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
- Roc Rock Biotechnology (Shenzhen), Shenzhen, 518118, China
| |
Collapse
|
47
|
Rocha GIY, Gomes JEM, Leite ML, da Cunha NB, Costa FF. Epigenome-Driven Strategies for Personalized Cancer Immunotherapy. Cancer Manag Res 2023; 15:1351-1367. [PMID: 38058537 PMCID: PMC10697012 DOI: 10.2147/cmar.s272031] [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: 05/13/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023] Open
Abstract
Fighting cancer remains one of the greatest challenges for science in the 21st century. Advances in immunotherapy against different types of cancer have greatly contributed to the treatment, remission, and cure of patients. In this context, knowledge of epigenetic phenomena, their relationship with tumor cells and how the immune system can be epigenetically modulated represent some of the greatest advances in the development of anticancer therapies. Epigenetics is a rapidly growing field that studies how environmental factors can affect gene expression without altering DNA sequence. Epigenomic changes include DNA methylation, histone modifications, and non-coding RNA regulation, which impact cellular function. Epigenetics has shown promise in developing cancer therapies, such as immunotherapy, which aims to stimulate the immune system to attack cancer cells. For example, PD-1 and PD-L1 are biomarkers that regulate the immune response to cancer cells and recent studies have shown that epigenetic modifications can affect their expression, potentially influencing the efficacy of immunotherapy. New therapies targeting epigenetic modifications, such as histone deacetylases and DNA methyltransferases, are being developed for cancer treatment, and some have shown promise in preclinical studies and clinical trials. With growing understanding of epigenetic regulation, we can expect more personalized and effective cancer immunotherapies in the future. This review highlights key advances in the use of epigenetic and epigenomic tools and modern immuno-oncology strategies to treat several types of tumors.
Collapse
Affiliation(s)
| | | | - Michel Lopes Leite
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Department of Cell Biology, Institute of Biological Sciences, Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
| | - Nicolau B da Cunha
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Faculty of Agronomy and Veterinary Medicine (FAV), Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
- Graduate Program in Agronomy, Campus Darcy Ribeiro, University of Brasilia (UnB), Brasília, DF, Brazil
| | - Fabricio F Costa
- Genomic Sciences and Biotechnology Program, Catholic University of Brasilia, Brasília, DF, Brazil
- Cancer Biology and Epigenomics Program, Northwestern University’s Feinberg School of Medicine, Chicago, IL, USA
- Genomic Enterprise, San FranciscoCA, USA
| |
Collapse
|
48
|
Pecharromán I, Solé L, Álvarez‐Villanueva D, Lobo‐Jarne T, Alonso‐Marañón J, Bertran J, Guillén Y, Montoto Á, Martínez‐Iniesta M, García‐Hernández V, Giménez G, Salazar R, Santos C, Garrido M, Borràs E, Sabidó E, Bonfill‐Teixidor E, Iurlaro R, Seoane J, Villanueva A, Iglesias M, Bigas A, Espinosa L. IκB kinase-α coordinates BRD4 and JAK/STAT signaling to subvert DNA damage-based anticancer therapy. EMBO J 2023; 42:e114719. [PMID: 37737566 PMCID: PMC10620764 DOI: 10.15252/embj.2023114719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/28/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023] Open
Abstract
Activation of the IκB kinase (IKK) complex has recurrently been linked to colorectal cancer (CRC) initiation and progression. However, identification of downstream effectors other than NF-κB has remained elusive. Here, analysis of IKK-dependent substrates in CRC cells after UV treatment revealed that phosphorylation of BRD4 by IKK-α is required for its chromatin-binding at target genes upon DNA damage. Moreover, IKK-α induces the NF-κB-dependent transcription of the cytokine LIF, leading to STAT3 activation, association with BRD4 and recruitment to specific target genes. IKK-α abrogation results in defective BRD4 and STAT3 functions and consequently irreparable DNA damage and apoptotic cell death upon different stimuli. Simultaneous inhibition of BRAF-dependent IKK-α activity, BRD4, and the JAK/STAT pathway enhanced the therapeutic potential of 5-fluorouracil combined with irinotecan in CRC cells and is curative in a chemotherapy-resistant xenograft model. Finally, coordinated expression of LIF and IKK-α is a poor prognosis marker for CRC patients. Our data uncover a functional link between IKK-α, BRD4, and JAK/STAT signaling with clinical relevance.
Collapse
Affiliation(s)
- Irene Pecharromán
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Laura Solé
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Daniel Álvarez‐Villanueva
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
| | - Teresa Lobo‐Jarne
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Josune Alonso‐Marañón
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Joan Bertran
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Faculty of Science and TechnologyUniversity of Vic – Central University of CataloniaVicSpain
| | - Yolanda Guillén
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Ángela Montoto
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - María Martínez‐Iniesta
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
| | - Violeta García‐Hernández
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Gemma Giménez
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Ramon Salazar
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)‐CIBERONCL'Hospitalet de LlobregatBarcelonaSpain
| | - Cristina Santos
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)‐CIBERONCL'Hospitalet de LlobregatBarcelonaSpain
| | - Marta Garrido
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| | - Eva Borràs
- Proteomics Unit, Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Proteomics UnitUniversitat Pompeu FabraBarcelonaSpain
| | - Eduard Sabidó
- Proteomics Unit, Centre for Genomic Regulation (CRG)Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Proteomics UnitUniversitat Pompeu FabraBarcelonaSpain
| | - Ester Bonfill‐Teixidor
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Raffaella Iurlaro
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
| | - Joan Seoane
- Vall d'Hebron Institute of Oncology (VHIO), CIBERONCVall d'Hebron University Hospital, Universitat Autònoma de BarcelonaBarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet del LlobregatBarcelonaSpain
- Xenopat S.L., Parc Cientific de Barcelona (PCB)BarcelonaSpain
| | - Mar Iglesias
- Department of Pathology, Institut Mar d'Investigacions Mèdiques, CIBERONCUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Anna Bigas
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
- Josep Carreras Leukemia Research InstituteBadalonaSpain
| | - Lluís Espinosa
- Cancer Research Program, Institut Mar d'Investigacions Mèdiques, CIBERONCHospital del MarBarcelonaSpain
| |
Collapse
|
49
|
Soler MF, Abaurrea A, Azcoaga P, Araujo AM, Caffarel MM. New perspectives in cancer immunotherapy: targeting IL-6 cytokine family. J Immunother Cancer 2023; 11:e007530. [PMID: 37945321 PMCID: PMC10649711 DOI: 10.1136/jitc-2023-007530] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
Chronic inflammation has been recognized as a canonical cancer hallmark. It is orchestrated by cytokines, which are master regulators of the tumor microenvironment (TME) as they represent the main communication bridge between cancer cells, the tumor stroma, and the immune system. Interleukin (IL)-6 represents a keystone cytokine in the link between inflammation and cancer. Many cytokines from the IL-6 family, which includes IL-6, oncostatin M, leukemia inhibitory factor, IL-11, IL-27, IL-31, ciliary neurotrophic factor, cardiotrophin 1, and cardiotrophin-like cytokine factor 1, have been shown to elicit tumor-promoting roles by modulating the TME, making them attractive therapeutic targets for cancer treatment.The development of immune checkpoint blockade (ICB) immunotherapies has radically changed the outcome of some cancers including melanoma, lung, and renal, although not without hurdles. However, ICB shows limited efficacy in other solid tumors. Recent reports support that chronic inflammation and IL-6 cytokine signaling are involved in resistance to immunotherapy. This review summarizes the available preclinical and clinical data regarding the implication of IL-6-related cytokines in regulating the immune TME and the response to ICB. Moreover, the potential clinical benefit of combining ICB with therapies targeting IL-6 cytokine members for cancer treatment is discussed.
Collapse
Affiliation(s)
- Maria Florencia Soler
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Andrea Abaurrea
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Peio Azcoaga
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Angela M Araujo
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Maria M Caffarel
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
50
|
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, Yu Z, Wu B, Liu S, Xu W, Zheng C, Schuchter LM, Karakousis GC, Mitchell TC, Amaravadi R, Flowers AJ, Gimotty PA, Xiao M, Mills G, Herlyn M, Dong H, Mitchell MJ, Kim J, Xu X, Guo W. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep 2023; 42:113224. [PMID: 37805922 PMCID: PMC10697782 DOI: 10.1016/j.celrep.2023.113224] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 06/15/2023] [Accepted: 09/20/2023] [Indexed: 10/10/2023] Open
Abstract
Macrophages play a pivotal role in tumor immunity. We report that reprogramming of macrophages to tumor-associated macrophages (TAMs) promotes the secretion of exosomes. Mechanistically, increased exosome secretion is driven by MADD, which is phosphorylated by Akt upon TAM induction and activates Rab27a. TAM exosomes carry high levels of programmed death-ligand 1 (PD-L1) and potently suppress the proliferation and function of CD8+ T cells. Analysis of patient melanoma tissues indicates that TAM exosomes contribute significantly to CD8+ T cell suppression. Single-cell RNA sequencing analysis showed that exosome-related genes are highly expressed in macrophages in melanoma; TAM-specific RAB27A expression inversely correlates with CD8+ T cell infiltration. In a murine melanoma model, lipid nanoparticle delivery of small interfering RNAs (siRNAs) targeting macrophage RAB27A led to better T cell activation and sensitized tumors to anti-programmed cell death protein 1 (PD-1) treatment. Our study demonstrates tumors use TAM exosomes to combat CD8 T cells and suggests targeting TAM exosomes as a potential strategy to improve immunotherapies.
Collapse
Affiliation(s)
- Wenqun Zhong
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Youtao Lu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhiyuan Qin
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Zhang
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ziyan Yu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Wu
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron J Flowers
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phyllis A Gimotty
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Gordon Mills
- Division of Oncological Science, School of Medicine, and Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haidong Dong
- Departments of Urology and Immunology, Mayo College of Medicine and Science, Rochester, MN 55905, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhyong Kim
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|