1
|
Chen F, Guo S, Li Y, Lu Y, Liu L, Chen S, An J, Zhang G. Fusobacterium nucleatum-driven CX3CR1 + PD-L1 + phagocytes route to tumor tissues and reshape tumor microenvironment. Gut Microbes 2025; 17:2442037. [PMID: 39710592 DOI: 10.1080/19490976.2024.2442037] [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: 09/01/2024] [Revised: 11/18/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
The intracellular bacterium Fusobacterium nucleatum (Fn) mediates tumorigenesis and progression in colorectal cancer (CRC). However, the origin of intratumoral Fn and the role of Fn-infected immunocytes in the tumor microenvironment remain unclear. Here, we observed that Fn-infected neutrophils/macrophages (PMNs/MΦs), especially PMNs, accumulate in tumor tissues and fecal Fn abundance correlates positively with an abundance of blood PD-L1+ PMNs in CRC patients. Moreover, Fn accumulates in tumor tissues of tumor-bearing mice via intragingival infection and intravenous injection. Mechanistically, Fn can survive inside PMNs by reducing intracellular ROS levels and producing H2S. Specifically, the lysozyme inhibitor Fn1792 as a novel virulence factor of Fn suppressed apoptosis of phagocytes by inducing CX3CR1 expression. Furthermore, Fn-driven CX3CR1+PD-L1+ phagocytes transfer intracellular Fn to tumor cells, which recruit PMNs/MΦs through the CXCL2/8-CXCR2 and CCL5/CCR5 axes. Consequently, CX3CR1+PD-L1+ PMNs infiltration promotes CRC metastasis and weakens the efficacy of immunotherapy. Treatment with the doxycycline eradicated intracellular Fn, thereby reducing the CX3CR1+PD-L1+ PMNs populations and slowing Fn-promoted tumor growth and metastasis in mice. These results suggest phagocytes as Fn-presenting cells use mutualistic strategies to home to tumor tissues and induce immunosuppression, and treatment with ROS-enhanced antibiotics can inhibit Fn-positive tumor progression.
Collapse
Affiliation(s)
- Fangfang Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Songhe Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yiqiu Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yongfan Lu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Le Liu
- Department of Gastroenterology, Integrated Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Shengxin Chen
- Graduate School, Chinese PLA General Hospital, Beijing, China
| | - Jun An
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
2
|
Lepsenyi M, Valdimarsson V, Algethami N, Thorlacius H, Ghanipour L, Cashin P, Asplund D, Lindskog EB, Palmer GJ, Nilsson PJ, Syk I. Postoperative leukopenia after cytoreductive surgery and hypertherm intraperitoneal chemotherapy for colorectal carcinomatosis- causes and implication on outcomes in a population-based study. World J Surg Oncol 2025; 23:173. [PMID: 40301901 PMCID: PMC12042315 DOI: 10.1186/s12957-025-03821-2] [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: 02/21/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Leukocytes have been reported to have tumor stimulating effects in colorectal cancer, among other malignancies. In line with this, earlier research has shown improved disease-free survival in patients with postoperative neutropenia compared to non-neutropenic patients following cytoreductive surgery (CRS) and hypertherm intraperitoneal chemotherapy (HIPEC). AIM To evaluate the impact of postoperative leukopenia after CRS and HIPEC on recurrence rate, survival, and risk of complications. METHODS All CRS and HIPEC-procedures for colorectal adenocarcinoma in the national Swedish HIPEC-registry since 2015 and local registries in Uppsala and Malmö since 2003 until December 31st, 2021, were included (n = 921). Patients who did not complete a full CRS and HIPEC procedure (n = 99), had incomplete macroscopic cytoreduction (n = 25) or a lack of information on leukocyte count (n = 213) were excluded, resulting in 584 analyzed cases. Primary outcome was overall recurrence rate. Secondary outcomes were overall survival, recurrence-free survival, and perioperative complications. RESULTS Postoperative leukopenia was observed in 54 (9.2%) cases of which 32 (5.5%) developed severe leukopenia. No differences in patient characteristics were noted between those with or without leukopenia. There were no differences in 3-year recurrence rate, overall survival or 3-year recurrence-free survival, between the groups. Neoadjuvant chemotherapy treatment, HR 1.32 (95% CI: 1.02-1.71), higher PCI-score, HR 1.50 (95% CI: 1.09-2.05) and higher pN-stage HR 2.52 (95% CI: 1.74-3.65) were associated with higher 3-year recurrence rate. 3-year mortality was associated with neoadjuvant chemotherapy treatment, HR 1.82 (95% CI: 1.06-3.11), severe postoperative complication, HR 2.39 (95% CI: 1.39-4.13) and high PCI-score, HR 2.60 (95% CI: 1.31-5.14). Treatment with combined oxaliplatin/irinotecan, HR 12.34 (95% CI: 4.51-33.74) was associated with developing postoperative leukopenia. Longer operation time, HR 2.30 (95% CI: 1.55-3.42), and severe leukopenia, HR 3.50 (95% CI: 1.25-9.77) were associated with postoperative complication. CONCLUSIONS Postoperative leukopenia did not impact recurrence rate or long-term survival in a statistically significant manner. Neoadjuvant chemotherapy and high PCI-score were associated with both recurrent disease and mortality within 3 years.
Collapse
Affiliation(s)
- Mattias Lepsenyi
- Department of Clinical Sciences Malmö, Section of Surgery, Lund University, Skåne University Hospital, Inga Marie Nilssons gata 47, Malmö, 20502, Sweden.
| | - Valentinus Valdimarsson
- Department of Clinical Sciences Malmö, Section of Surgery, Lund University, Skåne University Hospital, Inga Marie Nilssons gata 47, Malmö, 20502, Sweden
| | - Nader Algethami
- Department of Clinical Sciences Malmö, Section of Surgery, Lund University, Skåne University Hospital, Inga Marie Nilssons gata 47, Malmö, 20502, Sweden
| | - Henrik Thorlacius
- Department of Clinical Sciences Malmö, Section of Surgery, Lund University, Skåne University Hospital, Inga Marie Nilssons gata 47, Malmö, 20502, Sweden
| | - Lana Ghanipour
- Department of Surgical Sciences, Section of Surgery, Uppsala University, Uppsala, Akademiska sjukhuset, Sweden
| | - Peter Cashin
- Department of Surgical Sciences, Section of Surgery, Uppsala University, Uppsala, Akademiska sjukhuset, Sweden
| | - Dan Asplund
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, dept of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elinor Bexe Lindskog
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, dept of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Gabriella Jansson Palmer
- Department of Pelvic cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Per J Nilsson
- Department of Pelvic cancer, GI Oncology and Colorectal Surgery Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ingvar Syk
- Department of Clinical Sciences Malmö, Section of Surgery, Lund University, Skåne University Hospital, Inga Marie Nilssons gata 47, Malmö, 20502, Sweden
| |
Collapse
|
3
|
Correa E, Rendón JP, Bedoya-Betancur V, Montoya J, Duque JM, Naranjo TW. Standardization of a Preclinical Colon Cancer Model in Male and Female BALB/c Mice: Macroscopic and Microscopic Characterization from Pre-Neoplastic to Tumoral Lesions. Biomedicines 2025; 13:939. [PMID: 40299505 PMCID: PMC12024726 DOI: 10.3390/biomedicines13040939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objetives: This study standardized a chemically induced colorectal cancer (CRC) model using azoxymethane (AOM) and dextran sodium sulfate (DSS) in BALB/c mice, replicating the progression from preneoplastic lesions to adenocarcinoma observed in human colorectal carcinogenesis. Methods: The CCR-AOM/DSS model was standardized in male and female BALB/c mice. Two protocols were tested. Subsequently, the positive control group was established with nine evaluation points. Tumor progression was characterized histopathologically and corroborated by methylene blue staining and scanning electron microscopy. Results: Two cycles of 2% DSS combined with a single injection of AOM (10 mg/kg) were necessary to induce adenocarcinoma in 100% of the mice, with no significant sex-based differences in tumor development. Females showed earlier tumor susceptibility under certain protocols. Inflammatory processes played a critical role in tumorigenesis, with neutrophil infiltration and fibrosis observed. Conclusions: The findings align with previous reports, emphasizing the influence of DSS cycles, molecular weight, and mouse strain on model outcomes. This standardized model provides a reliable platform for the preclinical evaluation of novel preventive and therapeutic strategies for CRC.
Collapse
Affiliation(s)
- Elizabeth Correa
- Medical and Experimental Mycology Group, CIB-UPB-UdeA-UDES, Corporación para Investigaciones Biológicas, Carrera 72 A # 78B-141, Medellin 050034, Colombia; (E.C.); (J.P.R.); (V.B.-B.); (J.M.)
| | - Juan Pablo Rendón
- Medical and Experimental Mycology Group, CIB-UPB-UdeA-UDES, Corporación para Investigaciones Biológicas, Carrera 72 A # 78B-141, Medellin 050034, Colombia; (E.C.); (J.P.R.); (V.B.-B.); (J.M.)
| | - Vanesa Bedoya-Betancur
- Medical and Experimental Mycology Group, CIB-UPB-UdeA-UDES, Corporación para Investigaciones Biológicas, Carrera 72 A # 78B-141, Medellin 050034, Colombia; (E.C.); (J.P.R.); (V.B.-B.); (J.M.)
| | - Juliana Montoya
- Medical and Experimental Mycology Group, CIB-UPB-UdeA-UDES, Corporación para Investigaciones Biológicas, Carrera 72 A # 78B-141, Medellin 050034, Colombia; (E.C.); (J.P.R.); (V.B.-B.); (J.M.)
| | - Julian Muñoz Duque
- Pathobiology Research Group QUIRON, Faculty of Agricultural Sciences, Universidad de Antioquia, Calle 70 # 52-21, Medellin 050036, Colombia;
| | - Tonny W. Naranjo
- Medical and Experimental Mycology Group, CIB-UPB-UdeA-UDES, Corporación para Investigaciones Biológicas, Carrera 72 A # 78B-141, Medellin 050034, Colombia; (E.C.); (J.P.R.); (V.B.-B.); (J.M.)
- School of Health Sciences, Universidad Pontificia Bolivariana, Calle 78 B 72 A-109, Medellin 050034, Colombia
| |
Collapse
|
4
|
Tang J, Chen L, Shen X, Xia T, Li Z, Chai X, Huang Y, Yang S, Peng X, Lai J, Li R, Xie L. Exploring the Role of Cellular Interactions in the Colorectal Cancer Microenvironment. J Immunol Res 2025; 2025:4109934. [PMID: 40255905 PMCID: PMC12008489 DOI: 10.1155/jimr/4109934] [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: 08/23/2024] [Accepted: 02/22/2025] [Indexed: 04/22/2025] Open
Abstract
Colorectal cancer (CRC) stands as one of the tumors with globally high incidence and mortality rates. In recent years, researchers have extensively explored the role of the tumor immune microenvironment (TME) in CRC, highlighting the crucial influence of immune cell populations in driving tumor progression and shaping therapeutic outcomes. The TME encompasses an array of cellular and noncellular constituents, spanning tumor cells, immune cells, myeloid cells, and tumor-associated fibroblasts, among others. However, the cellular composition within the TME is highly dynamic, evolving throughout different stages of tumor progression. These shifts in cell subpopulation proportions lead to a gradual transition in the immune response, shifting from an early antitumor growth to a late-stage environment that supports tumor survival. Therefore, it is crucial to further investigate and understand the complex interactions among the various cell populations within the TME. In this review, we explore the key cellular components of varying origins, subpopulations with shared origins, and noncellular elements within the CRC TME, examining their interconnections and critical considerations for developing personalized and precise immunotherapy strategies.
Collapse
Affiliation(s)
- Jiadai Tang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Liuhan Chen
- Department of Head and Neck Surgery Section II, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xin Shen
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Tingrong Xia
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Zhengting Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xiaoying Chai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Yao Huang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Shaoqiong Yang
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Xinjun Peng
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Junbo Lai
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Rui Li
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| | - Lin Xie
- Department of Gastrointestinal Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, Yunnan, China
| |
Collapse
|
5
|
Isermann T, Schneider KL, Wegwitz F, De Oliveira T, Conradi LC, Volk V, Feuerhake F, Papke B, Stintzing S, Mundt B, Kühnel F, Moll UM, Schulz-Heddergott R. Enhancement of colorectal cancer therapy through interruption of the HSF1-HSP90 axis by p53 activation or cell cycle inhibition. Cell Death Differ 2025:10.1038/s41418-025-01502-x. [PMID: 40204953 DOI: 10.1038/s41418-025-01502-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025] Open
Abstract
The stress-associated chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing oncoproteins. Most inhibitors target the key component, heat-shock protein 90 (HSP90). Although HSP90 inhibitors are highly tumor-selective, they fail in clinical trials. These failures are partly due to interference with a negative regulatory feedback loop in the heat-shock response (HSR): in response to HSP90 inhibition, there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock-factor 1 (HSF1). We recently identified that wild-type p53 reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here, we test whether in HSP90-based therapies, simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. We found that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids, and patient-derived organoids (PDOs). Mechanistically, upon combination therapy, CRC cells upregulate p53-associated pathways, apoptosis, and inflammatory pathways. Likewise, in a CRC mouse model, dual HSF1-HSP90 inhibition represses tumor growth and remodels immune cell composition. Importantly, inhibition of the cyclin-dependent kinases 4/6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Moreover, in p53-deficient CRC cells, HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR and reduces cancer growth. Likewise, p53-mutated PDOs respond to dual HSF1-HSP90 inhibition, providing a strategy to target CRC independent of the p53 status. In sum, we provide new options to improve HSP90-based therapies to enhance CRC therapies.
Collapse
Affiliation(s)
- Tamara Isermann
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kim Lucia Schneider
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Valery Volk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Björn Papke
- Laboratory of Molecular Tumor Pathology and Systems Biology, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Oncology and Cancer Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina Mundt
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
6
|
Wahnou H, El Kebbaj R, Hba S, Ouadghiri Z, El Faqer O, Pinon A, Liagre B, Limami Y, Duval RE. Neutrophils and Neutrophil-Based Drug Delivery Systems in Anti-Cancer Therapy. Cancers (Basel) 2025; 17:1232. [PMID: 40227814 PMCID: PMC11988188 DOI: 10.3390/cancers17071232] [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: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
Neutrophils, the most abundant white blood cells, play a dual role in cancer progression. While they can promote tumor growth, metastasis, and immune suppression, they also exhibit anti-tumorigenic properties by attacking cancer cells and enhancing immune responses. This review explores the complex interplay between neutrophils and the tumor microenvironment (TME), highlighting their ability to switch between pro- and anti-tumor phenotypes based on external stimuli. Pro-tumorigenic neutrophils facilitate tumor growth through mechanisms such as neutrophil extracellular traps (NETs), secretion of pro-inflammatory cytokines, and immune evasion strategies. They contribute to angiogenesis, tumor invasion, and metastasis by releasing vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs). Conversely, anti-tumor neutrophils enhance cytotoxicity by generating reactive oxygen species (ROS), promoting antibody-dependent cell-mediated cytotoxicity (ADCC), and activating other immune cells such as cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells. Recent advances in neutrophil-based drug delivery systems have harnessed their tumor-homing capabilities to improve targeted therapy. Neutrophil-mimicking nanoparticles and membrane-coated drug carriers offer enhanced drug accumulation in tumors, reduced systemic toxicity, and improved therapeutic outcomes. Additionally, strategies to modulate neutrophil activity, such as inhibiting their immunosuppressive functions or reprogramming them towards an anti-tumor phenotype, are emerging as promising approaches in cancer immunotherapy. Understanding neutrophil plasticity and their interactions with the TME provides new avenues for therapeutic interventions. Targeting neutrophil-mediated mechanisms could enhance existing cancer treatments and lead to the development of novel immunotherapies, ultimately improving patient survival and clinical outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Riad El Kebbaj
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | - Soufyane Hba
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Zaynab Ouadghiri
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Othman El Faqer
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, B.P 2693, Maarif, Casablanca 20100, Morocco; (H.W.); (S.H.); (Z.O.); (O.E.F.)
| | - Aline Pinon
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Bertrand Liagre
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France; (A.P.); (B.L.)
| | - Youness Limami
- Sciences and Engineering of Biomedicals, Biophysics and Health Laboratory, Higher Institute of Health Sciences, Hassan First University, Settat 26000, Morocco;
| | | |
Collapse
|
7
|
Zhong R, He H, Wang X. Novel neutrophil targeting platforms in treating Glioblastoma: Latest evidence and therapeutic approaches. Int Immunopharmacol 2025; 150:114173. [PMID: 39938169 DOI: 10.1016/j.intimp.2025.114173] [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: 10/08/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal type of primary brain tumor, characterized by its rapid growth, resistance to conventional therapies, and a highly immunosuppressive tumor microenvironment (TME). Recent studies have highlighted the critical role of neutrophils in the progression of GBM, where they contribute to tumor growth, invasion, and treatment resistance. As a result, neutrophils have emerged as a promising target for therapeutic intervention in GBM. Various strategies are being investigated to specifically target neutrophils within the GBM environment, including using small molecules, antibodies, and nanoparticle-based methods. These approaches aim to regulate neutrophils' recruitment, activation, and functions. This study reviews the latest findings regarding the involvement of neutrophils in GBM, explores potential techniques targeting neutrophils for therapeutic purposes, and discusses current clinical studies and prospects in this rapidly evolving field. By studying the diverse functions of neutrophils in GBM, these innovative therapeutic strategies can help address some of the most significant challenges in treating this malignancy.
Collapse
Affiliation(s)
- Rui Zhong
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Hongmei He
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China
| | - Xiande Wang
- Department of Neurosurgery, The First People's Hospital of Lin'an District, Hangzhou 311300, China.
| |
Collapse
|
8
|
Chen Z, Fang Y, Zhong S, Lin S, Yang X, Chen S. ITGB5 is a prognostic factor in colorectal cancer and promotes cancer progression and metastasis through the Wnt signaling pathway. Sci Rep 2025; 15:9225. [PMID: 40097546 PMCID: PMC11914080 DOI: 10.1038/s41598-025-93081-7] [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: 04/15/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Integrin beta5 (ITGB5) expression levels are dysregulated in a variety of cancers. However, the mechanism and clinical value of ITGB5 in colorectal cancer (CRC) remain unclear. The Gene Expression Omnibus (GEO) database, real-time PCR, Western blotting and immunohistochemistry were utilized to evaluate ITGB5 expression levels in CRC tissue. Clinical data from the GEO database were obtained to further explore the associations of ITGB5 with clinical features and patient survival. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and gene set enrichment analysis (GSEA) were performed to explore the functions and signaling pathways of ITGB5. In addition, ITGB5 expression was inhibited by siRNA, and the roles of ITGB5 in SW480 and RKO cell growth, migration and invasion, as well as in the Wnt/β-catenin signaling pathway, were investigated. Pancancer studies have shown that ITGB5 is highly expressed in a variety of cancers. Moreover, ITGB5 expression is significantly increased in CRC tissues and is correlated with TNM stage, invasion depth, lymph node metastasis and distant metastasis stage. Kaplan-Meier analysis and meta-analysis of the GSE39582 and GSE17538 datasets indicated that a high level of ITGB5 is a high risk factor for overall survival (OS) and disease-free survival (DFS). In addition, receiver operating characteristic (ROC) curve analysis revealed the value of ITGB5 in predicting DFS, and univariate and multivariate analyses showed that ITGB5 may be an independent prognostic factor for DFS. GO and KEGG analyses indicated that many GO terms related to the extracellular matrix (ECM), focal adhesion and ECM-receptor interaction pathways were enriched. GSEA revealed focal adhesion, cancer pathways, ECM-receptor interactions and Wnt signaling pathways in the samples with high ITGB5 expression. Correlation analysis revealed that high ITGB5 expression is significantly correlated with the TGF-β/EMT pathway and WNT targets. Silencing of ITGB5 inhibited SW480 and RKO cell proliferation, invasion and migration. Mechanistically, downregulated ITGB5 expression blocked the Wnt/β-catenin signaling pathway and epithelial-mesenchymal transition (EMT) in CRC cells. Moreover, ITGB5 expression was related to M0 macrophages, M2 macrophages, neutrophils and plasma cell fractions. ITGB5 may be associated with poor prognosis and metastasis in patients with CRC. ITGB5 may hold promise as a prognostic biomarker and a new potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China
| | - Yuan Fang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
| | - Shuwu Zhong
- Intensive Care Unit (ICU), The Second Affiliated Hospital of University of South China, No. 35 Jiefang Avenue, Zhengxiang District, Hengyang, 421001, Hunan, China
| | - Suyong Lin
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China
| | - Xiaoyu Yang
- School of Basic Medicine Sciences, Fujian Medical University, No. 1, Xuefu North Road, Minhou County, Fuzhou, 350122, China.
| | - Shaoqin Chen
- Department of Gastrointestinal Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, No. 66, Jintang Road, Jianxin Town, Cangshan District, Fuzhou, 350002, Fujian, China.
| |
Collapse
|
9
|
Kawahara R, Kautto L, Bansal N, Dipta P, Chau TH, Liquet-Weiland B, Ahn SB, Thaysen-Andersen M. HEXB Drives Raised Paucimannosylation in Colorectal Cancer and Stratifies Patient Risk. Mol Cell Proteomics 2025; 24:100927. [PMID: 39947398 PMCID: PMC11932691 DOI: 10.1016/j.mcpro.2025.100927] [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: 10/11/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 03/28/2025] Open
Abstract
Noninvasive prognostic markers are needed to improve the survival of colorectal cancer (CRC) patients. Toward this goal, we applied untargeted systems glycobiology approaches to snap-frozen and formalin-fixed paraffin-embedded tumor tissues and peripheral blood mononuclear cells from CRC patients spanning different disease stages and matching controls to faithfully uncover molecular changes associated with CRC. Quantitative glycomics and immunohistochemistry revealed that noncanonical paucimannosidic N-glycans are elevated in CRC tumors relative to normal adjacent tissues. Cell origin-focused glycoproteomics enabled using the well-curated Human Protein Atlas combined with immunohistochemistry of CRC tumor tissues recapitulated these findings and indicated that the paucimannosidic proteins were in part from tumor-infiltrating monocytes (e.g., MPO, AZU1) and of CRC cell origin (e.g., LGALS3BP, PSAP). Biosynthetically explaining these observations, N-acetyl-β-D-hexosaminidase (Hex) subunit β (HEXB) was found to be overexpressed in CRC tissues relative to normal adjacent colorectal tissues and colocalization and enzyme inhibition studies confirmed that HEXB facilitates paucimannosidic protein biosynthesis in CRC cells. Employing a sensitive, quick, and robust enzyme activity assay, we then showed that Hex activity was elevated in plasma and peripheral blood mononuclear cells from patients with advanced CRC relative to controls and those with early-stage disease. Surveying a large donor cohort, the plasma Hex activity was found to be raised in CRC patients relative to normal controls and correlated with the 5-year survival of CRC patients indicating that elevated plasma Hex activity is a potential disease risk marker for patient outcome. Our glycoproteomics-driven findings open avenues for better prognostication and disease risk stratification in CRC.
Collapse
Affiliation(s)
- Rebeca Kawahara
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, Japan.
| | - Liisa Kautto
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Naaz Bansal
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Priya Dipta
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - The Huong Chau
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Benoit Liquet-Weiland
- School of Mathematical and Physical Sciences, Macquarie University, Sydney, New South Wales, Australia; Université de Pau et Pays de L'Adour, Laboratoire de Mathématiques et de leurs Applications de PAU, CNRS, E2S-UPPA, Pau, France
| | - Seong Beom Ahn
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Morten Thaysen-Andersen
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi, Japan.
| |
Collapse
|
10
|
Li Z, Deng L, Cheng M, Ye X, Yang N, Fan Z, Sun L. Emerging role of bile acids in colorectal liver metastasis: From molecular mechanism to clinical significance (Review). Int J Oncol 2025; 66:24. [PMID: 39981904 PMCID: PMC11844338 DOI: 10.3892/ijo.2025.5730] [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: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
Liver metastasis is the leading cause of colorectal cancer (CRC)‑related mortality. Microbiota dysbiosis serves a role in the pathogenesis of colorectal liver metastases. Bile acids (BAs), cholesterol metabolites synthesized by intestinal bacteria, contribute to the metastatic cascade of CRC, encompassing colorectal invasion, migration, angiogenesis, anoikis resistance and the establishment of a hepatic pre‑metastatic niche. BAs impact inflammation and modulate the immune landscape within the tumor microenvironment by activating signaling pathways, which are used by tumor cells to facilitate metastasis. Given the widespread distribution of BA‑activated receptors in both tumor and immune cells, strategies aimed at restoring BA homeostasis and blocking metastasis‑associated signaling are of importance in cancer therapy. The present study summarizes the specific role of BAs in each step of colorectal liver metastasis, elucidating the association between BA and CRC progression to highlight the potential of BAs as predictive biomarkers for colorectal liver metastasis and their therapeutic potential in developing novel treatment strategies.
Collapse
Affiliation(s)
- Zhaoyu Li
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, P.R. China
| | - Lingjun Deng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Mengting Cheng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Xiandong Ye
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China, P.R. China
| | - Zaiwen Fan
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| | - Li Sun
- Department of Oncology, Air Force Medical Center of People's Liberation Army, Air Force Medical University, Beijing 100010, P.R. China
| |
Collapse
|
11
|
Khizar H, Ali K, Wang J. From silent partners to potential therapeutic targets: macrophages in colorectal cancer. Cancer Immunol Immunother 2025; 74:121. [PMID: 39998578 PMCID: PMC11861851 DOI: 10.1007/s00262-025-03965-w] [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: 11/15/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Cancer cells grow and survive in the tumor microenvironment, which is a complicated process. As a key part of how colorectal cancer (CRC) progresses, tumor-associated macrophages (TAMs) exhibit a double role. Through angiogenesis, this TAM can promote the growth of cancers. Although being able to modify and adjust immune cells is a great advantage, these cells can also exhibit anti-cancer properties including direct killing of cancer cells, presenting antigens, and aiding T cell-mediated responses. The delicate regulatory mechanisms between the immune system and tumors are composed of a complex network of pathways regulated by several factors including hypoxia, metabolic reprogramming, cytokine/chemokine signaling, and cell interactions. Decoding and figuring out these complex systems become significant in building targeted treatment programs. Targeting TAMs in CRC involves disrupting chemokine signaling or adhesion molecules, reprogramming them to an anti-tumor phenotype using TLR agonists, CD40 agonists, or metabolic modulation, and selectively removing TAM subsets that promote tumor growth. Multi-drug resistance, the absence of an accurate biomarker, and drug non-specificity are also major problems. Combining macrophage-targeted therapies with chemotherapy and immunotherapy may revolutionize treatment. Macrophage studies will advance with new technology and multi-omics methodologies to help us understand CRC and build specific and efficient treatments.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Kamran Ali
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou, 310009, China.
| |
Collapse
|
12
|
Tozzi M, Fiore A, Travaglione S, Marcon F, Rainaldi G, Germinario EAP, Laterza I, Donati S, Macchia D, Spada M, Leoni O, Quattrini MC, Pietraforte D, Tomasoni S, Torrigiani F, Verin R, Matarrese P, Gambardella L, Spadaro F, Carollo M, Pietrantoni A, Carlini F, Panebianco C, Pazienza V, Colella F, Lucchetti D, Sgambato A, Sistigu A, Moschella F, Guidotti M, Vincentini O, Maroccia Z, Biffoni M, De Angelis R, Bracci L, Fabbri A. E. Coli cytotoxic necrotizing factor-1 promotes colorectal carcinogenesis by causing oxidative stress, DNA damage and intestinal permeability alteration. J Exp Clin Cancer Res 2025; 44:29. [PMID: 39876002 PMCID: PMC11776187 DOI: 10.1186/s13046-024-03271-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: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Bacterial toxins are emerging as promising hallmarks of colorectal cancer (CRC) pathogenesis. In particular, Cytotoxic Necrotizing Factor 1 (CNF1) from E. coli deserves special consideration due to the significantly higher prevalence of this toxin gene in CRC patients with respect to healthy subjects, and to the numerous tumor-promoting effects that have been ascribed to the toxin in vitro. Despite this evidence, a definitive causal link between CNF1 and CRC was missing. Here we investigated whether CNF1 plays an active role in CRC onset by analyzing pro-carcinogenic key effects specifically induced by the toxin in vitro and in vivo. METHODS Viability assays, confocal microscopy of γH2AX and 53BP1 molecules and cytogenetic analysis were carried out to assess CNF1-induced genotoxicity on non-neoplastic intestinal epithelial cells. Caco-2 monolayers and 3D Caco-2 spheroids were used to evaluate permeability alterations specifically induced by CNF1, either in the presence or in the absence of inflammation. In vivo, an inflammatory bowel disease (IBD) model was exploited to evaluate the carcinogenic potential of CNF1. Immunohistochemistry and immunofluorescence stainings of formalin-fixed paraffin-embedded (FFPE) colon tissue were carried out as well as fecal microbiota composition analysis by 16 S rRNA gene sequencing. RESULTS CNF1 induces the release of reactive oxidizing species and chromosomal instability in non-neoplastic intestinal epithelial cells. In addition, CNF1 modifies intestinal permeability by directly altering tight junctions' distribution in 2D Caco-2 monolayers, and by hindering the differentiation of 3D Caco-2 spheroids with an irregular arrangement of these junctions. In vivo, repeated intrarectal administration of CNF1 induces the formation of dysplastic aberrant crypt foci (ACF), and produces the formation of colorectal adenomas in an IBD model. These effects are accompanied by the increased neutrophilic infiltration in colonic tissue, by a mixed pro-inflammatory and anti-inflammatory cytokine milieu, and by the pro-tumoral modulation of the fecal microbiota. CONCLUSIONS Taken together, our results support the hypothesis that the CNF1 toxin from E. coli plays an active role in colorectal carcinogenesis. Altogether, these findings not only add new knowledge to the contribution of bacterial toxins to CRC, but also pave the way to the implementation of current screening programs and preventive strategies.
Collapse
Affiliation(s)
- Michela Tozzi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessia Fiore
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Travaglione
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Marcon
- Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriella Rainaldi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Ilenia Laterza
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Donati
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Spada
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Omar Leoni
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Sofia Tomasoni
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, BCA-University of Padua, Legnaro, PD, Italy
| | - Paola Matarrese
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Maria Carollo
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | | | - Francesca Carlini
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Concetta Panebianco
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, FG, Italy
| | - Filomena Colella
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Donatella Lucchetti
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Sgambato
- Multiplex Spatial Profiling Center, Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Sistigu
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "A. Gemelli" - IRCCS, Rome, Italy
| | - Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Guidotti
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Olimpia Vincentini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Rome, Italy
| | - Zaira Maroccia
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberta De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Bracci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
13
|
Wang Q, Yu M, Zhang S. The characteristics of the tumor immune microenvironment in colorectal cancer with different MSI status and current therapeutic strategies. Front Immunol 2025; 15:1440830. [PMID: 39877377 PMCID: PMC11772360 DOI: 10.3389/fimmu.2024.1440830] [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: 05/30/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Colorectal cancer (CRC) remains a significant cause of cancer-related mortality worldwide. Despite advancements in surgery, chemotherapy, and radiotherapy, the effectiveness of these conventional treatments is limited, particularly in advanced cases. Therefore, transition to novel treatment is urgently needed. Immunotherapy, especially immune checkpoint inhibitors (ICIs), has shown promise in improving outcomes for CRC patients. Notably, patients with deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) tumors often benefit from ICIs, while the majority of CRC cases, which exhibit proficient mismatch repair (pMMR) or microsatellite-stable (MSS) status, generally show resistance to this approach. It is assumed that the MSI phenotype cause some changes in the tumor microenvironment (TME), thus triggering antitumor immunity and leading to response to immunotherapy. Understanding these differences in the TME relative to MSI status is essential for developing more effective therapeutic strategies. This review provides an overview of the TME components in CRC and explores current approaches aimed at enhancing ICI efficacy in MSS CRC.
Collapse
Affiliation(s)
- Qingzhe Wang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Yu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuang Zhang
- Department of Targeting Therapy and Immunology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Masui H, Kawada K, Obama K. Neutrophil and Colorectal Cancer. Int J Mol Sci 2024; 26:6. [PMID: 39795864 PMCID: PMC11720084 DOI: 10.3390/ijms26010006] [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: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Colorectal cancer (CRC) is often associated with metastasis and recurrence and is the leading cause of cancer-related mortality. In the progression of CRC, recent studies have highlighted the critical role of neutrophils, particularly tumor-associated neutrophils (TANs). TANs have both tumor-promoting and tumor-suppressing activities, contributing to metastasis, immunosuppression, angiogenesis, and epithelial-to-mesenchymal transition. Tumor-promoting TANs promote tumor growth by releasing proteases, reactive oxygen species, and cytokines, whereas tumor-suppressing TANs enhance immune responses by activating T cells and natural killer cells. Understanding the mechanisms underlying TAN mobilization, plasticity, and their role in the tumor microenvironment has revealed potential therapeutic targets. This review provides a comprehensive overview of TAN biology in CRC and discusses both the tumor-promoting and tumor-suppressing functions of neutrophils. Novel therapeutic approaches targeting TANs, such as chemokine receptor antagonists, aim to modulate neutrophil reprogramming and offer promising avenues for improving treatment outcomes of CRC.
Collapse
Affiliation(s)
- Hideyuki Masui
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
- Department of Surgery, Hirakata Kohsai Hospital, Osaka 573-0153, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
- Department of Surgery, Kurashiki Central Hospital, Okayama 710-8602, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (H.M.); (K.O.)
| |
Collapse
|
15
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
16
|
Koržinek M, Ćelap I, Fabijanec M, Žanić T, Ljubičić N, Baršić N, Verbanac D, Barišić K, Rajković MG. Complete blood count parameters and inflammation-related biomarkers in patients with colorectal carcinoma. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2024; 74:739-749. [PMID: 39560441 DOI: 10.2478/acph-2024-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/14/2024] [Indexed: 11/20/2024]
Abstract
The aim of this study was to determine whether there are differences in complete blood count parameters (CBC) and inflammation-related biomarkers, MPV/PC, PLR, NLR, LWR, LMR, NMR, and LCR, among patients with colorectal carcinoma (CRC) and patients with colorectal adenomas. The study included 155 patients who were divided into two groups according to histopathological analysis - 74 adenomas patients and 81 CRC patients. A routine examination of CBC was conducted on Sysmex XN1000 whereas CRP was measured on Alinity ci-series. Statistical analysis was performed by ROC curve analysis using MedCalc Statistical Software. In CRC patients, hemoglobin concentration, hematocrit, MCV, MCH, and MCHC were lower, while RDW was higher (p < 0.001), compared to patients with adenomas. Total leukocyte count (p = 0 .006), absolute neutrophils (p = 0.005), and absolute monocytes (p = 0.007) were lower while relative eosinophils (p = 0.001) and relative basophils (p = 0.001) were higher in CRC patients. Platelet count (p < 0.001) was significantly higher and MPV (p = 0.003) was significantly lower in CRC patients. Furthermore, MPV/PC (p < 0.001) was significantly lower and PLR (p < 0.001) was significantly higher in CRC. Moreover, Receiver Operating Characteristic (ROC) analysis revealed poor diagnostic accuracy, for all tested parameters (AUC was 0.7 or less). PC, MPV, MPV/PC, and PLR were significantly different between study groups, but ROC analysis revealed poor diagnostic accuracy. Lower hemo globin levels in CRC patients are possibly due to more frequent and excessive bleeding. Higher levels of basophils and eosinophils in CRC patients are indicators of inflammatory reaction, which is linked to CRC.
Collapse
Affiliation(s)
- Martha Koržinek
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| | - Ivana Ćelap
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
- Clinical Hospital Centre Sestre milosrdnice, Department of Clinical Chemistry, 10000 Zagreb, Croatia
| | - Marija Fabijanec
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| | - Tena Žanić
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| | - Neven Ljubičić
- Clinical Hospital Centre Sestre milosrdnice, Department of Gastroenterology and Hepatology 10000 Zagreb, Croatia
| | - Neven Baršić
- Clinical Hospital Centre Sestre milosrdnice, Department of Gastroenterology and Hepatology 10000 Zagreb, Croatia
| | - Donatella Verbanac
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| | - Karmela Barišić
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| | - Marija Grdić Rajković
- University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medical Biochemistry and Hematology 10000 Zagreb, Croatia
| |
Collapse
|
17
|
Mi X, Duan Y, Sun J, Tai Q, Yao H, Meng L, Yang X, Shi X, Shi B, Chen J, Sun L, Zhou D, Xiao S, Yao Y, He S. The ketogenic diet modulates tumor-associated neutrophil polarization via the AMOT-YAP/TAZ axis to inhibit colorectal cancer progression. Pharmacol Res 2024; 210:107494. [PMID: 39510146 DOI: 10.1016/j.phrs.2024.107494] [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: 08/01/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Despite significant advances in the diagnosis and treatment of colorectal cancer (CRC), the prognosis for late-stage patients remains poor, highlighting the urgent need for new preventive and therapeutic strategies. Recent studies have focused on the ketogenic diet (KD) and its metabolite, β-hydroxybutyrate (BHB), for their tumor-suppressive effects and modulation of inflammatory responses. Using the azoxymethane (AOM) / dextran sulfate sodium (DSS)-induced mouse CRC model, we found that the ketogenic diet and BHB inhibit pro-tumor N2-type tumor-associated neutrophils (TANs) while promoting the polarization of TANs towards the anti-tumor N1 type. This shift in TANs polarization affects tumor growth and metastasis. The underlying mechanism involves BHB acting on the intracellular receptor histone deacetylases 3 (HDAC3), which modulates the activation of the AMOT-YAP/TAZ axis, leading to the inhibition of pro-carcinogenic factor transcription and release. Moreover, clinical cohort data corroborate these findings, showing that CRC patients with elevated BHB levels have significantly lower rates of lymph node involvement, which is associated with a higher infiltration ratio of anti-carcinogenic N1-type TANs in the tumor microenvironment (TME). These results suggest that BHB levels could serve as a prognostic biomarker for CRC. In conclusion, our findings indicate that BHB derived from KD regulates TANs polarization in CRC via the HDAC3-AMOT-YAP/TAZ axis, effectively inhibiting tumor growth and metastasis. These insights establish a novel theoretical basis for employing the KD in the treatment of CRC and for developing cancer adjuvant immunotherapy strategy based on the polarization of neutrophils.
Collapse
Affiliation(s)
- Xiuwei Mi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Yudong Duan
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiying Sun
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China; Department of Respiratory Diseases, Children's Hospital of Soochow University, Suzhou, Jiangsu 215025, China
| | - Qingliang Tai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Lijun Meng
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xiaoshan Yang
- Advanced Molecular Pathology Institute of Soochow University and SANO, & SANO Medical Laboratories Suzhou, Jiangsu 215000, China
| | - Xinyu Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Bo Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Junjie Chen
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215299, China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
18
|
Zeng J, Chen H, Liu X, Xia H, Chen L, Lin D, Wang N, Weng C, Guan G, Zheng Y. Cuproptosis in microsatellite stable colon cancer cells affects the cytotoxicity of CD8 +T through the WNT signaling pathway. Chem Biol Interact 2024; 403:111239. [PMID: 39306268 DOI: 10.1016/j.cbi.2024.111239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 08/29/2024] [Accepted: 09/09/2024] [Indexed: 09/29/2024]
Abstract
The microsatellite stable (MSS) colon cancer (CC) has long been considered resistant to immunotherapy. Cuproptosis, as a novel form of cell death, may interact with tumor immunity. This project focused on the impact of cuproptosis on the cytotoxicity of CD8+T in MSS CC, aiming to provide effective clues for improving the treatment strategy of MSS CC. The study developed an MSS CC cuproptosis model using 50 nM elesclomol and 1 μM CuCl2. Cuproptotic SW480 cells were directly co-cultured with CD8+ T cells. Cuproptosis levels were assessed via intracellular copper ion detection, Western blot, and confocal laser scanning microscopy. CCK-8, Hochest/PI staining, CFSE cell proliferation assay, LDH cytotoxicity detection, and ELISA were used to evaluate CD8+ T cell immune activity and cytotoxicity. Transcriptome sequencing and bioinformatics analysis identified regulated signals in cuproptotic SW480 cells. A rescue experiment utilized a WNT pathway activator (BML-284). PD-L1 expression in cells/membranes was analyzed using qRT-PCR, Western blot, and flow cytometry. NSG mice were immunoreconstituted, and the effects of cuproptosis on immune infiltration and cancer progression in MSS CC mice were assessed using ELISA and immunohistochemistry (IHC). Treatment with 50 nM elesclomol and 1 μM CuCl2 significantly increased cuproptosis in SW480 cells. Co-culture with CD8+ T cells enhanced their cytotoxicity. Sequencing revealed cuproptosis-mediated modulation of immune and inflammatory pathways, including WNT signaling. Rescue experiments showed downregulation of WNT signaling in cuproptotic SW480 cells. Indirectly, CD8+ T cell immune function was enhanced by reducing PD-L1 expression. In mice, cuproptosis resulted in increased infiltration of CD8+ T cells in tumor tissue, leading to delayed cancer progression compared to the control group. Cuproptosis in MSS CC cells enhances the cytotoxicity of CD8+ T cells, which may be achieved through downregulation of the WNT signaling pathway and decreased expression of PD-L1. In the future, drugs that can induce cuproptosis may be a promising approach to improve MSS CC immunotherapy.
Collapse
Affiliation(s)
- Jintao Zeng
- Department of Colorectal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, China; Department of Colorectal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China; Fujian Abdominal Surgery Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China
| | - Hong Chen
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xing Liu
- Department of Colorectal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, China
| | - Haoyun Xia
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Liqi Chen
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Dajia Lin
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Naisen Wang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Chong Weng
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Guoxian Guan
- Department of Colorectal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, China; Department of Colorectal Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China; Fujian Abdominal Surgery Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350001, China.
| | - Yu Zheng
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
19
|
Flory M, Bravo P, Alam A. Impact of gut microbiota and its metabolites on immunometabolism in colorectal cancer. IMMUNOMETABOLISM (COBHAM, SURREY) 2024; 6:e00050. [PMID: 39624362 PMCID: PMC11608621 DOI: 10.1097/in9.0000000000000050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/17/2024] [Indexed: 01/25/2025]
Abstract
Colorectal cancer (CRC) is highly prevalent, accounting for approximately one-tenth of cancer cases and deaths globally. It stands as the second most deadly and third most common cancer type. Although the gut microbiota has been implicated in CRC carcinogenesis for the last several decades, it remains one of the least understood risk factors for CRC development, as the gut microbiota is highly diverse and variable. Many studies have uncovered unique microbial signatures in CRC patients compared with healthy matched controls, with variations dependent on patient age, disease stage, and location. In addition, mechanistic studies revealed that tumor-associated bacteria produce diverse metabolites, proteins, and macromolecules during tumor development and progression in the colon, which impact both cancer cells and immune cells. Here, we summarize microbiota's role in tumor development and progression, then we discuss how the metabolic alterations in CRC tumor cells, immune cells, and the tumor microenvironment result in the reprogramming of activation, differentiation, functions, and phenotypes of immune cells within the tumor. Tumor-associated microbiota also undergoes metabolic adaptation to survive within the tumor environment, leading to immune evasion, accumulation of mutations, and impairment of immune cells. Finally, we conclude with a discussion on the interplay between gut microbiota, immunometabolism, and CRC, highlighting a complex interaction that influences cancer development, progression, and cancer therapy efficacy.
Collapse
Affiliation(s)
- Madison Flory
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Paloma Bravo
- Department of Biology, Carleton College, Northfield, MN, USA
| | - Ashfaqul Alam
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
20
|
Salesse L, Duval A, Sauvanet P, Da Silva A, Barnich N, Godfraind C, Dalmasso G, Nguyen HTT. ATG16L1 in myeloid cells limits colorectal tumor growth in ApcMin/+ mice infected with colibactin-producing Escherichia coli via decreasing inflammasome activation. Autophagy 2024; 20:2186-2204. [PMID: 38818900 PMCID: PMC11423662 DOI: 10.1080/15548627.2024.2359770] [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: 11/06/2023] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Escherichia coli strains producing the genotoxin colibactin, designated as CoPEC (colibactin-producing E. coli), have emerged as an important player in the etiology of colorectal cancer (CRC). Here, we investigated the role of macroautophagy/autophagy in myeloid cells, an important component of the tumor microenvironment, in the tumorigenesis of a susceptible mouse model infected with CoPEC. For that, a preclinical mouse model of CRC, the ApcMin/+ mice, with Atg16l1 deficiency specifically in myeloid cells (ApcMin/+/Atg16l1[∆MC]) and the corresponding control mice (ApcMin/+), were infected with a clinical CoPEC strain 11G5 or its isogenic mutant 11G5∆clbQ that does not produce colibactin. We showed that myeloid cell-specific Atg16l1 deficiency led to an increase in the volume of colonic tumors in ApcMin/+ mice under infection with 11G5, but not with 11G5∆clbQ. This was accompanied by increased colonocyte proliferation, enhanced inflammasome activation and IL1B/IL-1β secretion, increased neutrophil number and decreased total T cell and cytotoxic CD8+ T cell numbers in the colonic mucosa and tumors. In bone marrow-derived macrophages (BMDMs), compared to uninfected and 11G5∆clbQ-infected conditions, 11G5 infection increased inflammasome activation and IL1B secretion, and this was further enhanced by autophagy deficiency. These data indicate that ATG16L1 in myeloid cells was necessary to inhibit colonic tumor growth in CoPEC-infected ApcMin/+ mice via inhibiting colibactin-induced inflammasome activation and modulating immune cell response in the tumor microenvironment. Abbreviation: AOM, azoxymethane; APC, APC regulator of WNT signaling pathway; ATG, autophagy related; Atg16l1[∆MC] mice, mice deficient for Atg16l1 specifically in myeloid cells; CASP1, caspase 1; BMDM, bone marrow-derived macrophage; CFU, colony-forming unit; CoPEC, colibactin-producing Escherichia coli; CRC, colorectal cancer; CXCL1/KC, C-X-C motif chemokine ligand 1; ELISA, enzyme-linked immunosorbent assay; IL, interleukin; MC, myeloid cell; MOI, multiplicity of infection; PBS, phosphate-buffered saline; pks, polyketide synthase; qRT-PCR, quantitative real-time reverse-transcription polymerase chain reaction; siRNA, small interfering RNA; TME, tumor microenvironment; TNF/TNF-α, tumor necrosis factor.
Collapse
Affiliation(s)
- Laurène Salesse
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| | - Angéline Duval
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| | - Pierre Sauvanet
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
- Department of Digestive and Hepatobiliary Surgery, CHU, Clermont-Ferrand, France
| | - Alison Da Silva
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| | - Nicolas Barnich
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| | - Catherine Godfraind
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
- Department of Pathology, CHU Gabriel Montpied, Clermont-Ferrand, France
| | - Guillaume Dalmasso
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| | - Hang Thi Thu Nguyen
- M2iSH, UMR 1071 Inserm, University of Clermont Auvergne, INRAE USC 1382, CRNH, Clermont-Ferrand, France
| |
Collapse
|
21
|
Garcia-Flores LA, Dawid De Vera MT, Pilo J, Rego A, Gomez-Casado G, Arranz-Salas I, Hierro Martín I, Alcaide J, Torres E, Ortega-Gomez A, Boughanem H, Macias-Gonzalez M. Increased neutrophil counts are associated with poor overall survival in patients with colorectal cancer: a five-year retrospective analysis. Front Immunol 2024; 15:1415804. [PMID: 39376564 PMCID: PMC11456424 DOI: 10.3389/fimmu.2024.1415804] [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: 04/11/2024] [Accepted: 08/30/2024] [Indexed: 10/09/2024] Open
Abstract
Background Colorectal cancer (CRC) continues to be a major health concern in today's world. Despite conflictive findings, evidence supports systemic inflammation's impact on CRC patients' survival rates. Therefore, this study aims to assess the prognostic role of the innate immune system in patients with CRC. Method A total of 449 patients were included, with a 5-year follow-up period, and absolute neutrophil counts and their related ratios were measured. Results The non-survival group had increased levels of white blood cells, neutrophils (both p<0.001), and monocytes (p=0.038), compared to the survival group, along with other neutrophil-related ratios. We observed increased mortality risk in patients in the highest tertile of white blood cells [HR=1.85 (1.09-3.13), p<0.05], neutrophils [HR=1.78 (95% CI: 1.07-2.96), p<0.05], and monocytes [HR=2.11 (95% CI: 1.22-3.63)], compared to the lowest tertile, after adjusting for all clinicopathological variables. Random forest analysis identified neutrophils as the most crucial variable in predicting survival rates, having an AUC of 0.712, considering all clinicopathological variables. A positive relationship between neutrophil counts and metastasis was observed when neutrophil counts are considered continuous (β=0.92 (0.41), p<0.05) and tumor size (width) when neutrophils were considered as logistic variable (T1 vs T3) [OR=1.42, (95% CI: 1.05-1.98), p<0.05]. Conclusion This study offers comprehensive insights into the immune factors that impact the prognosis of CRC, emphasizing the need for personalized prognostic tools.
Collapse
Affiliation(s)
- Libia Alejandra Garcia-Flores
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
| | - María Teresa Dawid De Vera
- Unidad de Gestión Clínica Intercentros (UGCI) de Anatomía Patológica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Jesus Pilo
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
| | - Alejandro Rego
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
| | - Gema Gomez-Casado
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
| | - Isabel Arranz-Salas
- Unidad de Gestión Clínica Intercentros (UGCI) de Anatomía Patológica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Isabel Hierro Martín
- Unidad de Gestión Clínica Intercentros (UGCI) de Anatomía Patológica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Julia Alcaide
- Medical Oncology Service, Hospital Regional Universitario de Málaga, Biomedical Research Institute of Malaga (IBIMA), Málaga, Spain
| | - Esperanza Torres
- Unidad de Gestión Clínica Intercentros (UGCI) de Oncología Médica, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Almudena Ortega-Gomez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Hatim Boughanem
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofia, Cordoba, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba, Spain
| | - Manuel Macias-Gonzalez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain
- Institute of Biomedical Research in Malaga (IBIMA)-Bionand Platform, University of Malaga, Málaga, Spain
- Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Cañellas-Socias A, Sancho E, Batlle E. Mechanisms of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol 2024; 21:609-625. [PMID: 38806657 DOI: 10.1038/s41575-024-00934-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
Despite extensive research and improvements in understanding colorectal cancer (CRC), its metastatic form continues to pose a substantial challenge, primarily owing to limited therapeutic options and a poor prognosis. This Review addresses the emerging focus on metastatic CRC (mCRC), which has historically been under-studied compared with primary CRC despite its lethality. We delve into two crucial aspects: the molecular and cellular determinants facilitating CRC metastasis and the principles guiding the evolution of metastatic disease. Initially, we examine the genetic alterations integral to CRC metastasis, connecting them to clinically marked characteristics of advanced CRC. Subsequently, we scrutinize the role of cellular heterogeneity and plasticity in metastatic spread and therapy resistance. Finally, we explore how the tumour microenvironment influences metastatic disease, emphasizing the effect of stromal gene programmes and the immune context. The ongoing research in these fields holds immense importance, as its future implications are projected to revolutionize the treatment of patients with mCRC, hopefully offering a promising outlook for their survival.
Collapse
Affiliation(s)
- Adrià Cañellas-Socias
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Elena Sancho
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
23
|
Kromidas E, Geier A, Weghofer A, Liu HY, Weiss M, Loskill P. Immunocompetent PDMS-Free Organ-on-Chip Model of Cervical Cancer Integrating Patient-Specific Cervical Fibroblasts and Neutrophils. Adv Healthc Mater 2024; 13:e2302714. [PMID: 38029413 DOI: 10.1002/adhm.202302714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/04/2023] [Indexed: 12/01/2023]
Abstract
Despite preventive measures and available treatments, cervical cancer still ranks as the fourth most prevalent cancer among women worldwide and remains the leading cause of cancer death in women in many developing countries. To gain further insights into pathogenesis and to develop novel (immuno)therapies, more sophisticated human models recreating patient heterogeneities and including aspects of the tumor microenvironment are urgently required. A novel polydimethylsiloxane-free microfluidic platform, designed specifically for the generation and ccultivation of cervical cancerous tissue, is introduced. The microscale open-top tissue chambers of the cervical cancer-on-chip (CCoC) enable facile generation and long-term cultivation of SiHa spheroids in co-culture with donor-derived cervical fibroblasts. The resulting 3D tissue emulates physiological architecture and allows dissection of distinct effects of the stromal tissue on cancer viability and growth. Treatment with cisplatin at clinically-relevant routes of administration and dosing highlights the platform's applicability for drug testing. Moreover, the model is amenable for integration and recruitment of donor-derived neutrophils from the microvasculature-like channel into the tissue, all while retaining their ability to produce neutrophil extracellular traps. In the future, the immunocompetent CCoC featuring donor-specific primary cells and tumor spheroids has the potential to contribute to the development of new (immuno)therapeutic options.
Collapse
Affiliation(s)
- Elena Kromidas
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Alicia Geier
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Adrian Weghofer
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Hui-Yu Liu
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
| | - Martin Weiss
- Department for Biomedicine and Materials Science, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- Department for Women's Health, Faculty of Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, 72074, Tübingen, Germany
- Department for Biomedicine and Materials Science, NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770, Reutlingen, Germany
- 3R Center Tübingen for In Vitro Models and Alternatives to Animal Testing, 72074, Tübingen, Germany
| |
Collapse
|
24
|
Burgos-Molina AM, Téllez Santana T, Redondo M, Bravo Romero MJ. The Crucial Role of Inflammation and the Immune System in Colorectal Cancer Carcinogenesis: A Comprehensive Perspective. Int J Mol Sci 2024; 25:6188. [PMID: 38892375 PMCID: PMC11172443 DOI: 10.3390/ijms25116188] [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: 05/04/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic inflammation drives the growth of colorectal cancer through the dysregulation of molecular pathways within the immune system. Infiltration of immune cells, such as macrophages, into tumoral regions results in the release of proinflammatory cytokines (IL-6; IL-17; TNF-α), fostering tumor proliferation, survival, and invasion. Tumors employ various mechanisms to evade immune surveillance, effectively 'cloaking' themselves from detection and subsequent attack. A comprehensive understanding of these intricate molecular interactions is paramount for advancing novel strategies aimed at modulating the immune response against cancer.
Collapse
Affiliation(s)
- Antonio Manuel Burgos-Molina
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
| | - Teresa Téllez Santana
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
- Research Network on Chronic Diseases, Primary Care, and Health Promotion (RICAPPS), Carlos III Health Institute (Instituto de Salud Carlos III), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- Málaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28, 29010 Málaga, Spain
| | - Maximino Redondo
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
- Research Network on Chronic Diseases, Primary Care, and Health Promotion (RICAPPS), Carlos III Health Institute (Instituto de Salud Carlos III), Av. de Monforte de Lemos, 5, 28029 Madrid, Spain
- Málaga Biomedical Research Institute (Instituto de Investigación Biomédica de Málaga, IBIMA), Calle Doctor Miguel Díaz Recio, 28, 29010 Málaga, Spain
- Research Unit, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Spain
| | - María José Bravo Romero
- Surgery, Biochemistry and Immunology Department, School of Medicine, University of Malaga, 29010 Málaga, Spain; (A.M.B.-M.); (T.T.S.); (M.J.B.R.)
| |
Collapse
|
25
|
Lopez-Perez D, Prados-Lopez B, Galvez J, Leon J, Carazo A. Eosinophils in Colorectal Cancer: Emerging Insights into Anti-Tumoral Mechanisms and Clinical Implications. Int J Mol Sci 2024; 25:6098. [PMID: 38892286 PMCID: PMC11172675 DOI: 10.3390/ijms25116098] [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] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Eosinophils are myeloid effector cells whose main homing is the gastrointestinal tract. There, they take part in type I and type II immune responses. They also contribute to other non-immunological homeostatic functions like mucus production, tissue regeneration, and angiogenesis. In colorectal cancer (CRC), eosinophils locate in the center of the tumor and in the front of invasion and play an anti-tumoral role. They directly kill tumor cells by releasing cytotoxic compounds and eosinophil extracellular traps or indirectly by activating other immune cells via cytokines. As CRC progresses, the number of infiltrating eosinophils decreases. Although this phenomenon is not fully understood, it is known that some changes in the microenvironmental milieu and microbiome can affect eosinophil infiltration. Importantly, a high number of intratumoral eosinophils is a favorable prognostic factor independent from the tumor stage. Moreover, after immunotherapy, responding patients usually display eosinophilia, so eosinophils could be a good biomarker candidate to monitor treatment outcomes. Finally, even though eosinophils seem to play an interesting anti-tumoral role in CRC, much more research is needed to fully understand their interactions in the CRC microenvironment. This review explores the multifaceted roles of eosinophils in colorectal cancer, highlighting their anti-tumoral effects, prognostic significance, and potential as a biomarker for treatment outcomes.
Collapse
Affiliation(s)
- David Lopez-Perez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Belen Prados-Lopez
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
| | - Julio Galvez
- Department of Pharmacology, Faculty of Pharmacy, University of Granada, 18012 Granada, Spain
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Centro de Investigación Biomédica en Red para Enfermedades Hepáticas y Digestivas (CIBER-EHD), Center for Biomedical Research, University of Granada, 18012 Granada, Spain
| | - Josefa Leon
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión Clínica de Aparato Digestivo, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| | - Angel Carazo
- Research Unit, Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18016 Granada, Spain
- Unidad de Gestión de Microbiología, Hospital Universitario San Cecilio de Granada, 18016 Granada, Spain
| |
Collapse
|
26
|
Ming L, Tang J, Qin F, Qin Y, Wang D, Huang L, Cao Y, Huang Z, Yin Y. Exosome secretion related gene signature predicts chemoresistance in patients with colorectal cancer. Pathol Res Pract 2024; 257:155313. [PMID: 38642509 DOI: 10.1016/j.prp.2024.155313] [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: 11/06/2023] [Revised: 03/14/2024] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a highly heterogeneous malignancy, and patients often have different responses to treatment. In this study, the genetic characteristics related to exosome formation and secretion procedure were used to predict chemoresistance and guide the individualized treatment of patients. METHODS Firstly, seven microarray datasets in Gene Expression Omnibus (GEO) and RNA-Seq dataset from the Cancer Genome Atlas (TCGA) were used to analysis the transcriptome profiles and associated characteristics of CRC patients. Then, a predictive model based on gene features linked to exosome formation and secretion was created and validated using Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis and Support Vector Machine-Recursive Feature Elimination (SVM-RFE) machine learning. Finally, we evaluated the model using chemoresistant/chemosensitive cells and tissues by immunofluorescence (IF), western blot (WB), quantitative real-time PCR (qRT-PCR) and immunocytochemistry (IHC) experiments, and the predictive value of integrated model in the clinical validation cohort were performed by Receiver Operating Characteristic (ROC) and Kaplan-Meier (K-M) curves analyses. RESULTS We established a risk score signature based on three genes related to exosome secretion in CRC. Better Overall Survival (OS) and greater chemosensitivity were seen in the low-risk group, whereas the high-risk group exhibited chemoresistance and a subpar response to immune checkpoint blockade (ICB) therapy. Higher expression of the model genes EXOC2, EXOC3 and STX4 were observed in chemoresistant cells and specimens. The AUC of 5-year disease-free survival (DFS) was 0.804. Compared with that in the low-risk group, patients' DFS was found to be significantly worse in the high-risk group. CONCLUSIONS In summary, the gene signature related to exosome formation and secretion could reliably predict patients' chemosensitivity and ICB treatment response, which providing new independent biomarkers for the treatment of CRC.
Collapse
Affiliation(s)
- Liang Ming
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Junhui Tang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Feiyu Qin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yan Qin
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Duo Wang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Liuying Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yulin Cao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
27
|
Isermann T, Schneider KL, Wegwitz F, De Oliveira T, Conradi LC, Volk V, Feuerhake F, Papke B, Stintzing S, Mundt B, Kühnel F, Moll UM, Schulz-Heddergott R. Enhancement of colorectal cancer therapy through interruption of the HSF1-HSP90 axis by p53 activation or cell cycle inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581507. [PMID: 38464125 PMCID: PMC10925225 DOI: 10.1101/2024.02.22.581507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The stress-associated molecular chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing hundreds of oncoproteins and disturbing the stoichiometry of protein complexes. Most inhibitors target the key component heat-shock protein 90 (HSP90). However, although classical HSP90 inhibitors are highly tumor-selective, they fail in phase 3 clinical oncology trials. These failures are at least partly due to an interference with a negative feedback loop by HSP90 inhibition, known as heat-shock response (HSR): in response to HSP90 inhibition there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock factor 1 (HSF1). We recently identified that wildtype p53 (p53) actively reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here we test the hypothesis that in HSP90-based therapies simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. Indeed, we find that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids and patient-derived organoids (PDOs). Mechanistically, upon combination therapy human CRC cells strongly upregulate p53-associated pathways, apoptosis, and inflammatory immune pathways. Likewise, in the chemical AOM/DSS CRC model in mice, dual HSF1-HSP90 inhibition strongly represses tumor growth and remodels immune cell composition, yet displays only minor toxicities in mice and normal mucosa-derived organoids. Importantly, inhibition of the cyclin dependent kinases 4 and 6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Even more important, in p53-deficient (mutp53-harboring) CRC cells, an HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR system and reduces cancer growth. Likewise, p53-mutated PDOs strongly respond to dual HSF1-HSP90 pathway inhibition and thus, providing a strategy to target CRC independent of the p53 status. In sum, activating p53 (in p53-proficient cancer cells) or inhibiting CDK4/6 (independent of the p53 status) provide new options to improve the clinical outcome of HSP90-based therapies and to enhance colorectal cancer therapy.
Collapse
Affiliation(s)
- Tamara Isermann
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
- Charité – Universitätsmedizin Berlin, Institute of Pathology, Laboratory of Molecular Tumor Pathology and Systems Biology, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kim Lucia Schneider
- Department of Molecular Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Wegwitz
- Department of Gynecology and Obstetrics, University Medical Center Göttingen, Göttingen, Germany
| | - Tiago De Oliveira
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, Germany
| | - Valery Volk
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | | | - Björn Papke
- Charité – Universitätsmedizin Berlin, Institute of Pathology, Laboratory of Molecular Tumor Pathology and Systems Biology, Berlin, Germany
- German Cancer Consortium (DKTK); Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Stintzing
- Charité – Universitätsmedizin Berlin, Department of Hematology, Oncology, and Cancer Immunology, Berlin, Germany
| | - Bettina Mundt
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ute M. Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY
| | | |
Collapse
|
28
|
Xu M, Zhao X, Wen T, Qu X. Unveiling the role of KRAS in tumor immune microenvironment. Biomed Pharmacother 2024; 171:116058. [PMID: 38171240 DOI: 10.1016/j.biopha.2023.116058] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/03/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Kirsten rats sarcoma viral oncogene (KRAS), the first discovered human oncogene, has long been recognized as "undruggable". KRAS mutations frequently occur in multiple human cancers including non-small cell lung cancer(NSCLC), colorectal cancer(CRC) and pancreatic ductal adenocarcinoma(PDAC), functioning as a "molecule switch" determining the activation of various oncogenic signaling pathways. Except for its intrinsic pro-tumorigenic role, KRAS alteration also exhibits an unique immune signature characterized by elevated PD-L1 level and high tumor mutational burden(TMB). KRAS mutation shape an immune suppressive microenvironment by impeding effective T cells infiltration and recruiting suppressive immune cells including myeloid-derived suppressor cells(MDSCs), regulatory T cells(Tregs), cancer associated fibroblasts(CAFs). In immune checkpoint inhibitor(ICI) era, NSCLC patients with mutated KRAS tend to be more responsive to ICI than patients with intact KRAS. The hallmark for KRAS mutation is the existence of multiple kinds of co-mutations. Different types of co-alterations have distinct tumor microenvironment(TME) signatures and responses to ICI. TP53 co-mutation possess a "hot" TME and achieve higher response to immunotherapy while other loss of function mutation correlated with a "colder" TME and a poor outcome to ICI-based therapy. The groundbreaking discovery of KRAS G12C inhibitors significantly improved outcomes for this KRAS subtype even though efficacy was limited to NSCLC patients. KRAS G12C inhibitors also restore the suppressive TME, creating an opportunity for combinations with ICI. However, an inevitable challenge to KRAS inhibitors is drug resistance. Promising combination strategies such as combination with SHP2 is an approach deserve further exploration because of their immune modulatory effect.
Collapse
Affiliation(s)
- Miao Xu
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China
| | - Xing Zhao
- Department of Pediatrics, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, 155 North Nanjing Street, Shenyang, Liaoning, China; Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Provinces, The First Hospital of China Medical University, Shenyang, Liaoning, China; Clinical Cancer Research Center of Shenyang, the First Hospital of China Medical University, Shenyang, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, Shenyang, Liaoning, China.
| |
Collapse
|
29
|
Yue M, Chen MM, Zhang B, Wang Y, Li P, Zhao Y. The functional roles of chemokines and chemokine receptors in colorectal cancer progression. Biomed Pharmacother 2024; 170:116040. [PMID: 38113624 DOI: 10.1016/j.biopha.2023.116040] [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: 09/27/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Colorectal cancer is a common malignancy with significant rates of morbidity and mortality. A number of factors, including the tumor microenvironment, chemokines, the inflammatory response, have an impact on the development of colorectal cancer. A critical component of the tumor microenvironment is chemokines. Various cell subsets are attracted to the tumor microenvironment through interactions with chemokine receptors. These cells have varying effects on the development of the tumor and the effectiveness of treatment. Additionally, chemokines can participate in inflammatory processes and have effects that are either pro- or anti-tumor. Chemokines can be exploited as targets for medication resistance and treatment in colorectal cancer. In this review, we discuss the expression of chemokines and chemokine receptors, and their relationship with immune cells in the tumor microenvironment. At the same time, we also collect and discuss the significance of chemokines and chemokine receptors in colorectal cancer progression, and their potential as molecular targets for CRC treatment.
Collapse
Affiliation(s)
- Mingli Yue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Meng-Meng Chen
- Key Laboratory of Cancer and Immune Cells of Qingdao, Qingdao 266021, China; Qingdao Restore Biotechnology Co., Ltd., Qingdao, Shandong 266111, PR China
| | - Bingqiang Zhang
- Key Laboratory of Cancer and Immune Cells of Qingdao, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yi Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province affiliated to Qingdao University, Shandong Province, China.
| |
Collapse
|
30
|
Wu J, Dong W, Pan Y, Wang J, Wu M, Yu Y. Crosstalk between gut microbiota and metastasis in colorectal cancer: implication of neutrophil extracellular traps. Front Immunol 2023; 14:1296783. [PMID: 37936694 PMCID: PMC10626548 DOI: 10.3389/fimmu.2023.1296783] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Primary colorectal cancer (CRC) often leads to liver metastasis, possibly due to the formation of pre-metastatic niche (PMN) in liver. Thus, unravelling the key modulator in metastasis is important for the development of clinical therapies. Gut microbiota dysregulation is a key event during CRC progression and metastasis. Numerous studies have elucidated the correlation between specific gut bacteria strains (e.g., pks + E. coli and Bacteroides fragilis) and CRC initiation, and gut bacteria translocation is commonly witnessed during CRC progression. Gut microbiota shapes tumor microenvironment (TME) through direct contact with immune cells or through its functional metabolites. However, how gut microbiota facilitates CRC metastasis remains controversial. Meanwhile, recent studies identify the dissemination of bacteria from gut lumen to liver, suggesting the role of gut microbiota in shaping tumor PMN. A pro-tumoral PMN is characterized by the infiltration of immunosuppressive cells and increased pro-inflammatory immune responses. Notably, neutrophils form web-like structures known as neutrophil extracellular traps (NETs) both in primary TME and metastatic sites, NETs are involved in cancer progression and metastasis. In this review, we focus on the role of gut microbiota in CRC progression and metastasis, highlight the multiple functions of different immune cell types in TME, especially neutrophils and NETs, discuss the possible mechanisms of gut microbiota in shaping PMN formation, and provide therapeutical indications in clinic.
Collapse
Affiliation(s)
- Jiawei Wu
- Department of General Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Wenyan Dong
- Department of General Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yayun Pan
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jingjing Wang
- Department of Burn and Plastic Surgery, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yue Yu
- Department of General Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
31
|
Pan Q, Fan X, Xie L, Wu D, Liu R, Gao W, Luo K, He B, Pu Y. Nano-enabled colorectal cancer therapy. J Control Release 2023; 362:548-564. [PMID: 37683732 DOI: 10.1016/j.jconrel.2023.09.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
Colorectal cancer (CRC), one of the most common and deadliest diseases worldwide, poses a great health threat and social burden. The clinical treatments of CRC encompassing surgery, chemotherapy, and radiotherapy are challenged with toxicity, therapy resistance, and recurrence. In the past two decades, targeted therapy and immunotherapy have greatly improved the therapeutic benefits of CRC patients but they still suffer from drug resistance and low response rates. Very recently, gut microbiota regulation has exhibited a great potential in preventing and treating CRC, as well as in modulating the efficacy and toxicity of chemotherapy and immunotherapy. In this review, we provide a cutting-edge summary of nanomedicine-based treatment in colorectal cancer, highlighting the recent progress of oral and systemic tumor-targeting and/or tumor-activatable drug delivery systems as well as novel therapeutic strategies against CRC, including nano-sensitizing immunotherapy, anti-inflammation, gut microbiota modulation therapy, etc. Finally, the recent endeavors to address therapy resistance, metastasis, and recurrence in CRC were discussed. We hope this review could offer insight into the design and development of nanomedicines for CRC and beyond.
Collapse
Affiliation(s)
- Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Li Xie
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Di Wu
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Rong Liu
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China.
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and molecular imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
32
|
Kong X, Zhang Y, Xiang L, You Y, Duan Y, Zhao Y, Li S, Wu R, Zhang J, Zhou L, Duan L. Fusobacterium nucleatum-triggered neutrophil extracellular traps facilitate colorectal carcinoma progression. J Exp Clin Cancer Res 2023; 42:236. [PMID: 37684625 PMCID: PMC10492297 DOI: 10.1186/s13046-023-02817-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Fusobacterium nucleatum (Fn) acts as a procarcinogenic bacterium in colorectal carcinoma (CRC) by regulating the inflammatory tumor microenvironment (TME). Neutrophil extracellular traps (NETs), which can be generated by persistent inflammation, have been recently considered to be significant contributors in promoting cancer progression. However, whether NETs are implicated in Fn-related carcinogenesis is still poorly characterized. Here, we explored the role of NETs in Fn-related CRC as well as their potential clinical significance. METHODS Fn was measured in tissue specimens and feces samples from CRC patients. The expression of NET markers were also detected in tissue specimens, freshly isolated neutrophils and blood serum from CRC patients, and the correlation of circulating NETs levels with Fn was evaluated. Cell-based experiments were conducted to investigate the mechanism by which Fn modulates NETs formation. In addition, we clarified the functional mechanism of Fn-induced NETs on the growth and metastasis of CRC in vitro and in vivo experiments. RESULTS Tissue and blood samples from CRC patients, particularly those from Fn-infected CRC patients, exhibited greater neutrophil infiltration and higher NETs levels. Fn infection induced abundant NETs production in in vitro studies. Subsequently, we demonstrated that Fn-induced NETs indirectly accelerated malignant tumor growth through angiopoiesis, and facilitated tumor metastasis, as manifested by epithelial-mesenchymal transition (EMT)-related cell migration, matrix metalloproteinase (MMP)-mediated basement membrane protein degradation, and trapping of CRC cells. Mechanistically, the Toll-like receptor (TLR4)-reactive oxygen species (ROS) signaling pathway and NOD-like receptor (NOD1/2)-dependent signaling were responsible for Fn-stimulated NETs formation. More importantly, circulating NETs combined with carcinoembryonic antigen (CEA) could predict CRC occurrence and metastasis, with areas under the ROC curves (AUCs) of 0.92 and 0.85, respectively. CONCLUSIONS Our findings indicated that Fn-induced NETs abundance by activating TLR4-ROS and NOD1/2 signalings in neutrophils facilitated CRC progression. The combination of circulating NETs and CEA was identified as a novel screening strategy for predicting CRC occurrence and metastasis.
Collapse
Affiliation(s)
- Xuehua Kong
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Linwei Xiang
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yan You
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yaqian Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China
| | - Yuqing Zhao
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Shue Li
- Department of Academic Research, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rui Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of Chonqing Medical University, Chongqing, 400016, China
| | - Jiangbo Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Zhou
- Department of Laboratory Medicine, Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Chongqing Medical University, No. 1 of Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Liang Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, No.74 Linjiang Road, Yu Zhong District, Chongqing, 400010, China.
| |
Collapse
|
33
|
Zhang X, Yang L, Deng Y, Huang Z, Huang H, Wu Y, He B, Hu F. Single-cell RNA-Seq and bulk RNA-Seq reveal reliable diagnostic and prognostic biomarkers for CRC. J Cancer Res Clin Oncol 2023; 149:9805-9821. [PMID: 37247080 DOI: 10.1007/s00432-023-04882-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/19/2023] [Indexed: 05/30/2023]
Abstract
PURPOSE The potential role of epithelium-specific genes through the adenoma-carcinoma sequence in the development of colorectal cancer (CRC) remains unknown. Therefore, we integrated single-cell RNA sequencing and bulk RNA sequencing data to select diagnosis and prognosis biomarkers for CRC. METHODS The CRC scRNA-seq dataset was used to describe the cellular landscape of normal intestinal mucosa, adenoma and CRC and to further select epithelium-specific clusters. Differentially expressed genes (DEGs) of epithelium-specific clusters were identified between intestinal lesion and normal mucosa in the scRNA-seq data throughout the adenoma-carcinoma sequence. Diagnostic biomarkers and prognostic biomarker (the risk score) for CRC were selected in the bulk RNA-seq dataset based on DEGs shared by the adenoma epithelium-specific cluster and the CRC epithelium-specific cluster (shared-DEGs). RESULTS Among the 1063 shared-DEGs, we selected 38 gene expression biomarkers and 3 methylation biomarkers that had promising diagnostic power in plasma. Multivariate Cox regression identified 174 shared-DEGs as prognostic genes for CRC. We combined 1000 times LASSO-Cox regression and two-way stepwise regression to select 10 prognostic shared-DEGs to construct the risk score in the CRC meta-dataset. In the external validation dataset, the 1- and 5-year AUCs of the risk score were higher than those of stage, the pyroptosis-related genes (PRG) score and the cuproptosis-related genes (CRG) score. In addition, the risk score was closely associated with the immune infiltration of CRC. CONCLUSION The combined analysis of the scRNA-seq dataset and the bulk RNA-seq dataset in this study provides reliable biomarkers for the diagnosis and prognosis of CRC.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Epidemiology, The School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Longkun Yang
- Department of Epidemiology, The School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Ying Deng
- Department of Epidemiology, The School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Zhicong Huang
- Department of Epidemiology, The School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China
| | - Hao Huang
- Department of Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen University Medical School, Shenzhen, 518061, Guangdong Province, People's Republic of China
| | - Yuying Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, People's Republic of China
| | - Baochang He
- Department of Epidemiology, The School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, People's Republic of China.
| | - Fulan Hu
- Department of Epidemiology, School of Public Health, Shenzhen University Health Science Center, Shenzhen University Medical School, Shenzhen, 518061, Guangdong Province, People's Republic of China.
| |
Collapse
|
34
|
Cheng X, Wang H, Wang Z, Zhu B, Long H. Tumor-associated myeloid cells in cancer immunotherapy. J Hematol Oncol 2023; 16:71. [PMID: 37415162 PMCID: PMC10324139 DOI: 10.1186/s13045-023-01473-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Tumor-associated myeloid cells (TAMCs) are among the most important immune cell populations in the tumor microenvironment, and play a significant role on the efficacy of immune checkpoint blockade. Understanding the origin of TAMCs was found to be the essential to determining their functional heterogeneity and, developing cancer immunotherapy strategies. While myeloid-biased differentiation in the bone marrow has been traditionally considered as the primary source of TAMCs, the abnormal differentiation of splenic hematopoietic stem and progenitor cells, erythroid progenitor cells, and B precursor cells in the spleen, as well as embryo-derived TAMCs, have been depicted as important origins of TAMCs. This review article provides an overview of the literature with a focus on the recent research progress evaluating the heterogeneity of TAMCs origins. Moreover, this review summarizes the major therapeutic strategies targeting TAMCs with heterogeneous sources, shedding light on their implications for cancer antitumor immunotherapies.
Collapse
Affiliation(s)
- Xinyu Cheng
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Huilan Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Zhongyu Wang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| | - Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Chongqing, 400037, China.
| |
Collapse
|
35
|
Wang Y, Liu Y, Zhang Z, Lu B, Gao Y, Tong L, Hu M, Lin PP, Li B, Zhang T. Post-therapeutic circulating tumor cell-associated white blood cell clusters predict poor survival in patients with advanced driver gene-negative non-small cell lung cancer. BMC Cancer 2023; 23:578. [PMID: 37349714 DOI: 10.1186/s12885-023-10985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
PURPOSE This study aimed to investigate the clinical utility of diverse aneuploid circulating tumor cell (CTC) subtypes and particularly CTC-associated white blood cell (CTC-WBC) clusters in predicting treatment response, prognosis and real-time monitoring disease progression in advanced driver gene-negative non-small lung cancer (NSCLC) patients. MATERIALS AND METHODS A total of 74 eligible patients were prospectively enrolled and serial blood samples were collected at pre-treatment(t0), after two cycles of therapy (t1) and at post-four-to-six treatment cycles (t2). Co-detection of diverse subtypes of aneuploid CTCs and CTC-WBC clusters was conducted in advanced NSCLC patients receiving first-line treatment. RESULTS At baseline, CTCs were detected in 69 (93.24%) patients and CTC-WBC clusters were detected in 23 (31.08%) patients. Patients with CTCs < 5/6ml or with CTC-WBC clusters undetectable exhibited a better treatment response than patients with pre-therapeutic aneuploid CTCs ≥ 5/6ml or harboring CTC-WBC clusters (p = 0.034 and p = 0.012, respectively). Before treatment, patients bearing tetraploid CTCs ≥ 1/6ml showed significantly inferior progression-free survival (PFS) [hazard ratio (HR):2.420, 95% confidence interval (CI): 1.426-4.106; p = 0.001] and overall survival (OS) compared to patients with tetraploid CTCs < 1/6ml (HR:1.907, 95%CI: 1.119-3.251; p = 0.018). A longitudinal study demonstrated that post-therapeutic patients harboring CTC-WBC clusters displayed the reduced PFS and OS compared with those without CTC-WBC clusters, and subgroup analysis showed that the presence of CTC-WBC clusters indicated a worse prognosis in both lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) patients. After adjusting for multiple significant factors, post-therapeutic CTC-WBC clusters were the only independent predictor of both PFS (HR:2.872, 95% CI: 1.539-5.368; p = 0.001) and OS (HR:2.162, 95% CI: 1.168-4.003; p = 0.014). CONCLUSIONS In addition to CTCs, longitudinal detection of CTC-WBC clusters provided a feasible tool to indicate initial treatment response, dynamically monitor disease progression and predict survival in driver gene-negative advanced NSCLC patients.
Collapse
Affiliation(s)
- Ying Wang
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yanxia Liu
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Zhiyun Zhang
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
- Department of Cancer Research Center, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Baohua Lu
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yuan Gao
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Li Tong
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Mingming Hu
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | | | - Baolan Li
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Tongmei Zhang
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| |
Collapse
|
36
|
Lucarini V, Nardozi D, Angiolini V, Benvenuto M, Focaccetti C, Carrano R, Besharat ZM, Bei R, Masuelli L. Tumor Microenvironment Remodeling in Gastrointestinal Cancer: Role of miRNAs as Biomarkers of Tumor Invasion. Biomedicines 2023; 11:1761. [PMID: 37371856 DOI: 10.3390/biomedicines11061761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Gastrointestinal (GI) cancers are the most frequent neoplasm, responsible for half of all cancer-related deaths. Metastasis is the leading cause of death from GI cancer; thus, studying the processes that regulate cancer cell migration is of paramount importance for the development of new therapeutic strategies. In this review, we summarize the mechanisms adopted by cancer cells to promote cell migration and the subsequent metastasis formation by highlighting the key role that tumor microenvironment components play in deregulating cellular pathways involved in these processes. We, therefore, provide an overview of the role of different microRNAs in promoting tumor metastasis and their role as potential biomarkers for the prognosis, monitoring, and diagnosis of GI cancer patients. Finally, we relate the possible use of nutraceuticals as a new strategy for targeting numerous microRNAs and different pathways involved in GI tumor invasiveness.
Collapse
Affiliation(s)
- Valeria Lucarini
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Daniela Nardozi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Valentina Angiolini
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
- Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, via di Sant'Alessandro 8, 00131 Rome, Italy
| | - Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Raffaele Carrano
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Zein Mersini Besharat
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
37
|
Giacomelli M, Monti M, Pezzola DC, Lonardi S, Bugatti M, Missale F, Cioncada R, Melocchi L, Giustini V, Villanacci V, Baronchelli C, Manenti S, Imberti L, Giurisato E, Vermi W. Immuno-Contexture and Immune Checkpoint Molecule Expression in Mismatch Repair Proficient Colorectal Carcinoma. Cancers (Basel) 2023; 15:3097. [PMID: 37370706 DOI: 10.3390/cancers15123097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/01/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal carcinoma (CRC) represents a lethal disease with heterogeneous outcomes. Only patients with mismatch repair (MMR) deficient CRC showing microsatellite instability and hyper-mutated tumors can obtain clinical benefits from current immune checkpoint blockades; on the other hand, immune- or target-based therapeutic strategies are very limited for subjects with mismatch repair proficient CRC (CRCpMMR). Here, we report a comprehensive typing of immune infiltrating cells in CRCpMMR. We also tested the expression and interferon-γ-modulation of PD-L1/CD274. Relevant findings were subsequently validated by immunohistochemistry on fixed materials. CRCpMMR contain a significantly increased fraction of CD163+ macrophages (TAMs) expressing TREM2 and CD66+ neutrophils (TANs) together with decrease in CD4-CD8-CD3+ double negative T lymphocytes (DNTs); no differences were revealed by the analysis of conventional and plasmacytoid dendritic cell populations. A fraction of tumor-infiltrating T-cells displays an exhausted phenotype, co-expressing PD-1 and TIM-3. Remarkably, expression of PD-L1 on fresh tumor cells and TAMs was undetectable even after in vitro stimulation with interferon-γ. These findings confirm the immune suppressive microenvironment of CRCpMMR characterized by dense infiltration of TAMs, occurrence of TANs, lack of DNTs, T-cell exhaustion, and interferon-γ unresponsiveness by host and tumor cells. Appropriate bypass strategies should consider these combinations of immune escape mechanisms in CRCpMMR.
Collapse
Affiliation(s)
- Mauro Giacomelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Diego Cesare Pezzola
- Department of Surgery, Surgery Division II, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mattia Bugatti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Antoni Van Leeuwenhoek-Nederlands Kanker Instituut, 1066 CX Amsterdam, The Netherlands
| | - Rossella Cioncada
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Laura Melocchi
- Department of Pathology, Fondazione Poliambulanza, 25124 Brescia, Italy
| | - Viviana Giustini
- CREA Laboratory, AIL Center for Hemato-Oncologic Research, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Vincenzo Villanacci
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Carla Baronchelli
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Stefania Manenti
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Luisa Imberti
- Section of Microbiology, University of Brescia, 25123 Brescia, Italy
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - William Vermi
- Department of Pathology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
- Department of Pathology and Immunology, School of Medicine, Washington University, Saint Louis, MO 63130, USA
| |
Collapse
|
38
|
Levy JJ, Zavras JP, Veziroglu EM, Nasir-Moin M, Kolling FW, Christensen BC, Salas LA, Barney RE, Palisoul SM, Ren B, Liu X, Kerr DA, Pointer KB, Tsongalis GJ, Vaickus LJ. Identification of Spatial Proteomic Signatures of Colon Tumor Metastasis: A Digital Spatial Profiling Approach. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:778-795. [PMID: 37037284 PMCID: PMC10284031 DOI: 10.1016/j.ajpath.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 02/24/2023] [Indexed: 04/12/2023]
Abstract
Over 150,000 Americans are diagnosed with colorectal cancer (CRC) every year, and annually >50,000 individuals are estimated to die of CRC, necessitating improvements in screening, prognostication, disease management, and therapeutic options. CRC tumors are removed en bloc with surrounding vasculature and lymphatics. Examination of regional lymph nodes at the time of surgical resection is essential for prognostication. Developing alternative approaches to indirectly assess recurrence risk would have utility in cases where lymph node yield is incomplete or inadequate. Spatially dependent, immune cell-specific (eg, tumor-infiltrating lymphocytes), proteomic, and transcriptomic expression patterns inside and around the tumor-the tumor immune microenvironment-can predict nodal/distant metastasis and probe the coordinated immune response from the primary tumor site. The comprehensive characterization of tumor-infiltrating lymphocytes and other immune infiltrates is possible using highly multiplexed spatial omics technologies, such as the GeoMX Digital Spatial Profiler. In this study, machine learning and differential co-expression analyses helped identify biomarkers from Digital Spatial Profiler-assayed protein expression patterns inside, at the invasive margin, and away from the tumor, associated with extracellular matrix remodeling (eg, granzyme B and fibronectin), immune suppression (eg, forkhead box P3), exhaustion and cytotoxicity (eg, CD8), Programmed death ligand 1-expressing dendritic cells, and neutrophil proliferation, among other concomitant alterations. Further investigation of these biomarkers may reveal independent risk factors of CRC metastasis that can be formulated into low-cost, widely available assays.
Collapse
Affiliation(s)
- Joshua J Levy
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire; Department of Dermatology, Dartmouth Health, Lebanon, New Hampshire; Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire; Program in Quantitative Biomedical Sciences, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire.
| | | | - Eren M Veziroglu
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire
| | | | | | - Brock C Christensen
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire; Department of Molecular and Systems Biology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire; Department of Community and Family Medicine, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire
| | - Lucas A Salas
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire; Department of Molecular and Systems Biology, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire; Integrative Neuroscience at Dartmouth Graduate Program, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire
| | - Rachael E Barney
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Scott M Palisoul
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Bing Ren
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Xiaoying Liu
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Darcy A Kerr
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| | - Kelli B Pointer
- Section of Radiation Oncology, Department of Medicine, Dartmouth College Geisel School of Medicine, Hanover, New Hampshire
| | - Gregory J Tsongalis
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire.
| | - Louis J Vaickus
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, New Hampshire
| |
Collapse
|
39
|
Bi Y, Yang Q, Li Z, Wang Y, Wang Y, Jia A, Pan Z, Yang R, Liu G. Aryl hydrocarbon receptor nuclear translocator limits the recruitment and function of regulatory neutrophils against colorectal cancer by regulating the gut microbiota. J Exp Clin Cancer Res 2023; 42:53. [PMID: 36859266 PMCID: PMC9976387 DOI: 10.1186/s13046-023-02627-y] [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: 11/14/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Although the role and mechanism of neutrophils in tumors have been widely studied, the precise effects of aryl hydrocarbon receptor nuclear translocator (ARNT) on neutrophils remain unclear. In this study, we investigated the roles of ARNT in the function of CD11b+Gr1+ neutrophils in colitis-associated colorectal cancer. METHODS Wild-type (WT), ARNT myeloid-specific deficient mice and a colitis-associated colorectal cancer mouse model were used in this study. The level and functions of CD11b+Gr1+ cells were evaluated by flow cytometry and confocal microscopy. RESULTS We found that ARNT deficiency drives neutrophils recruitment, neutrophil extracellular trap (NET) development, inflammatory cytokine secretion and suppressive activities when cells enter the periphery from bone marrow upon colorectal tumorigenesis. ARNT deficiency displays similar effects to aryl hydrocarbon receptor (AHR) deficiency in neutrophils. CXCR2 is required for NET development, cytokine production and recruitment of neutrophils but not the suppressive activities induced by Arnt-/- in colorectal cancer. The gut microbiota is essential for functional alterations in Arnt-/- neutrophils to promote colorectal cancer growth. The colorectal cancer effects of Arnt-/- neutrophils were significantly restored by mouse cohousing or antibiotic treatment. Intragastric administration of the feces of Arnt-/- mice phenocopied their colorectal cancer effects. CONCLUSION Our results defined a new role for the transcription factor ARNT in regulating neutrophils recruitment and function and the gut microbiota with implications for the future combination of gut microbiota and immunotherapy approaches in colorectal cancer.
Collapse
Affiliation(s)
- Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 100071, Beijing, China.
| | - Qiuli Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Zhengchao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 100071, Beijing, China
| | - Yuexin Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Yufei Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Anna Jia
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China
| | - Zhiyuan Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 100071, Beijing, China
| | - Ruifu Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, 100071, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, 100875, Beijing, China.
| |
Collapse
|
40
|
Wei X, Leng X, Li G, Wang R, Chi L, Sun D. Advances in research on the effectiveness and mechanism of Traditional Chinese Medicine formulas for colitis-associated colorectal cancer. Front Pharmacol 2023; 14:1120672. [PMID: 36909166 PMCID: PMC9995472 DOI: 10.3389/fphar.2023.1120672] [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: 12/10/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Inflammatory bowel disease (IBD) can progress into colitis-associated colorectal cancer (CAC) through the inflammation-cancer sequence. Although the mechanism of carcinogenesis in IBD has not been fully elucidated, the existing research indicates that CAC may represent a fundamentally different pathogenesis pattern of colorectal cancer. At present, there is no proven safe and effective medication to prevent IBD cancer. In recent years, Chinese medicine extracts and Chinese medicine monomers have been the subject of numerous articles about the prevention and treatment of CAC, but their clinical application is still relatively limited. Traditional Chinese Medicine (TCM) formulas are widely applied in clinical practice. TCM formulas have demonstrated great potential in the prevention and treatment of CAC in recent years, although there is still a lack of review. Our work aimed to summarize the effects and potential mechanisms of TCM formulas for the prevention and treatment of CAC, point out the issues and limitations of the current research, and provide recommendations for the advancement of CAC research in the future. We discovered that TCM formulas regulated many malignant biological processes, such as inflammation-mediated oxidative stress, apoptosis, tumor microenvironment, and intestinal microecology imbalance in CAC, through a review of the articles published in databases such as PubMed, SCOPUS, Web of Science, Embase, and CNKI. Several major signal transduction pathways, including NF-κB, STAT3, Wnt/β-catenin, HIF-1α, and Nrf2, were engaged. TCM formula may be a promising treatment candidate to control the colitis-cancer transformation, however further high-quality research is required.
Collapse
Affiliation(s)
- Xiunan Wei
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaohui Leng
- Weifang Traditional Chinese Hospital, Weifang, China
| | - Gongyi Li
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruting Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lili Chi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dajuan Sun
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
41
|
Lv J, Liu J, Chao G, Zhang S. PARs in the inflammation-cancer transformation of CRC. Clin Transl Oncol 2022; 25:1242-1251. [PMID: 36547764 DOI: 10.1007/s12094-022-03052-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the common malignancies with a global trend of increasing incidence and mortality. There is an urgent need to identify new predictive markers and therapeutic targets for the treatment of CRC. Protease-activated receptors (PARs) are a class of G-protein-coupled receptors, with currently identified subtypes including PAR1, PAR2, PAR3 and PAR4. Increasingly, studies suggest that PARs play an important role in the growth and metastasis of CRC. By targeting multiple signaling pathways may contribute to the pathogenesis of CRC. In this review, we first describe recent studies on the role of PARs in CRC inflammation-cancer transformation, focusing on the important role of PARs in signaling pathways associated with inflammation-cancer transformation, and summarize the progress of research on PARs-targeted drugs.
Collapse
Affiliation(s)
- Jianyu Lv
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Jinguo Liu
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University, Hanghou, China.
| | - Shuo Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Zhejiang, China.
| |
Collapse
|
42
|
Complement and Fungal Dysbiosis as Prognostic Markers and Potential Targets in PDAC Treatment. Curr Oncol 2022; 29:9833-9854. [PMID: 36547187 PMCID: PMC9777542 DOI: 10.3390/curroncol29120773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is still hampered by a dismal prognosis. A better understanding of the tumor microenvironment within the pancreas and of the factors affecting its composition is of utmost importance for developing new diagnostic and treatment tools. In this context, the complement system plays a prominent role. Not only has it been shown to shape a T cell-mediated immune response, but it also directly affects proliferation and apoptosis of the tumor cells, influencing angiogenesis, metastatic spread and therapeutic resistance. This makes complement proteins appealing not only as early biomarkers of PDAC development, but also as therapeutic targets. Fungal dysbiosis is currently the new kid on the block in tumorigenesis with cancer-associated mycobiomes extracted from several cancer types. For PDAC, colonization with the yeast Malassezia seems to promote cancer progression, already in precursor lesions. One responsible mechanism appears to be complement activation via the lectin pathway. In the present article, we review the role of the complement system in tumorigenesis, presenting observations that propose it as the missing link between fungal dysbiosis and PDAC development. We also present the results of a small pilot study supporting the crucial interplay between the complement system and Malassezia colonization in PDAC pathogenesis.
Collapse
|