1
|
Kwak JW, Houghton AM. Targeting neutrophils for cancer therapy. Nat Rev Drug Discov 2025:10.1038/s41573-025-01210-8. [PMID: 40374764 DOI: 10.1038/s41573-025-01210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/18/2025]
Abstract
Neutrophils are among the most abundant immune cell types in the tumour microenvironment and have been associated with poor outcomes across multiple cancer types. Yet despite mounting evidence of their role in tumour progression, therapeutic strategies targeting neutrophils have only recently gained attention and remain limited in scope. This is probably due to the increasing number of distinct neutrophil subtypes identified in cancer and the limited understanding of the mechanisms by which these subsets influence tumour progression and immune evasion. In this Review, we discuss the spectrum of neutrophil subtypes - including those with antitumour activity - and their potential to polarize towards tumour-suppressive phenotypes. We explore the molecular pathways and effector functions by which neutrophils modulate cancer progression, with an emphasis on identifying tractable therapeutic targets. Finally, we examine emerging clinical trials aimed at modulating neutrophil lineages and consider their implications for patient outcomes.
Collapse
Affiliation(s)
- Jeff W Kwak
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Translational Science and Therapeutics Division and Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Szlachetko JA, Hofmann-Vega F, Budeus B, Schröder LJ, Dumitru CA, Schmidt M, Deuss E, Vollmer S, Hanschmann EM, Busch M, Kehrmann J, Lang S, Dünker N, Hussain T, Brandau S. Tumor cells that resist neutrophil anticancer cytotoxicity acquire a prometastatic and innate immune escape phenotype. Cell Mol Immunol 2025; 22:527-540. [PMID: 40155451 PMCID: PMC12041228 DOI: 10.1038/s41423-025-01283-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: 09/23/2024] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
In the tumor host, neutrophils may exhibit protumor or antitumor activity. It is hypothesized that in response to host-derived or therapy-induced factors, neutrophils adopt diverse functional states to ultimately execute these differential functions. Here, we provide an alternative scenario in which the response of an individual tumor cell population determines the overall protumor versus antitumor outcome of neutrophil‒tumor interactions. Experimentally, we show that human neutrophils, which are sequentially stimulated with bacteria and secreted factors from tumor cells, kill a certain proportion of tumor target cells. However, the majority of the tumor cells remained resistant to this neutrophil-mediated killing and underwent a functional, phenotypic and transcriptomic switch that was reminiscent of partial epithelial‒to-mesenchymal transition. This cell biological switch was associated with physical escape from NK-mediated killing and resulted in enhanced metastasis to the lymph nodes in a preclinical orthotopic mouse model. Mechanistically, we identified the antimicrobial neutrophil granule proteins neutrophil elastase (NE) and matrix metalloprotease-9 (MMP-9) as the molecular mediators of this functional switch. We validated these data in patients with head and neck cancer and identified bacterially colonized intratumoral niches that were enriched for mesenchymal tumor cells and neutrophils expressing NE and MMP-9. Our data reveal the parallel execution of tumor cytotoxic and prometastatic activity by activated neutrophils and identify NE and MMP-9 as mediators of lymph node metastasis. The identified mechanism explains the functional dichotomy of tumor-associated neutrophils at the level of the tumor target cell response and has implications for superinfected cancers and the dysbiotic tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Bettina Budeus
- Institute of Cell Biology, University Hospital Essen, Essen, 45147, Germany
| | - Lara-Jasmin Schröder
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Institute of Pathology, Medical School Hannover, Hannover, 30625, Germany
| | - Claudia Alexandra Dumitru
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, 39106, Germany
| | - Mathias Schmidt
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Eric Deuss
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Sebastian Vollmer
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Eva-Maria Hanschmann
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Medical Faculty, Essen, 45147, Germany
| | - Jan Kehrmann
- Institute of Medical Microbiology, University Hospital Essen, Essen, 45147, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Medical Faculty, Essen, 45147, Germany
| | - Timon Hussain
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany
- Department of Otorhinolaryngology, Klinikum rechts der Isar, Technical University Munich, Munich, 81675, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, Essen, 45147, Germany.
- German Cancer Consortium, DKTK, Partner Site Essen-Düsseldorf, Essen, 45147, Germany.
| |
Collapse
|
3
|
Rys RN, Calcinotto A. Senescent neutrophils: a hidden role in cancer progression. Trends Cell Biol 2025; 35:399-411. [PMID: 39362804 DOI: 10.1016/j.tcb.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Neutrophils have recently received increased attention in cancer because they contribute to all stages of cancer. Neutrophils are so far considered to have a short half-life. However, a growing body of literature has shown that tumor-associated neutrophils (TANs) acquire a prolonged lifespan. This review discusses recent work surrounding the mechanisms by which neutrophils can persist in the tumor microenvironment (TME). It also highlights different scenarios for therapeutic targeting of protumorigenic neutrophils, supporting the idea that, in tumors, inhibition of neutrophil recruitment is not sufficient because these cells can persist and remain hidden from current interventions. Hence, the elimination of long-lived neutrophils should be pursued to increase the efficacy of standard therapy.
Collapse
Affiliation(s)
- Ryan N Rys
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland
| | - Arianna Calcinotto
- Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland; Università della Svizzera Italiana, Faculty of Biomedical Sciences, 6900 Lugano, Switzerland.
| |
Collapse
|
4
|
Cai W, Fan T, Xiao C, Deng Z, Liu Y, Li C, He J. Neutrophils in cancer: At the crucial crossroads of anti-tumor and pro-tumor. Cancer Commun (Lond) 2025. [PMID: 40296668 DOI: 10.1002/cac2.70027] [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: 09/19/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Neutrophils are important components of the immune system and play a key role in defending against pathogenic infections and responding to inflammatory cues, including cancer. Their dysregulation indicates potential disease risk factors. However, their functional importance in disease progression has often been underestimated due to their short half-life, especially as there is limited information on the role of intratumoral neutrophils. Recent studies on their prominent role in cancer have led to a paradigm shift in our understanding of the functional diversity of neutrophils. These studies highlight that neutrophils have emerged as key components of the tumor microenvironment, where they can play a dual role in promoting and suppressing cancer. Moreover, several approaches to therapeutically target neutrophils have emerged, and clinical trials are investigating their efficacy. In this review, we discussed the involvement of neutrophils in cancer initiation and progression. We summarized recent advances in therapeutic strategies targeting neutrophils and, most importantly, suggested future research directions that could facilitate the manipulation of neutrophils for therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
5
|
Yang Y, Hu Z, Ye Y, Wu H, Sun W, Wang N. Association of aggregate index of systemic inflammation with increased all-cause and cardiovascular mortality in female cancer patients. Front Oncol 2025; 15:1552341. [PMID: 40365348 PMCID: PMC12069043 DOI: 10.3389/fonc.2025.1552341] [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: 01/02/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background Cancer is a leading cause of death, especially among women, with cancers like breast, ovarian, and cervical cancer presenting unique diagnostic and treatment challenges. Systemic inflammation plays a significant role in cancer progression, affecting both tumor development and therapeutic outcomes. Despite the established link between inflammation and cancer, comprehensive studies on the prognostic value of the Aggregate Index of Systemic Inflammation (AISI) in female cancer patients are lacking. This study explores the association between AISI and mortality outcomes, including all-cause and cardiovascular mortality, in female cancer patients. Methods This study analyzes data from the NHANES database and Dandong Central Hospital. Kaplan-Meier survival curves and multivariable Cox proportional hazards regression analyses were used to assess the relationship between AISI and all-cause and cardiovascular mortality. Restricted cubic spline plots and subgroup analyses were applied to explore potential interactions. Results Elevated AISI levels were strongly associated with increased all-cause and cardiovascular mortality. Patients in the highest AISI quartile demonstrated significantly higher mortality risks compared to those in the lowest quartile. ROC curve analysis indicated superior predictive performance of AISI over SII. Restricted cubic spline plots revealed a linear relationship, with mortality risk notably increasing when AISI levels were elevated. Conclusion AISI is a robust predictor of all-cause and cardiovascular mortality in female cancer patients. Its ease of measurement and strong prognostic value make it a valuable tool for risk assessment and management in this population.
Collapse
Affiliation(s)
- Ying Yang
- Dalian Medical University, Dalian, China
- The Dandong Central Hospital, Dandong, China
| | - Zelin Hu
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Yuqin Ye
- Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Haoqi Wu
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Wei Sun
- The Dandong Central Hospital, Dandong, China
| | - Ning Wang
- The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Zhang J, Miao C, Zhang H. Targeting neutrophil extracellular traps in cancer progression and metastasis. Theranostics 2025; 15:5846-5869. [PMID: 40365275 PMCID: PMC12068306 DOI: 10.7150/thno.111096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Neutrophils serve as pivotal effectors and regulators of the intricate immune system. Their contributions are indispensable, encompassing the obliteration of pathogens and a significant role in both cancer initiation and progression. Conversely, malignancies profoundly affect neutrophil activity, maturation, and lifespans. Cancer cells manipulate their biology to enhance or suppress the key functions of neutrophils. This manipulation is one of the most remarkable defensive mechanisms used by neutrophils, including the formation of neutrophil extracellular traps (NETs). NETs are filamentous structures comprising DNA, histones, and proteins derived from cytotoxic granules. In this review, we discuss the bidirectional interplay in which cancer elicits NET formation, and NETs concurrently facilitate cancer progression. Here, we discuss how vascular dysfunction and thrombosis induced by neutrophils and NETs contribute to an elevated risk of mortality from cardiovascular complications in patients with cancer. Ultimately, we propose a series of therapeutic strategies that hold promise for effectively targeting NETs in clinical settings.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
- Department of Anesthesiology, Shanghai Medical College, Fudan University, China
| |
Collapse
|
7
|
Lin S, Li Y, Liu W, Du Y, Tao T. Metabolic Implications of Elevated Neutrophil Extracellular Traps in Polycystic Ovary Syndrome: A Focus on Hepatic Glycolysis. Biomolecules 2025; 15:572. [PMID: 40305335 PMCID: PMC12025135 DOI: 10.3390/biom15040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Polycystic ovary syndrome (PCOS) is a leading cause of infertility but also a metabolic disorder, frequently associated with obesity, insulin resistance, and diabetes. However, its etiology remains inadequately understood. Recent studies have increasingly implicated neutrophil extracellular traps (NETs) in the pathogenesis of metabolic diseases. Methods: Serum and follicular fluid samples were collected from patients with PCOS and control populations to assess NETs levels. The effects of NETs were investigated using DNase I to reduce NETs in dehydroepiandrosterone sulfate (DHEAS)-induced PCOS rats. Metabolic differences were further analyzed by untargeted metabolomics, and in vitro studies were conducted using primary bone marrow-derived neutrophils and normal mouse liver cell lines. Results: Markers of NETs in both the serum and follicular fluid of patients with PCOS were significantly higher than those in the control group. PCOS rats treated with DNase I exhibited significant improvements in glucose metabolism. Untargeted metabolomics analysis of liver tissue from these rats revealed alterations in the glycolysis pathway. Subsequent in vitro experiments demonstrated that treatment with NETs-conditioned medium (NETs CM) led to reduced insulin sensitivity, glucose uptake, and glucose utilization in liver cells, accompanied by varying degrees of decline in the transcription, translation, and function of glycolysis pathway proteins. Conclusions: NETs may be involved in the regulation of insulin resistance pathogenesis in PCOS by downregulating glycolytic pathways in the liver. Our study offers a novel strategy for insulin resistance intervention in PCOS.
Collapse
Affiliation(s)
- Siyu Lin
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Yushan Li
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Wei Liu
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| | - Yanzhi Du
- Center for Reproductive Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200135, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China
| | - Tao Tao
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (S.L.)
| |
Collapse
|
8
|
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
|
9
|
Yu L, Liebenberg K, Shen Y, Liu F, Xu Z, Hao X, Wu L, Zhang W, Chan HL, Wei B, Lorenzi PL, Gao Y, Bado I, Becerra-Dominguez L, Rivas CH, Aguirre S, Pingel BC, Wu YH, Ding Y, Liu J, Edwards DG, Eberlin LS, Zhang XHF. Tumor-derived arachidonic acid reprograms neutrophils to promote immune suppression and therapy resistance in triple-negative breast cancer. Immunity 2025; 58:909-925.e7. [PMID: 40157359 PMCID: PMC11981829 DOI: 10.1016/j.immuni.2025.03.002] [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: 10/07/2024] [Revised: 12/08/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
The combination of immune checkpoint blockade and chemotherapies is the standard of care for triple-negative breast cancer (TNBC). However, initially, responsive tumors can still develop recurrences, suggesting acquired resistance mechanisms that remain poorly understood. Herein, we discover that TNBC cells surviving anti-programmed cell death protein-1 (anti-PD-1) and chemotherapy treatment accumulate neutral lipids. Disrupting lipid droplet formation in cancer cells reverses resistance and mitigates the immunosuppressive microenvironment. Single-cell RNA sequencing reveals a subset of neutrophils exhibiting a lipid-laden phenotype similar to adjacent tumor cells. Mechanistically, tumor-derived extracellular vesicles carrying lipids, including arachidonic acid (AA), mediate neutrophil reprogramming. Blocking dietary intake of omega-6 fatty acids or inhibiting fatty acid elongation for AA synthesis restores anti-tumor immunity and re-sensitizes the resistant tumors to anti-PD-1 and chemotherapy treatment. In human patients, AA metabolism-related pathways correlates with neutrophil enrichment. Overall, we demonstrate how lipid accumulation in TNBC cells leads to immune suppression and therapy resistance.
Collapse
Affiliation(s)
- Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Keziah Liebenberg
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luis Becerra-Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bradley C Pingel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Livia S Eberlin
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
10
|
Huang C, Qiu H, Xu C, Tan Z, Jin M, Hu J, Huang Z, Zhou Y, Ge S, Hu X. Downregulation of tropomyosin 2 promotes the progression of lung adenocarcinoma by regulating neutrophil infiltration through neutrophil elastase. Cell Death Dis 2025; 16:264. [PMID: 40199876 PMCID: PMC11978998 DOI: 10.1038/s41419-025-07531-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Lung adenocarcinoma (LUAD) is a common malignant tumor in the lung that seriously endangers the health of people worldwide. The neutrophil-associated inflammatory microenvironment contributes to the activation of tumor cells. In this study, we report a role of tumor-associated neutrophils (TANs) promote tumor progression of LUAD by crosstalk between neutrophils and tumor cells. Mechanistically, in co-culture with tumor cells, downregulation of TPM2 on tumor cells increases neutrophil elastase (ELANE) levels in neutrophils regulated by p38/ MAPK signaling activation, and ELANE promotes tumor cell progression through the Hippo pathway. Furthermore, downregulation of TPM2 activates ELANE of neutrophils to facilitate ERK1/2 activation, thus enhancing IL1β and IL8 secretion for chemoattraction of more neutrophils to tumor microenvironment. The new studies identify an accomplice role for the interaction between TPM2 and ELANE in promoting LUAD progression and provide potential strategies in the prevention and/or treatment of LUAD and other cancers.
Collapse
Affiliation(s)
- Caixiu Huang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Hao Qiu
- The First People's Hospital of Changde City, Changde, PR China
| | - Changting Xu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | - Zilong Tan
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Mei Jin
- Pingxiang People's Hospital, Pingxiang, PR China
| | - Jing Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China
| | | | - Yuwei Zhou
- Nanchang University Second Affiliated Hospital, Nanchang, PR China
| | - Shengyou Ge
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| | - Xiaoyuan Hu
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, PR China.
| |
Collapse
|
11
|
HAO X, FENG Y, LU A, SUN Y, XIA J, MEI X, FENG L, JIANG M, WANG B, YANG H. [Research Progress of Neutrophil Extracellular Traps in Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2025; 28:201-212. [PMID: 40210480 PMCID: PMC11986667 DOI: 10.3779/j.issn.1009-3419.2025.106.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Indexed: 04/12/2025]
Abstract
Neutrophil extracellular traps (NETs), intricate reticular structures released by activated neutrophils, play a pivotal regulatory role in the pathogenesis of malignant tumors. Lung cancer is one of the most prevalent malignancies globally, with persistently high incidence and mortality rates. Recent studies have revealed that NETs dynamically modulate the tumor microenvironment through unique pathological mechanisms, exhibiting complex immunoregulatory characteristics during the progression of lung cancer, and this discovery has increasingly become a focal point in tumor immunology research. This paper provides a comprehensive review of the latest advancements in NETs research related to lung cancer, offering an in-depth analysis of their impact on lung cancer progression, their potential diagnostic value, and the current state of research on targeting NETs for lung cancer prevention and treatment. The aim is to propose novel strategies to enhance therapeutic outcomes and improve the prognosis for lung cancer patients.
.
Collapse
|
12
|
Li S, Dong P, Wu X, Kang Z, Yan G. Global trends in tumor-associated neutrophil research: a bibliometric and visual analysis. Front Immunol 2025; 16:1478092. [PMID: 40160822 PMCID: PMC11949894 DOI: 10.3389/fimmu.2025.1478092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Background Tumor-associated neutrophils (TANs) play crucial roles in tumor progression, immune response modulation, and the therapeutic outcomes. Despite significant advancements in TAN research, a comprehensive bibliometric analysis that objectively presents the current status and trends in this field is lacking. This study aims to fill this gap by visually analyzing global trends in TANs research using bibliometric and knowledge mapping techniques. Methods We retrieved articles and reviews related to TANs from the Web of Science core collection database, spanning the period from 2012 to2024. The data was analyzed using bibliometric tools such as Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio) to identify key trends, influential countries and institutions, collaborative networks. and citation patterns. Results A total of 6l5 publications were included in the bibliometric analysis, showing a significant upward trend in TANs research over the last two decades. The United States and China emerged as the leading contributors with the highest number of publications and citations. The journal with the most publications in this field is Frontiers in Immunology, Prominent authors such as Fridlender ZG was identified as the key contributor, with his works frequently cited. The analysis highlighted major research themes. including the role of TANs in tumor microenvironment modulation, their dual functions in tumor promotion and suppression, and the exploration of TANs-targeted therapies, Emerging research hotspots include studies on TANs plasticity and their interactions with other immune cells. Conclusion This study is the first to employ bibliometric methods to visualize trends and frontiers in TANs research. The findings provide valuable insights into the evolution of the field, highlighting critical areas for future investigation and potential collaborative opportunities. This comprehensive analysis serves as a crucial resource for researchers and practitioners aiming to advance TAN research and its application in cancer therapy.
Collapse
Affiliation(s)
- Shaodong Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Peng Dong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhenhua Kang
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Guoqiang Yan
- Department of Colorectal & Anal Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Kaya TA, Stein KP, Schaufler A, Neyazi B, Rashidi A, Kahlert UD, Mawrin C, Sandalcioglu IE, Dumitru CA. The tumor-neutrophil interactions in the microenvironment of brain metastases with different primary sites. J Leukoc Biol 2025; 117:qiae248. [PMID: 39565891 DOI: 10.1093/jleuko/qiae248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/07/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Abstract
Brain metastases originating from lung and breast cancer can recruit and activate neutrophils to acquire a tumor-promoting phenotype. It is currently unclear if this phenomenon also occurs in brain metastases arising from other primary sites. Here, we investigated the effect of tumor cells isolated from melanoma, lung cancer, and gastrointestinal cancer brain metastases on neutrophil biology and functions. We found that lung and gastrointestinal but not melanoma brain metastasis cells produced CXCL8/IL-8 and promoted neutrophil recruitment. Similarly, lung and gastrointestinal but not melanoma brain metastasis cells prolonged the survival of neutrophils and stimulated them to release MMP9 and CCL4/MIP1β. In situ, lung and gastrointestinal brain metastasis tissues contained significantly higher numbers of tumor-infiltrating neutrophils compared to melanoma brain metastases. The levels of neutrophil infiltration significantly correlated with the proliferation index of these tumors. Our findings identify variabilities in the immune microenvironment of brain metastases with different primary sites, which may ultimately affect their pathophysiology and progression.
Collapse
Affiliation(s)
- Tamer A Kaya
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Klaus-Peter Stein
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Anna Schaufler
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Belal Neyazi
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Ali Rashidi
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, University Clinic for General-, Visceral-, Vascular- and Transplantation Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Christian Mawrin
- Department of Neuropathology, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - I Erol Sandalcioglu
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Claudia A Dumitru
- Department of Neurosurgery, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
14
|
Lee D, Oh S, Lawler S, Kim Y. Bistable dynamics of TAN-NK cells in tumor growth and control of radiotherapy-induced neutropenia in lung cancer treatment. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2025; 22:744-809. [PMID: 40296792 DOI: 10.3934/mbe.2025028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neutrophils play a crucial role in the innate immune response as a first line of defense in many diseases, including cancer. Tumor-associated neutrophils (TANs) can either promote or inhibit tumor growth in various steps of cancer progression via mutual interactions with cancer cells in a complex tumor microenvironment (TME). In this study, we developed and analyzed mathematical models to investigate the role of natural killer cells (NK cells) and the dynamic transition between N1 and N2 TAN phenotypes in killing cancer cells through key signaling networks and how adjuvant therapy with radiation can be used in combination to increase anti-tumor efficacy. We examined the complex immune-tumor dynamics among N1/N2 TANs, NK cells, and tumor cells, communicating through key extracellular mediators (Transforming growth factor (TGF-$ \beta $), Interferon gamma (IFN-$ \gamma $)) and intracellular regulation in the apoptosis signaling network. We developed several tumor prevention strategies to eradicate tumors, including combination (IFN-$ \gamma $, exogenous NK, TGF-$ \beta $ inhibitor) therapy and optimally-controlled ionizing radiation in a complex TME. Using this model, we investigated the fundamental mechanism of radiation-induced changes in the TME and the impact of internal and external immune composition on the tumor cell fate and their response to different treatment schedules.
Collapse
Affiliation(s)
- Donggu Lee
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
| | - Sunju Oh
- Department of Biological Sciences, Konkuk University, Seoul 05029, Republic of Korea
| | - Sean Lawler
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
- Department of Pathology and Laboratory Medicine, Legorreta Brown Cancer Center, Brown University, Providence, RI 02912, USA
| |
Collapse
|
15
|
Xu X, Tian M, Ding CC, Xu H, Wang H, Jin X. Skeletal Muscle Index-Based Cachexia Index as a Predictor of Prognosis in Patients With Cancer: A Meta-Analysis and Systematic Review. Nutr Rev 2025; 83:e852-e865. [PMID: 39001797 DOI: 10.1093/nutrit/nuae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024] Open
Abstract
CONTEXT Cachexia is associated with poor survival rates. In the clinical setting, the diagnosis of cancer cachexia is challenging. The cachexia index (CXI), a new index for predicting survival time, is a promising tool for diagnosing cancer cachexia; however, its efficacy in predicting patient survival has not been validated. OBJECTIVE This meta-analysis and systematic review aimed to explore the CXI's prognostic value in patients with cancer. DATA SOURCES The PubMed, Embase, MEDLINE, and Cochrane Library databases were searched for relevant studies to determine the association between CXI findings and prognosis. DATA EXTRACTION The outcomes were overall survival (OS), progression-, disease-, and recurrence-free survival (PFS/DFS/RFS) rates, and the rate of complete response. DATA ANALYSIS The QUality In Prognostic Studies (QUIPS) tool was used to evaluate the quality of the included trials. This meta-analysis comprised 14 studies involving 2777 patients. A low CXI was associated with decreased OS (hazard ratio [HR] 2.34, 95% confidence interval [CI] 2.01-2.72; P < .001), PFS/DFS/RFS (HR 1.93, 95% CI 1.68-2.22; P < .001), and complete response (odds ratio [OR] 0.49, 95% CI 0.36-0.66; P < .001). Patients with a low CXI had a lower body mass index (mean difference [MD] -0.75, 95% CI -1.00 to 0.50; P < .001), skeletal muscle index (standardized MD -0.80, 95% CI -0.98 to -0.61; P < .001), and serum albumin level (MD -0.23, 95% CI -0.26 to -0.20; P < .001); and a higher neutrophil-lymphocyte ratio (MD 1.88, 95% CI 1.29-2.47; P < .001) and more advanced disease stages (OR 0.80, 95% CI 0.71-0.91; P = .001). CONCLUSION A low CXI was found to be associated with poor survival in patients with cancer. While the CXI is a promising marker for predicting cancer cachexia, further studies are required to verify its usefulness.
Collapse
Affiliation(s)
- Xintian Xu
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Mengxing Tian
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Chen Chen Ding
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Huiting Xu
- Department of Abdominal Oncology 1, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Huifen Wang
- Nursing Department, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| | - Xin Jin
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430079, China
| |
Collapse
|
16
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [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: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
17
|
Oi H, Taki T, Kuroe T, Sakamoto N, Sakashita S, Kojima M, Sugiyama E, Umemura S, Sakai T, Izumi H, Zenke Y, Matsumoto S, Yoh K, Ishii M, Tsuboi M, Goto K, Ishii G. NETosis in pulmonary pleomorphic carcinoma. Cancer Sci 2025; 116:524-532. [PMID: 39555736 PMCID: PMC11786310 DOI: 10.1111/cas.16332] [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: 04/07/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 11/19/2024] Open
Abstract
Pulmonary pleomorphic carcinoma (PC) is a rare non-small-cell lung carcinoma (NSCLC) with a poor prognosis, characterized by tumor necrosis (TN). NETosis is a form of neutrophil-specific cell death, which is morphologically characterized by prominent neutrophil infiltration and cell detritus in the necrotic foci. Seventy-six patients with pulmonary PC who underwent complete resection were enrolled. Tumor necrosis was evaluated using digitally scanned resected specimens. The regions of NETosis were quantified using citrullinated histone H3 (citH3)- and myeloperoxidase-positive regions. We examined the association between the NETosis area and the prognostic outcomes and assessed the correlation between the NETosis area and systemic inflammation. Tumor necrosis was observed in 70 patients (92%). In all the cases, the TN region was accompanied by a citH3-positive region. The patients with high NETosis area (n = 54) had significantly shorter overall survival than those with low NETosis area (n = 16) (p = 0.013). Furthermore, a high NETosis area was an independent poor prognostic factor in the multivariate analyses. Systemic inflammatory markers, including C-reactive protein (CRP), CRP-to-albumin ratio, and neutrophil-to-lymphocyte ratio, were significantly higher in patients with high NETosis area than in those with low NETosis area. Furthermore, the levels of these inflammatory markers were significantly decreased postsurgery. This study shows that in surgically resected pulmonary PC, patients with high NETosis areas have higher systemic inflammation and worse prognosis.
Collapse
Affiliation(s)
- Hajime Oi
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
- Department of Respiratory MedicineNagoya University Graduate School of MedicineNagoyaJapan
| | - Tetsuro Taki
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
| | - Takashi Kuroe
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
| | - Naoya Sakamoto
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
- Division of Pathology, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Shingo Sakashita
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
- Division of Pathology, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Motohiro Kojima
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
- Division of Pathology, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Eri Sugiyama
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Shigeki Umemura
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Tetsuya Sakai
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Hiroki Izumi
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Yoshitaka Zenke
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Shingo Matsumoto
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Kiyotaka Yoh
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Makoto Ishii
- Department of Respiratory MedicineNagoya University Graduate School of MedicineNagoyaJapan
| | - Masahiro Tsuboi
- Department of Thoracic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Koichi Goto
- Department of Thoracic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Genichiro Ishii
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital EastKashiwaJapan
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| |
Collapse
|
18
|
Pang J, Ding N, Liu X, He X, Zhou W, Xie H, Feng J, Li Y, He Y, Wang S, Xiao Z. Prognostic Value of the Baseline Systemic Immune-Inflammation Index in HER2-Positive Metastatic Breast Cancer: Exploratory Analysis of Two Prospective Trials. Ann Surg Oncol 2025; 32:750-759. [PMID: 39565489 DOI: 10.1245/s10434-024-16454-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024]
Abstract
PURPOSE The systemic immune-inflammation index (SII) is a hematological marker that reflects the immune status of the body. This study was designed to evaluate the prognostic significance of the baseline SII in HER2-positive metastatic breast cancer (MBC) patients receiving chemotherapy plus trastuzumab without or with pertuzumab. METHODS Data were collected from 774 patients from the CLEOPATRA trial, 196 patients from the H0648G trial, and 229 patients from six clinical centers in China. Patients were divided into the low and high SII subgroups according to the median SII value. The inverse probability of treatment weighting (IPTW) method was used to control bias. Associations between the SII and progression-free survival (PFS) and overall survival (OS) were analyzed. RESULTS In the CLEOPATRA trial, a lower SII was associated with better PFS (hazard ratio [HR] 1.31, 95% confidence interval [CI] 1.04-1.65, P = 0.02) and OS (HR 1.42, 95% CI 1.07-1.88, P = 0.02) in the trastuzumab and docetaxel groups, as well as improved PFS in the trastuzumab and pertuzumab and docetaxel groups (HR 1.39, 95% CI 1.10-1.77, P < 0.01) after IPTW. In the H0648G trial, a lower SII was associated with better PFS (P = 0.04) and OS (P = 0.02) in HER2-positive MBC patients receiving trastuzumab-based therapy. According to real-world data, a lower SII predicted an improvement in PFS for patients treated with docetaxel plus trastuzumab without or with pertuzumab (P = 0.02 and 0.01, respectively). CONCLUSIONS A low baseline SII is associated with better survival outcomes among HER2-positive MBC patients receiving trastuzumab-based first-line therapy.
Collapse
Affiliation(s)
- Jian Pang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Nianhua Ding
- Department of Clinical Laboratory, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuan Liu
- Department of General Surgery, The First People's Hospital of Xiangtan, Xiangtan, China
| | - Xiongbin He
- Department of Breast and Thyroid Surgery, The First People's Hospital of Chenzhou, Chenzhou, China
| | - Wei Zhou
- Department of Breast Surgery, The Affiliated Zhuzhou Hospital of Xiangya School of Medicine, Zhuzhou, China
| | - Haiqing Xie
- Department of Breast and Thyroid Surgery, The Third People's Hospital of Chenzhou, Chenzhou, China
| | - Jianqi Feng
- Department of Breast and Thyroid Surgery, The First People's Hospital of Huaihua, Huaihua, China
| | - Yan Li
- Changsha Medical University, Changsha, Hunan, China
| | - Yingjian He
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education/Beijing), Breast Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shouman Wang
- Department of Breast Surgery, Multidisciplinary Breast Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Xiao
- Department of Breast Surgery, General Surgery, Clinical Research Center for Breast Cancer in Hunan Province, National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Hospital, Central South University, Xiangya, China.
| |
Collapse
|
19
|
Liu X, Yan G, Pang J, Xiao Z, Xie H. The Systemic Immune-Inflammation Index is a Predictor of Chemotherapy Sensitivity and Disease-Free Survival in Patients With Hormone Receptor-Positive Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer. Clin Breast Cancer 2025; 25:e190-e195. [PMID: 39448293 DOI: 10.1016/j.clbc.2024.09.016] [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: 04/21/2024] [Revised: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND The relationship between the systemic immune-inflammation index (SII), chemotherapy sensitivity, and prognosis in HR+HER2- breast cancer (BC) has not been extensively studied. PATIENTS AND METHODS Clinical data from 980 patients diagnosed with HR+HER2- BC between June 2012 and June 2016 were collected. Patients were divided into low- and high-SII groups according to median SII value. Differences among variables were assessed using the chi-squared test. The inverse probability of treatment weighting (IPTW) method was used to control bias. The associations between clinicopathological factors, baseline SII, and disease-free survival (DFS) were analyzed using Kaplan-Meier curves and Cox analyses. RESULTS The median follow-up period was 37 months (5-77). 480 patients underwent neoadjuvant chemotherapy, and low baseline SII values were associated with higher pathological complete response (pCR) rates than those in the high SII group (16.4% vs. 9.2%; P = .019). Multivariate analyses revealed that larger tumor size, more lymph node involvement, high Ki-67 score, and high baseline SII were independent prognostic factors for worse outcomes in patients with HR+HER2- BC. The risk for metastasis/recurrence was higher in the high SII group compared with that in the low SII group (hazard ratio 2.07 [95% CI, 1.35-3.19]; P = .001). After IPTW, a similar result was obtained, in that a high SII value was significantly associated with worse DFS among patients with HR+HER2- BC. CONCLUSION A low baseline SII was associated with higher pCR rates after neoadjuvant chemotherapy and was an independent prognostic factor for better DFS outcomes in patients with HR+HER2- BC.
Collapse
Affiliation(s)
- Xuan Liu
- Department of General Surgery, The First People's Hospital of Xiangtan, Xiangtan, China
| | - Guoqing Yan
- External Department 1, Guangdong Provincial General Hospital, Chinese People's Armed Police Force, Guangzhou, China
| | - Jian Pang
- Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Xiao
- Department of Breast Surgery, General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Haiqing Xie
- Department of Breast and Thyroid Surgery, The First People's Hospital of Chenzhou, Chenzhou, China.
| |
Collapse
|
20
|
Lu T, Li W. Neutrophil Engulfment in Cancer: Friend or Foe? Cancers (Basel) 2025; 17:384. [PMID: 39941753 PMCID: PMC11816126 DOI: 10.3390/cancers17030384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Neutrophils, the most abundant circulating white blood cells, are essential for the initial immune response to infection and injury. Emerging research reveals a dualistic function of neutrophils in cancer, where they can promote or inhibit tumor progression. This dichotomy is influenced by the tumor microenvironment, with neutrophils capable of remodeling the extracellular matrix, promoting angiogenesis, or alternatively inducing cancer cell death and enhancing immune responses. An intriguing yet poorly understood aspect of neutrophil-cancer interactions is the phenomenon of neutrophil engulfment by cancer cells, which has been observed across various cancers. This process, potentially mediated by LC3-associated phagocytosis (LAP), raises questions about whether it serves as a mechanism for immune evasion or contributes to tumor cell death through pathways like ferroptosis. This review examines current knowledge on neutrophil development, their roles in cancer, and the mechanisms of LAP in neutrophil engulfment by tumor cells. We discuss how manipulating LAP impacts cancer progression and may represent a therapeutic strategy. We also explore neutrophils' potential as delivery vehicles for cancer therapeutic agents. Understanding the complex functions of tumor-associated neutrophils (TANs) and the molecular mechanisms underlying LAP in cancer may open new avenues for effective therapeutic interventions and mitigate potential risks.
Collapse
Affiliation(s)
- Tong Lu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
21
|
Zhu M, Jia R, Zhang X, Xu P. The success of the tumor immunotherapy: neutrophils from bench to beside. Front Immunol 2025; 16:1524038. [PMID: 39925807 PMCID: PMC11802522 DOI: 10.3389/fimmu.2025.1524038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/07/2025] [Indexed: 02/11/2025] Open
Abstract
The present immune therapy was focused on the immune checkpoint blockade or Chimeric Antigen Receptor T-Cell Immunotherapy (CART) transfer, but how to activate the innate immune system to antitumor still lags out. Neutrophils are the most abundant circulating leukocytes in human, and heterogeneous neutrophils have been increasingly recognized as important players in tumor progression. They play double "edge-sward" by either supporting or suppressing the tumor growth, including driving angiogenesis, extracellular matrix remodeling to promote tumor growth, participating in antitumor adaptive immunity, or killing tumor cells directly to inhibit the tumor growth. The complex role of neutrophils in various tumors depends on the tumor microenvironment (TME) they are located, and emerging evidence has suggested that neutrophils may determine the success of tumor immunotherapy in the context of the immune checkpoint blockade, innate immune training, or drug-loaded extracellular microvesicles therapy, which makes them become an exciting target for tumor immunotherapy, but still with challenges. Here, we summarize the latest insights on how to activate neutrophils in antitumor immunity and discuss the advances of neutrophil-targeted immunotherapy strategies.
Collapse
Affiliation(s)
- Meng Zhu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ru Jia
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaojie Zhang
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Pingwei Xu
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
22
|
Liu Z, Dou Y, Lu C, Han R, He Y. Neutrophil extracellular traps in tumor metabolism and microenvironment. Biomark Res 2025; 13:12. [PMID: 39849606 PMCID: PMC11756210 DOI: 10.1186/s40364-025-00731-z] [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: 10/28/2024] [Accepted: 01/11/2025] [Indexed: 01/25/2025] Open
Abstract
Neutrophil extracellular traps (NETs) are intricate, web-like formations composed of DNA, histones, and antimicrobial proteins, released by neutrophils. These structures participate in a wide array of physiological and pathological activities, including immune rheumatic diseases and damage to target organs. Recently, the connection between NETs and cancer has garnered significant attention. Within the tumor microenvironment and metabolism, NETs exhibit multifaceted roles, such as promoting the proliferation and migration of tumor cells, influencing redox balance, triggering angiogenesis, and driving metabolic reprogramming. This review offers a comprehensive analysis of the link between NETs and tumor metabolism, emphasizing areas that remain underexplored. These include the interaction of NETs with tumor mitochondria, their effect on redox states within tumors, their involvement in metabolic reprogramming, and their contribution to angiogenesis in tumors. Such insights lay a theoretical foundation for a deeper understanding of the role of NETs in cancer development. Moreover, the review also delves into potential therapeutic strategies that target NETs and suggests future research directions, offering new perspectives on the treatment of cancer and other related diseases.
Collapse
Affiliation(s)
- Zhanrui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Rui Han
- Department of Respiratory Disease, Bishan hospital of Chongqing medical university, Chongqing, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
23
|
Luyang H, Zeng F, Lei Y, He Q, Zhou Y, Xu J. Bidirectional role of neutrophils in tumor development. Mol Cancer 2025; 24:22. [PMID: 39819428 PMCID: PMC11737241 DOI: 10.1186/s12943-025-02228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Neutrophils, traditionally considered as non-specific components of the innate immune system, have garnered considerable research interest due to their dual roles in both promoting and inhibiting tumor progression. This paper seeks to clarify the specific mechanisms by which neutrophils play a bidirectional role in tumor immunity and the factors that influence these roles. By conducting a comprehensive analysis and synthesis of a vast array of relevant literature, it has become evident that neutrophils can influence tumor development and invasive migration through various mechanisms, thereby exerting their anti-tumor effects. Conversely, they can also facilitate tumorigenesis and proliferation, as well as affect the normal physiological functions of other immune cells, thus exerting pro-tumor effects. Moreover, neutrophils are influenced by tumor cells and their unique microenvironment, which in turn affects their heterogeneity and plasticity. Neutrophils interact with tumor cells to regulate various aspects of their life activities precisely. This paper also identifies unresolved issues in the research concerning the bidirectional role of neutrophils in tumorigenesis and tumor development, offering new opportunities and challenges for advancing our understanding. This, in turn, can aid in the proper application of these insights to clinical treatment strategies.
Collapse
Affiliation(s)
- Haoxin Luyang
- Department of critical care medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Feng Zeng
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Yan Lei
- Department of Blood Transfusion, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
| | - Qian He
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of Medicine, Central South University, Changsha, 410011, Hunan, China.
| | - Juan Xu
- Department of critical care medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Hunan Cancer Hospital, Changsha, 410013, Hunan, China.
| |
Collapse
|
24
|
Eruslanov E, Nefedova Y, Gabrilovich DI. The heterogeneity of neutrophils in cancer and its implication for therapeutic targeting. Nat Immunol 2025; 26:17-28. [PMID: 39747431 PMCID: PMC12055240 DOI: 10.1038/s41590-024-02029-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Neutrophils have a pivotal role in safeguarding the host against pathogens and facilitating tissue remodeling. They possess a large array of tools essential for executing these functions. Neutrophils have a critical role in cancer, where they are largely associated with negative clinical outcome and resistance to therapy. However, the specific role of neutrophils in cancer is complex and controversial, owing to their high functional diversity and acute sensitivity to the microenvironment. In this Perspective, we discuss the accumulated evidence that suggests that the functional diversity of neutrophils can be ascribed to two principal functional states, each with distinct characteristics: classically activated neutrophils and pathologically activated immunosuppressive myeloid-derived suppressor cells. We discuss how the antimicrobial factors in neutrophils can contribute to tumor progression and the fundamental mechanisms that govern the pathologically activated myeloid-derived suppressor cells. These functional states play divergent roles in cancer and thus require separate consideration in therapeutic targeting.
Collapse
Affiliation(s)
- Evgeniy Eruslanov
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | |
Collapse
|
25
|
Koenderman L, Vrisekoop N. Neutrophils in cancer: from biology to therapy. Cell Mol Immunol 2025; 22:4-23. [PMID: 39653768 PMCID: PMC11686117 DOI: 10.1038/s41423-024-01244-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
The view of neutrophils has shifted from simple phagocytic cells, whose main function is to kill pathogens, to very complex cells that are also involved in immune regulation and tissue repair. These cells are essential for maintaining and regaining tissue homeostasis. Neutrophils can be viewed as double-edged swords in a range of situations. The potent killing machinery necessary for immune responses to pathogens can easily lead to collateral damage to host tissues when inappropriately controlled. Furthermore, some subtypes of neutrophils are potent pathogen killers, whereas others are immunosuppressive or can aid in tissue healing. Finally, in tumor immunology, many examples of both protumorigenic and antitumorigenic properties of neutrophils have been described. This has important consequences for cancer therapy, as targeting neutrophils can lead to either suppressed or stimulated antitumor responses. This review will discuss the current knowledge regarding the pro- and antitumorigenic roles of neutrophils, leading to the concept of a confused state of neutrophil-driven pro-/antitumor responses.
Collapse
Affiliation(s)
- Leo Koenderman
- Dept. Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Nienke Vrisekoop
- Dept. Respiratory Medicine and Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
26
|
Marco S, Braconi C. Stroma-tology: Predicting recurrence in cholangiocarcinoma: Editorial on "Development and validation of a stromal-immune signature to predict prognosis in intrahepatic cholangiocarcinoma". Clin Mol Hepatol 2025; 31:323-326. [PMID: 39468841 PMCID: PMC11791558 DOI: 10.3350/cmh.2024.0932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 10/30/2024] Open
Affiliation(s)
- Sergi Marco
- University of Glasgow – Wolfson Wohl Cancer Research Centre, Scotland, UK
| | - Chiara Braconi
- University of Glasgow – Wolfson Wohl Cancer Research Centre, Scotland, UK
| |
Collapse
|
27
|
Wang Y, Liang S, Hong Q, Mu J, Wu Y, Li K, Li Y, Wu Y, Lou X, Xu D, Cui W. Construction of a neutrophil extracellular trap formation-related gene model for predicting the survival of lung adenocarcinoma patients and their response to immunotherapy. Transl Lung Cancer Res 2024; 13:3407-3425. [PMID: 39830760 PMCID: PMC11736607 DOI: 10.21037/tlcr-24-463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
Background Lung adenocarcinoma (LUAD) is associated with high morbidity and mortality rates. Increasing evidence indicates that neutrophil extracellular traps (NETs) play a critical role in tumor progression, metastasis and immunosuppression in the LUAD tumor microenvironment (TME). Nevertheless, the use of NET formation-related genes (NFRGs) to predict LUAD patient survival and response to immunotherapy has not been explored. Therefore, this study aimed to construct a NFRGs-based prognostic signature for stratifying LUAD patients and informing individualized management strategies. Methods The cell composition of the LUAD TME was investigated using the single-cell sequencing data in Single-Cell Lung Cancer Atlas (LuCA). NFRGs were identified to construct a prognostic signature based on The Cancer Genome Atlas (TCGA) cohort which was validated in the Gene Expression Omnibus (GEO) dataset. The univariate Cox and least absolute shrinkage and selection operator (LASSO) Cox regression models, receiver operating characteristic (ROC) and Brier Score were applied to assess the prognostic model. A nomogram was established to facilitate the clinical application of the risk score. The Estimation of STromal and Immune cells in MAlignant Tumor tissues (ESTIMATE) and Tumor Immune Dysfunction and Exclusion (TIDE) algorithm were utilized to assess the TME and predict immunotherapy response. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to quantify the expression levels of four NFRGs in LUAD paired tissue samples. Results Single‑cell RNA sequence analysis showed the importance of neutrophils in LUAD TME. We developed and validated a 4-NFRG (CAT, CTSG, ENO1, TLR2) prognostic signature based on TCGA and GEO cohorts, which stratified patients into high-risk and low-risk groups. Univariate and multivariate analyses showed that our risk model could independently predict the survival of LUAD patients. Patients in the low-risk group exhibited a more active immune microenvironment, lower TIDE scores, lower half-maximal inhibitory concentration (IC50) values and higher immune checkpoint molecule expression. Our risk signature could serve as a biomarker for predicting immunotherapeutic benefits. Conclusions We developed a novel prognostic signature for LUAD patients based on NFRGs and emphasized the critical role of this signature in predicting LUAD patient survival and immunotherapy response.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang Liang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Hong
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juwei Mu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxin Wu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kexin Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiling Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Wu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoying Lou
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Danfei Xu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Zeng F, Shao Y, Wu J, Luo J, Yue Y, Shen Y, Wang Y, Shi Y, Wu D, Cata JP, Yang S, Zhang H, Miao C. Tumor metastasis and recurrence: The role of perioperative NETosis. Cancer Lett 2024; 611:217413. [PMID: 39725150 DOI: 10.1016/j.canlet.2024.217413] [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: 04/29/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Although surgical resection of tumor mass remains the mainstay of curative therapeutic management for solid tumors, accumulating studies suggest that these procedures promote tumor recurrence and metastasis. Regarded as the first immune cells to fight against infectious or inflammatory insults from surgery, neutrophils along with their ability of neutrophil extracellular traps (NETs) production has attracted much attention. A growing body of evidence suggests that NETs promote cancer metastasis by stimulating various stages, including local invasion, colonization, and growth. Therefore, we discussed the mechanism of NETosis induced by surgical stress and tumor cells, and the contribution of NETs on tumor metastasis: aid in the tumor cell migration and proliferation, evasion of immune surveillance, circulating tumor cell adhesion and establishment of a metastatic niche. Lastly, we summarized existing NET-targeting interventions, offering recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Fu Zeng
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuwen Shao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jingyi Wu
- Department of Anesthesiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Jingwen Luo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yang Shen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yanghanzhao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yuxin Shi
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Dan Wu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Pujian Road 160, Shanghai, 200127, China.
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
29
|
Abdalsalam NMF, Ibrahim A, Saliu MA, Liu TM, Wan X, Yan D. MDSC: a new potential breakthrough in CAR-T therapy for solid tumors. Cell Commun Signal 2024; 22:612. [PMID: 39702149 DOI: 10.1186/s12964-024-01995-y] [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: 09/23/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has shown remarkable success in hematologic malignancies but has encountered challenges in effectively treating solid tumors. One major obstacle is the presence of the immunosuppressive tumor microenvironment (TME), which is mainly built by myeloid-derived suppressor cells (MDSCs). Recent studies have shown that MDSCs have a detrimental effect on CAR-T cells due to their potent immunosuppressive capabilities. Targeting MDSCs has shown promising results to enhance CAR-T immunotherapy in preclinical solid tumor models. In this review, we first highlight that MDSCs increase tumor proliferation, transition, angiogenesis and encourage circulating tumor cells (CTCs) extravasation leading to tumor progression and metastasis. Moreover, we describe the main characteristics of the immunosuppressive activities of MDSCs on T cells in TME. Most importantly, we summarize targeting therapeutic strategies of MDSCs in CAR-T therapies against solid tumors. These strategies include (1) therapeutic targeting of MDSCs through small molecule inhibitors and large molecule antibodies; (2) CAR-T targeting cancer cell antigen combination with MDSC modulatory agents; (3) cytokine receptor antigen-targeted CAR-T indirectly or directly targeting MDSCs reshapes TME; (4) modified natural killer (NK) cells expressing activating receptor directly targeting MDSCs; and (5) CAR-T directly targeting MDSC selective antigens. In the near future, we are expected to witness the improvement of CAR-T cell therapies for solid tumors by targeting MDSCs in clinical practice.
Collapse
Affiliation(s)
- Nada Mohamady Farouk Abdalsalam
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Abdulrahman Ibrahim
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Muhammad Auwal Saliu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Tzu-Ming Liu
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macao SAR, Taipa, China.
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100864, China.
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100864, China.
| |
Collapse
|
30
|
Hasan S, Awasthi P, Malik S, Dwivedi M. Immunotherapeutic strategies to induce inflection in the immune response: therapy for cancer and COVID-19. Biotechnol Genet Eng Rev 2024; 40:3571-3610. [PMID: 36411974 DOI: 10.1080/02648725.2022.2147661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
Cancer has agonized the human race for millions of years. The present decade witnesses biological therapeutics to combat cancer effectively. Cancer Immunotherapy involves the use of therapeutics for manipulation of the immune system by immune agents like cytokines, vaccines, and transfection agents. Recently, this therapeutic approach has got vast attention due to the current pandemic COVID-19 and has been very effective. Concerning cancer, immunotherapy is based on the activation of the host's antitumor response by enhancing effector cell number and the production of soluble mediators, thereby reducing the host's suppressor mechanisms by induction of a tumour killing environment and by modulating immune checkpoints. In the present era, immunotherapies have gained traction and momentum as a pedestal of cancer treatment, improving the prognosis of many patients with a wide variety of haematological and solid malignancies. Food supplements, natural immunomodulatory drugs, and phytochemicals, with recent developments, have shown positive trends in cancer treatment by improving the immune system. The current review presents the systematic studies on major immunotherapeutics and their development for the effective treatment of cancers as well as in COVID-19. The focus of the review is to highlight comparative analytics of existing and novel immunotherapies in cancers, concerning immunomodulatory drugs and natural immunosuppressants, including immunotherapy in COVID-19 patients.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University, Ranchi, Jharkhand, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| |
Collapse
|
31
|
Kang W, Wang C, Wang M, Liu M, Hu W, Liang X, Yang J, Zhang Y. A key regulator of tumor-associated neutrophils: the CXCR2 chemokine receptor. J Mol Histol 2024; 55:1051-1061. [PMID: 39269537 DOI: 10.1007/s10735-024-10260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024]
Abstract
In recent years, with the advance of research, the role of tumor-associated neutrophils (TANs) in tumors has become a research hotspot. As important effector cells in the innate immune system, neutrophils play a key role in the immune and inflammatory responses of the body. As the first line of defense against bacterial and fungal infections, neutrophils have the ability to kill invading pathogens. In the pathological state of malignant tumors, the phenotype of neutrophils is altered and has an important regulatory function in tumor development. The C-X-C motif chemokine receptor 2(CXCR2) is a key molecule that mediates the migration and aggregation signaling pathway of immune cells, especially neutrophils. This review focuses on the regulation of CXCR2 on TANs in the process of tumorigenesis and development, and emphasizes the application significance of CXCR2 inhibitors in blocking the migration of TANs to tumors.
Collapse
Affiliation(s)
- Wenyan Kang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Chengkun Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Minhui Wang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Meiqi Liu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Wei Hu
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Xiaoqiu Liang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China
| | - Juanli Yang
- Department of Gynecology, The First Affiliated Hospital, Hengyang School of Medicine, University of South China, Hengyang, 421001, Hunan, P.R. China.
| | - Yang Zhang
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 420001, Hunan, China.
| |
Collapse
|
32
|
Ulas A, Temel B, Kos FT. Comparison of Prognostic Values of Seven Immune Indexes in Advanced Non-Small-Cell Lung Cancer Treated with Nivolumab: How Effective Can They Be Regarding Our Treatment Decisions? MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1792. [PMID: 39596977 PMCID: PMC11596302 DOI: 10.3390/medicina60111792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/27/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: In this study, we evaluated the impact of seven immune indexes on treatment response and survival outcomes in advanced non-small-cell lung cancer (NSCLC) patients receiving second-line and subsequent nivolumab treatment under real-life conditions. Materials and Methods: The pan-immune inflammation value (PIV), systemic immune inflammation value (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR), derived neutrophil-to-lymphocyte ratio (d-NLR), and prognostic nutritional index (PNI) were calculated. All immune indexes were classified as low and high based on cut-off values. Kaplan-Meier and Cox hazard models were used for survival analysis. Results: The median follow-up was 22.0 months (6.0-96.0). The median overall survival (OS) was 30.0 months and the median progression-free survival (PFS) was 7.0 months. In the univariate analysis, comorbidity (p = 0.03) and nivolumab use for more than eight cycles (p < 0.0001) were associated with an increase in PFS, while smoking history (p < 0.005) and d-NLR (p < 0.05) were more effective regarding OS. Patients who received more than eight cycles of nivolumab had longer median PFS (4 vs. 19 months, p < 0.001) and OS (23 vs. 43 months, p < 0.001). We found longer median OS in the PLR (45.7 vs. 75.4 months; p = 0.05), PIV (53.0 vs. 66.4 months; p = 0.19), SII (50.0 vs. 71.9 vs. months, p = 0.19), and NLR (49.9 vs. 74.55 months, p = 0.10) indexes in nivolumab long-term users (high vs. low groups, respectively). In short-term users of nivolumab, only d-NLR median OS (high vs. low, 19 vs. 75.2 months, p = 0.07) was different. Complete and partial response rates to nivolumab treatment were higher in the PNI-high group (p = 0.04). Conclusions: In these real-life data, we determined that the PLR, PIV, SII, and NLR indexes were effective in the prognosis of patients who received PD1 inhibitor nivolumab for a long time, and the d-NLR index was effective in those who developed progression in a short time. We found that the PNI was effective in patients who responded well to ICI treatment.
Collapse
Affiliation(s)
- Arife Ulas
- Department of Medical Oncology, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey
| | - Beyza Temel
- Department of Internal Medicine, University of Health Sciences, Bursa City Education and Research Hospital, 16059 Bursa, Turkey;
| | - Fahriye Tugba Kos
- Department of Medical Oncology, University of Health Sciences, Ankara City Education and Research Hospital, 06290 Ankara, Turkey;
| |
Collapse
|
33
|
Du H, Xu E, Xu Y, Xue Q, Xu H, Song J. 3D DNAzyme Motor Nanodevice With Self-Powered FRET Amplifier and Self-Supplied H 2O 2 for Enhancing Human Neutrophil Elastase Profiling and Chemodynamic Therapy in Lung Tumor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406599. [PMID: 39348241 PMCID: PMC11600284 DOI: 10.1002/advs.202406599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/05/2024] [Indexed: 10/02/2024]
Abstract
The development of theragnostic nanosystems integrating FRET (fluorescence resonance energy transfer) imaging and chemodynamic therapy (CDT) for accurate diagnosis and effective treatment of lung tumors is still a big challenge. Herein, a peptide-assembled 3D DNAzyme motor nanodevice is engineered for a self-powered FRET amplifier profiling human neutrophil elastase (HNE) and self-supplied H2O2 enhancing CDT. The nanodevice is prepared by depositing AuNPs on ZIF-8, in which ZIF-8 co-loaded the lysosomal targeting peptide-modified copper peroxides (PCPs) and hairpins (H1, H2, and H3), AuNPs are co-labeled by DNAzyme-peptide (DP) conjugate and H3. In the tumor micro-environment, HNE driven 3D DNAzyme walker followed by an exponential amplification constructed by a synergistic cross-activation between hybridization chain reaction and DNAzyme, generating a self-powered FRET amplifier. The FRET amplifier specifically measures HNE with a sensitivity of 0.026 pM, and successfully images exogenous HNE in living cells and monitors HNE in mouse models. Moreover, the PCPs can target lysosomes, reducing lysosome escape. The self-supplying H2O2 undertaken by PCPs improves the Cu (II)-catalyzed Fenton-like reaction, effectively causing cell apoptosis to inhibit tumor growth. Significantly, the nanodevice successfully screens inhibitors and discriminates the HNE level in normal and lung cancer tissues, suggesting that the nanodevice provides an effective tool for the diagnosis and treatment of lung tumors.
Collapse
Affiliation(s)
- Huiyan Du
- Department of ChemistryLiaocheng UniversityLiaochengShandong252059P. R. China
| | - Ensheng Xu
- Department of ChemistryLiaocheng UniversityLiaochengShandong252059P. R. China
| | - Yihan Xu
- Department of ChemistryLiaocheng UniversityLiaochengShandong252059P. R. China
| | - Qingwang Xue
- Department of ChemistryLiaocheng UniversityLiaochengShandong252059P. R. China
| | - Hongxia Xu
- Department of Clinical LaboratoryThe Third People's Hospital of LiaochengLiaochengShandong252059P. R. China
| | - Jibin Song
- State Key Laboratory of Chemical Resource EngineeringCollege of ChemistryBeijing University of Chemical TechnologyBeijing10010P. R. China
| |
Collapse
|
34
|
Pan X, Wang Q, Sun B. Multifaceted roles of neutrophils in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189231. [PMID: 39615862 DOI: 10.1016/j.bbcan.2024.189231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/14/2024]
Abstract
Neutrophils, the most abundant leukocyte population in circulation, play a crucial role in detecting and responding to foreign cells, such as pathogens and tumor cells. However, the impact of neutrophils on cancer pathogenesis has been overlooked because of their short lifespan, terminal differentiation, and limited transcriptional activity. Within the tumor microenvironment (TME), neutrophils can be influenced by tumor cells or other stromal cells to acquire either protumor or antitumor properties via the cytokine environment. Despite progress in neutrophil-related research, a comprehensive understanding of tissue-specific neutrophil diversity and adaptability in the TME is still lacking, which poses a significant obstacle to the development of neutrophil-based cancer therapies. This review evaluated the current studies on the dual roles of neutrophils in cancer progression, emphasizing their importance in predicting clinical outcomes, and explored various approaches for targeting neutrophils in cancer treatment, including their potential synergy with cancer immunotherapy.
Collapse
Affiliation(s)
- Xueyin Pan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Hefei, Anhui, China; Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, China.
| |
Collapse
|
35
|
Wu G, Pan B, Shi H, Yi Y, Zheng X, Ma H, Zhao M, Zhang Z, Cheng L, Huang Y, Guo W. Neutrophils' dual role in cancer: from tumor progression to immunotherapeutic potential. Int Immunopharmacol 2024; 140:112788. [PMID: 39083923 DOI: 10.1016/j.intimp.2024.112788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
The tumor microenvironment (TME) is intricately associated with cancer progression, characterized by dynamic interactions among various cellular and molecular components that significantly impact the carcinogenic process. Notably, neutrophils play a crucial dual role in regulating this complex environment. These cells oscillate between promoting and inhibiting tumor activity, responding to a multitude of cytokines, chemokines, and tumor-derived factors. This response modulates immune reactions and affects the proliferation, metastasis, and angiogenesis of cancer cells. A significant aspect of their influence is their interaction with the endoplasmic reticulum (ER) stress responses in cancer cells, markedly altering tumor immunodynamics by modulating the phenotypic plasticity and functionality of neutrophils. Furthermore, neutrophil extracellular traps (NETs) exert a pivotal influence in the progression of malignancies by enhancing inflammation, metastasis, immune suppression, and thrombosis, thereby exacerbating the disease. In the realm of immunotherapy, checkpoint inhibitors targeting PD-L1/PD-1 and CTLA-4 among others have underscored the significant role of neutrophils in enhancing therapeutic responses. Recent research has highlighted the potential of using neutrophils for targeted drug delivery through nanoparticle systems, which precisely control drug release and significantly enhance antitumor efficacy. This review thoroughly examines the diverse functions of neutrophils in cancer treatment, emphasizing their potential in regulating immune therapy responses and as drug delivery carriers, offering innovative perspectives and profound implications for the development of targeted diagnostic and therapeutic strategies in oncology.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Binyang Pan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanjun Yi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaobin Zheng
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, China
| | - Lin Cheng
- Regenerative Medicine Institute, School of Medicine, National University of Ireland (NUI), Galway, Ireland.
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weigang Guo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Zhang Q, Zhang J, Gu H, Yang Y, Zhang H, Miao C. Perioperative NETosis and Cancer Progression: Current Evidence and Future Perspectives. Curr Oncol Rep 2024; 26:1169-1175. [PMID: 39012468 DOI: 10.1007/s11912-024-01573-y] [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] [Accepted: 06/24/2024] [Indexed: 07/17/2024]
Abstract
PURPOSE OF REVIEW The process of neutrophil extracellular traps (NETs) formation, called NETosis, is a peculiar death modality of neutrophils, which was first observed as an immune response against bacterial infection. However, an ongoing and exaggerated NETs formation may have adverse clinical consequences and even promote cancer progression. This review will discuss the complex relationship between NETosis and cancer progression. RECENT FINDINGS NETs exhibits cancer-promoting effects by causing cancer metastaisis and tumor-associated thrombosis. Many studies have found that many mechanisms are involved in the process, and the corresponding targets could be applied for cancer therapy. Although NETs may have anti-bacteria effects, it is necessary to inhibit an excessive NETs formation, mostly showing cancer-promoting effects. The contribution of NETs to cancer progression has gained a growing appreciation and the approaches to targeting NETs deposition exhibited beneficial effects both in primary and metastatic tumors, which, however, has been challenged by a recent finding demonstrating an opposite effect of NETs to suppress tumor growth via the activation of immune response against tumor. This seeming discrepancy reflects we are in the early stage of NETs study facing fundamental questions and a better understanding of the underlying mechanism is urgently needed.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Jing Zhang
- Department of Anesthesiology, Hebei General Hospital, Shijiazhuang, China
| | - Haiyun Gu
- Department of Anesthesiology, Rizhao People's Hospital, Shandong, China
| | - Yan Yang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
37
|
Zhang WH, Zhao Y, Zhang CR, Huang JC, Lyu SC, Lang R. Preoperative systemic inflammatory response index as a prognostic marker for distal cholangiocarcinoma after pancreatoduodenectomy. World J Gastrointest Surg 2024; 16:2910-2924. [PMID: 39351557 PMCID: PMC11438816 DOI: 10.4240/wjgs.v16.i9.2910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND The relationship between preoperative inflammation status and tumorigenesis as well as tumor progression is widely acknowledged. AIM To assess the prognostic significance of preoperative inflammatory biomarkers in patients with distal cholangiocarcinoma (dCCA) who underwent pancreatoduodenectomy (PD). METHODS This single-center study included 216 patients with dCCA after PD between January 1, 2011, and December 31, 2022. The individuals were categorized into two sets based on their systemic inflammatory response index (SIRI) levels: A low SIRI group (SIRI < 1.5, n = 123) and a high SIRI group (SIRI ≥ 1.5, n = 93). Inflammatory biomarkers were evaluated for predictive accuracy using receiver operating characteristic curves. Both univariate and multivariate Cox proportional hazards analyses were performed to estimate SIRI for overall survival (OS) and recurrence-free survival (RFS). RESULTS The study included a total of 216 patients, with 58.3% being male and a mean age of 65.6 ± 9.6 years. 123 patients were in the low SIRI group and 93 were in the high SIRI group after PD for dCCA. SIRI had an area under the curve value of 0.674 for diagnosing dCCA, showing better performance than other inflammatory biomarkers. Multivariate analysis indicated that having a SIRI greater than 1.5 independently increased the risk of dCCA following PD, leading to lower OS [hazard ratios (HR) = 1.868, P = 0.006] and RFS (HR = 0.949, P < 0.001). Additionally, survival analysis indicated a significantly better prognosis for patients in the low SIRI group (P < 0.001). CONCLUSION It is determined that a high SIRI before surgery is a significant risk factor for dCCA after PD.
Collapse
Affiliation(s)
- Wen-Hui Zhang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| | - Cheng-Run Zhang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| | - Jin-Can Huang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| | - Shao-Cheng Lyu
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing 100020, China
| |
Collapse
|
38
|
Zhang Y, Wu J, Chen W, Liang X. Pretreatment System Inflammation Response Index (SIRI) is a Valuable Marker for Evaluating the Efficacy of Neoadjuvant Therapy in Breast Cancer Patients. Int J Gen Med 2024; 17:4359-4368. [PMID: 39346633 PMCID: PMC11439349 DOI: 10.2147/ijgm.s478000] [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: 05/12/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Objective Immune inflammatory response are involved in the development and progression of cancer. However, there are still inconsistent research results on the value of peripheral blood inflammatory indicators for evaluating the efficacy of neoadjuvant therapy (NAT) in breast cancer. The purpose of this study was to investigate the relationship between pretreatment systemic immune inflammatory response index (SII), systemic inflammatory response index (SIRI), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR) and NAT efficacy in breast cancer. Methods A retrospective analysis was performed on 326 patients with breast cancer who underwent NAT at Meizhou People's Hospital from November 2017 to October 2023. Clinicopathological data was collected, including gender, age, body mass index (BMI), hypertension, diabetes mellitus, family history of cancer, TNM stage, and the molecular subtypes of breast cancer. The optimal cutoff values of SII, SIRI, NLR, PLR, and LMR were calculated using receiver operating characteristic (ROC) curve, and the relationship between inflammatory indexes and other clinicopathological features and the efficacy of NAT was analyzed. Results In this study, 162 (49.7%) breast cancer patients did not respond to NAT and 164 (50.3%) patients responded to NAT. The levels of SII (p=0.002), SIRI (p<0.001), and NLR (p=0.006) in patients who responded to NAT were significantly higher than those in patients who did not. When the efficacy of NAT was considered as the endpoint of SII, SIRI, and NLR, the critical value of the SII, SIRI, and NLR was 572.53 (under the ROC curve (AUC)=0.598), 0.745 (AUC=0.630), and 2.325 (AUC=0.588), respectively. Logistic regression analysis showed that a high SIRI level (≥0.745/<0.745, OR: 2.447, 95% CI: 1.375-4.357, p=0.002) was an independent factor associated with the efficacy of NAT in breast cancer patients. Conclusion High SIRI levels (≥0.745) may be an independent factor associated with the efficacy of NAT in patients with breast cancer.
Collapse
Affiliation(s)
- Yunuo Zhang
- Department of Medical Oncology, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Jingna Wu
- Department of Medical Oncology, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Weiming Chen
- Data Center, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Xinhong Liang
- Radiology Department, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| |
Collapse
|
39
|
Zhang M, Qin H, Wu Y, Gao Q. Complex role of neutrophils in the tumor microenvironment: an avenue for novel immunotherapies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0192. [PMID: 39297568 PMCID: PMC11523270 DOI: 10.20892/j.issn.2095-3941.2024.0192] [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: 05/24/2024] [Accepted: 08/26/2024] [Indexed: 11/01/2024] Open
Abstract
Neutrophils, which originate from the bone marrow and are characterized by a segmented nucleus and a brief lifespan, have a crucial role in the body's defense against infections and acute inflammation. Recent research has uncovered the complex roles of neutrophils as regulators in tumorigenesis, during which neutrophils exhibit a dualistic nature that promotes or inhibits tumor progression. This adaptability is pivotal within the tumor microenvironment (TME). In this review, we provide a comprehensive characterization of neutrophil plasticity and heterogeneity, aiming to illuminate current research findings and discuss potential therapeutic avenues. By delineating the intricate interplay of neutrophils in the TME, this review further underscores the urgent need to understand the dual functions of neutrophils with particular emphasis on the anti-tumor effects to facilitate the development of effective therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Haokai Qin
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Yingcheng Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, State Key Laboratory of Genetic Engineering, Human Phenome Institute, Fudan University, Shanghai 200433, China
| |
Collapse
|
40
|
Wen J, Liu D, Zhu H, Shu K. Microenvironmental regulation of tumor-associated neutrophils in malignant glioma: from mechanism to therapy. J Neuroinflammation 2024; 21:226. [PMID: 39285276 PMCID: PMC11406851 DOI: 10.1186/s12974-024-03222-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/03/2024] [Indexed: 09/19/2024] Open
Abstract
Glioma is the most common primary intracranial tumor in adults, with high incidence, recurrence, and mortality rates. Tumor-associated neutrophils (TANs) are essential components of the tumor microenvironment (TME) in glioma and play a crucial role in glioma cell proliferation, invasion and proneural-mesenchymal transition. Besides the interactions between TANs and tumor cells, the multi-dimensional crosstalk between TANs and other components within TME have been reported to participate in glioma progression. More importantly, several therapies targeting TANs have been developed and relevant preclinical and clinical studies have been conducted in cancer therapy. In this review, we introduce the origin of TANs and the functions of TANs in malignant behaviors of glioma, highlighting the microenvironmental regulation of TANs. Moreover, we focus on summarizing the TANs-targeted methods in cancer therapy, aiming to provide insights into the mechanisms and therapeutic opportunities of TANs in the malignant glioma microenvironment.
Collapse
Affiliation(s)
- Jiayi Wen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Dan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Kai Shu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
41
|
Tharani PV, Rao KVB. A comprehensive review on microbial diversity and anticancer compounds derived from seaweed endophytes: a pharmacokinetic and pharmacodynamic approach. Arch Microbiol 2024; 206:403. [PMID: 39276253 DOI: 10.1007/s00203-024-04121-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/17/2024] [Accepted: 08/25/2024] [Indexed: 09/16/2024]
Abstract
Seaweed endophytes are a rich source of microbial diversity and bioactive compounds. This review provides a comprehensive analysis of the microbial diversity associated with seaweeds and their interaction between them. These diverse bacteria and fungi have distinct metabolic pathways, which result in the synthesis of bioactive compounds with potential applications in a variety of health fields. We examine many types of seaweed-associated microorganisms, their bioactive metabolites, and their potential role in cancer treatment using a comprehensive literature review. By incorporating recent findings, we hope to highlight the importance of seaweed endophytes as a prospective source of novel anticancer drugs and promote additional studies in this area. We also investigate the pharmacokinetic and pharmacodynamic profiles of these bioactive compounds because understanding their absorption, distribution, metabolism, excretion (ADMET), and toxicity profiles is critical for developing bioactive compounds with anticancer potential into effective cancer drugs. This knowledge ensures the safety and efficacy of proposed medications prior to clinical trials. This study not only provides promise for novel and more effective treatments for cancer with fewer side effects, but it also emphasizes the necessity of sustainable harvesting procedures and ethical considerations for protecting the delicate marine ecology during bioprospecting activities.
Collapse
Affiliation(s)
- P V Tharani
- Marine Biotechnology Laboratory, Department of Bio-Medical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - K V Bhaskara Rao
- Marine Biotechnology Laboratory, Department of Bio-Medical Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
42
|
Ding P, Yang J, Wu J, Wu H, Sun C, Chen S, Yang P, Tian Y, Guo H, Liu Y, Meng L, Zhao Q. Combined systemic inflammatory immune index and prognostic nutrition index as chemosensitivity and prognostic markers for locally advanced gastric cancer receiving neoadjuvant chemotherapy: a retrospective study. BMC Cancer 2024; 24:1014. [PMID: 39148031 PMCID: PMC11328362 DOI: 10.1186/s12885-024-12771-z] [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] [Accepted: 08/06/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND The prognosis nutritional index (PNI) and the systemic inflammatory immunological index (SII) are characteristic indicators of the nutritional state and the systemic inflammatory response, respectively. However, there is an unknown combined effect of these indicators in the clinic. Therefore, the practicality of using the SII-PNI score to predict prognosis and tumor response of locally advanced gastric cancer (LAGC) following chemotherapy was the main focus of this investigation. METHODS We retrospectively analyzed 181 patients with LAGC who underwent curative resection after neoadjuvant chemotherapy in a prospective study (NCT01516944). We divided these patients into tumour regression grade(TRG) 3 and non-TRG3 groups based on tumor response (AJCC/CAP guidelines). The SII and PNI were assessed and confirmed the cut-off values before treatment. The SII-PNI values varied from 0 to 2, with 2 being the high SII (≥ 471.5) as well as low PNI (≤ 48.6), a high SII or low PNI is represented by a 1 and neither is represented by a 0, respectively. RESULTS 51 and 130 samples had TRG3 and non-TRG3 tumor responses respectively. Patients with TRG3 had substantially higher SII-PNI scores than those without TRG3 (p < 0.0001). Patients with greater SII-PNI scores had a poorer prognosis (p < 0.0001). The SII-PNI score was found to be an independent predictor of both overall survival (HR = 4.982, 95%CI: 1.890-10.234, p = 0.001) and disease-free survival (HR = 4.763, 95%CI: 1.994-13.903, p = 0.001) in a multivariate analysis. CONCLUSION The clinical potential and accuracy of low-cost stratification based on SII-PNI score in forecasting tumor response and prognosis in LAGC is satisfactory.
Collapse
Affiliation(s)
- Ping'an Ding
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxuan Yang
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Haotian Wu
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Chenyu Sun
- AMITA Health Saint Joseph Hospital Chicago, 2900 N. Lake Shore Drive, Chicago, IL, 60657, USA
| | - Shuya Chen
- Newham University Hospital, Glen Road, Plaistow, London, E13 8SL, United Kingdom
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Yuan Tian
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Yang Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China
| | - Lingjiao Meng
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Key Laboratory of Tumour Prevention, Precision Diagnosis and Treatment in Hebei Province, Shijiazhuang, 050011, China.
| | - Qun Zhao
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
- Big Data Analysis and Mining Application for Precise Diagnosis and Treatment of Gastric Cancer Hebei Provincial Engineering Research Center, Shijiazhuang, 050011, China.
| |
Collapse
|
43
|
Noguchi A, Numata Y, Sugawara T, Miura H, Konno K, Adachi Y, Yamaguchi R, Ishida M, Kokumai T, Douchi D, Miura T, Ariake K, Nakayama S, Maeda S, Ohtsuka H, Mizuma M, Nakagawa K, Morikawa H, Akatsuka J, Maeda I, Unno M, Yamamoto Y, Furukawa T. Deep learning predicts the 1-year prognosis of pancreatic cancer patients using positive peritoneal washing cytology. Sci Rep 2024; 14:17059. [PMID: 39095474 PMCID: PMC11297136 DOI: 10.1038/s41598-024-67757-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Peritoneal washing cytology (CY) in patients with pancreatic cancer is mainly used for staging; however, it may also be used to evaluate the intraperitoneal status to predict a more accurate prognosis. Here, we investigated the potential of deep learning of CY specimen images for predicting the 1-year prognosis of pancreatic cancer in CY-positive patients. CY specimens from 88 patients with prognostic information were retrospectively analyzed. CY specimens scanned by the whole slide imaging device were segmented and subjected to deep learning with a Vision Transformer (ViT) and a Convolutional Neural Network (CNN). The results indicated that ViT and CNN predicted the 1-year prognosis from scanned images with accuracies of 0.8056 and 0.8009 in the area under the curve of the receiver operating characteristic curves, respectively. Patients predicted to survive 1 year or more by ViT showed significantly longer survivals by Kaplan-Meier analyses. The cell nuclei found to have a negative prognostic impact by ViT appeared to be neutrophils. Our results indicate that AI-mediated analysis of CY specimens can successfully predict the 1-year prognosis of patients with pancreatic cancer positive for CY. Intraperitoneal neutrophils may be a novel prognostic marker and therapeutic target for CY-positive patients with pancreatic cancer.
Collapse
Affiliation(s)
- Aya Noguchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yasushi Numata
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Takanori Sugawara
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Hiroshu Miura
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Kaori Konno
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Yuzu Adachi
- Department of Pathology, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Ruri Yamaguchi
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Masaharu Ishida
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takashi Kokumai
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Daisuke Douchi
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Takayuki Miura
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kyohei Ariake
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Shun Nakayama
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Shimpei Maeda
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hideo Ohtsuka
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Masamichi Mizuma
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Kei Nakagawa
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Hiromu Morikawa
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Jun Akatsuka
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
- Department of Urology, Nippon Medical School Hospital, Tokyo, 113-8603, Japan
| | - Ichiro Maeda
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
- Department of Pathology, Kitasato University Kitasato Institute Hospital, Tokyo, 108-0072, Japan
- Department of Pathology, Kitasato University School of Medicine, Kanagawa, 252-0373, Japan
| | - Michiaki Unno
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Yoichiro Yamamoto
- Pathology Informatics Team, RIKEN Center for Advanced Intelligence Project, Nihonbashi 1-chome Mitsui Bldg. 15F, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan.
- Mathematical Intelligence for Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan.
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryomachi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
44
|
Feng X, Sun Z, Huang Y, Zhang Y, Chen Y, Deng B. A Low Systemic Inflammatory Response Index Is Associated With Improved Survival in Adenoid Cystic Carcinoma Patients. J Oral Maxillofac Surg 2024; 82:999-1007. [PMID: 38615695 DOI: 10.1016/j.joms.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Predicting the long-term survival in adenoid cystic carcinoma (ACC) patients remains challenging. Inflammatory cell-based indices are emerging as prognostic indicators of oncology. PURPOSE This study aimed to determine the associations between the preoperative systemic inflammatory response index (SIRI) and the systemic immunoinflammatory index (SII) and the 10-year survival rates in patients with ACC of the head and neck (ACCHN). STUDY DESIGN, SETTING, SAMPLE This retrospective cohort study comprised ACCHN patients treated at the Chinese People's Liberation Army General Hospital between November 2003 and December 2020. PREDICTOR VARIABLE The inflammatory response, assessed using the SIRI and SII, was the predictor variable. The optimal cutoff values were based on the maximum Youden index values (sensitivity + specificity-1). The patients were divided into two groups each, based on the SIRI (low, ≤ 0.15) and (high, > 0.15), and SII (low, ≤ 562.8 and high, > 562.8) values. MAIN OUTCOME VARIABLE(S) Overall survival (OS), or the number of days, weeks, or months between treatment initiation and death (or the last follow-up date), was the primary outcome variable. COVARIATES The covariates were classified as demographic (age, gender, body mass index), medical (hypertension, diabetes), inflammatory (neutrophils, lymphocytes, monocytes, platelets, lymphocyte-monocyte ratio, platelet-lymphocyte ratio, neutrophil-lymphocyte ratio), and perioperative (tumor stage, lymph node metastasis, tumor size, treatment type). ANALYSES Descriptive, univariate, and multivariate Cox proportional risk regression analyses were performed to determine whether the SIRI and SII were independent prognostic factors for OS. Kaplan-Meier survival curves and log-rank tests were used to determine their associations with the OS. RESULTS The study sample comprised 162 patients (mean age, 52 ± 14; males, 39.5%). The median follow-up time was 6.81 ± 0.23, and the 10-year OS rate was 7.68 ± 0.25. The low and high SIRI groups comprised 109 and 53 patients, while the low and high SII groups comprised 116 and 46 patients, respectively. SIRI was identified as a prognostic factor (P < .01; hazard ratio, 2.45; 95% confidence interval, 1.35-4.45). CONCLUSION AND RELEVANCE The SIRI has the advantages of reproducibility, convenience, noninvasiveness, and affordability, making it a promising prognostic inflammatory index for patients with ACCHN.
Collapse
Affiliation(s)
- Xuanqi Feng
- Graduate Student, Medical School of Chinese PLA, Beijing, China
| | - Zhongyang Sun
- Resident Doctor, Department of Infectious Disease Management, The Second Affiliated Hospital of Xiamen Medical College (Xiamen Second Hospital), Xiamen, China
| | - Yang Huang
- Dentistry, Attending Physician, Department of Stomatology, The First Medical Center, Chinese PLA General, Medical School of Chinese PLA, Beijing, China
| | - Yu Zhang
- Graduate Student, Medical School of Chinese PLA, Beijing, China
| | - Yanru Chen
- Graduate Student, Medical School of Chinese PLA, Beijing, China
| | - Bin Deng
- Dentistry, Associate Chief Physician, Department of Stomatology, Medical School of Chinese PLA, The First Medical Center, Chinese PLA General, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
45
|
Rodríguez-Bejarano OH, Parra-López C, Patarroyo MA. A review concerning the breast cancer-related tumour microenvironment. Crit Rev Oncol Hematol 2024; 199:104389. [PMID: 38734280 DOI: 10.1016/j.critrevonc.2024.104389] [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: 03/11/2024] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Breast cancer (BC) is currently the most common malignant tumour in women and one of the leading causes of their death around the world. New and increasingly personalised diagnostic and therapeutic tools have been introduced over the last few decades, along with significant advances regarding the study and knowledge related to BC. The tumour microenvironment (TME) refers to the tumour cell-associated cellular and molecular environment which can influence conditions affecting tumour development and progression. The TME is composed of immune cells, stromal cells, extracellular matrix (ECM) and signalling molecules secreted by these different cell types. Ever deeper understanding of TME composition changes during tumour development and progression will enable new and more innovative therapeutic strategies to become developed for targeting tumours during specific stages of its evolution. This review summarises the role of BC-related TME components and their influence on tumour progression and the development of resistance to therapy. In addition, an account on the modifications in BC-related TME components associated with therapy is given, and the completed or ongoing clinical trials related to this topic are presented.
Collapse
Affiliation(s)
- Oscar Hernán Rodríguez-Bejarano
- Health Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222#55-37, Bogotá 111166, Colombia; Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia; PhD Programme in Biotechnology, Faculty of Sciences, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia
| | - Carlos Parra-López
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia.
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50#26-20, Bogotá 111321, Colombia; Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Carrera 45#26-85, Bogotá 111321, Colombia.
| |
Collapse
|
46
|
Gerashchenko T, Frolova A, Patysheva M, Fedorov A, Stakheyeva M, Denisov E, Cherdyntseva N. Breast Cancer Immune Landscape: Interplay Between Systemic and Local Immunity. Adv Biol (Weinh) 2024; 8:e2400140. [PMID: 38727796 DOI: 10.1002/adbi.202400140] [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: 03/12/2024] [Revised: 04/16/2024] [Indexed: 07/13/2024]
Abstract
Breast cancer (BC) is one of the most common malignancies in women worldwide. Numerous studies in immuno-oncology and successful trials of immunotherapy have demonstrated the causal role of the immune system in cancer pathogenesis. The interaction between the tumor and the immune system is known to have a dual nature. Despite cytotoxic lymphocyte activity against transformed cells, a tumor can escape immune surveillance and leverage chronic inflammation to maintain its own development. Research on antitumor immunity primarily focuses on the role of the tumor microenvironment, whereas the systemic immune response beyond the tumor site is described less thoroughly. Here, a comprehensive review of the formation of the immune profile in breast cancer patients is offered. The interplay between systemic and local immune reactions as self-sustaining mechanism of tumor progression is described and the functional activity of the main cell populations related to innate and adaptive immunity is discussed. Additionally, the interaction between different functional levels of the immune system and their contribution to the development of the pro- or anti-tumor immune response in BC is highlighted. The presented data can potentially inform the development of new immunotherapy strategies in the treatment of patients with BC.
Collapse
Affiliation(s)
- Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anastasia Frolova
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| | - Marina Patysheva
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Anton Fedorov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Marina Stakheyeva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
| | - Nadezda Cherdyntseva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Researc, Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk, 634009, Russia
- Tomsk State University, 36 Lenin Ave., Tomsk, 634050, Russia
| |
Collapse
|
47
|
Hu X, Wang N, Gao F, Ge S, Lin M, Zhang X, Li T, Li T, Xu C, Huang C, Liang G, Shang W, Xiang F, Feng Y. Prognostic significance of serum Chemerin and neutrophils levels in patients with oral squamous cell carcinoma. Heliyon 2024; 10:e32393. [PMID: 38975159 PMCID: PMC11225754 DOI: 10.1016/j.heliyon.2024.e32393] [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: 01/27/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Objectives Chemerin, as a novel multifunctional adipokine, is proposed to be involved in high cancer risk and mortality. The present study was aimed to evaluate the prognostic value of serum Chemerin and neutrophils in patients with oral squamous cell carcinoma (OSCC). Materials and methods 120 patients with OSCC were included in this prospective cohort study. The levels of serum Chemerin were measured by enzyme-linked immunosorbent assay (ELISA). We also explored the possible effects of Chemerin on neutrophils' chemokines in OSCC using a real-time PCR, western blotting. Results Levels of serum Chemerin, neutrophils and NLR were significantly higher among non-survivors compared to survivors of OSCC (both P < 0.05). Higher serum Chemerin levels were associated with advanced TNM stage, lymph node metastasis, differentiation and tumor recurrence (both P < 0.05). Serum Chemerin levels correlated with neutrophils and NLR levels (r = 0.708, r = 0.578, both P < 0.05). Based on ROC analysis, Chemerin + NLR predicted OSCC patient mortality with 81.54 % sensitivity and 87.27 % specificity, with an AUC of 0.8898. In a Kaplan-Meier analysis, high serum Chemerin levels, high neutrophil levels and high NLR levels were associated with shorter overall and disease-free survival (both P < 0.05). A univariate and multivariate Cox regression analysis showed that serum Chemerin and neutrophils were independent risk factors for OSCC. (both P < 0.05). QRT-PCR and western blotting results showed that Chemerin upregulated the expression of chemokines IL-17 and CXCL-5 in neutrophils (both P < 0.05). Conclusions Our study suggests that measurement of serum Chemerin and neutrophils might be a useful diagnostic and prognostic biomarker for OSCC patients. Chemerin may promote neutrophils infiltration in OSCC through upregulation of chemokines IL17 and CXCL-5.
Collapse
Affiliation(s)
- Xiaoyuan Hu
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Ning Wang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Fei Gao
- Deparment of Pathology, West China Hospital, Sichuan University, Sichuan, 610041, China
| | - Shengyou Ge
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| | - Mei Lin
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Xuan Zhang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Tongtong Li
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Tao Li
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Changting Xu
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Caixiu Huang
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Guicai Liang
- Biological Therapy Center, The Third Affiliated Hospital of Kunming Medical University, Kunzhou Road No. 519, Kunming, Yunnan, 650118, China
| | - Wei Shang
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| | - Fenggang Xiang
- Department of Pathology, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Yuanyong Feng
- Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Jiangsu Road No.16, Qingdao, Shandong, 266003, China
| |
Collapse
|
48
|
Zhu W, Zhao S, Cheng X, Wu C, Liu Z, Huang J. Chemokine‑ and chemokine receptor-based subtypes predict prognosis, immunotherapy and chemotherapy response in colorectal cancer patients. Int Immunopharmacol 2024; 134:112172. [PMID: 38703566 DOI: 10.1016/j.intimp.2024.112172] [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: 03/12/2024] [Revised: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND The clinical significance and comprehensive characteristics of chemokines and chemokine receptors in colorectal cancer (CRC) have not been previously reported. Our study aims to investigate the expression profiles of chemokines and chemokine receptors, as well as establish subtypes in CRC. METHODS 1009 CRC samples were enrolled in our study. Consensus unsupervised clustering analysis was conducted to establish subtypes, and a risk score model was developed using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses. 36 pairs of tissue specimens of CRC patients and two CRC cell lines were used to validate the subtypes and risk score in vitro. Quantitative real-time PCR and western blotting were employed to validate mRNA and protein expression levels, respectively. Flow cytometry was utilized for analyzing cell apoptosis, while cell viability assay and EdU assay were conducted to assess cell proliferation ability. RESULTS The Cluster B group shares similarities with the low-risk group in terms of exhibiting a higher level of immune cell infiltration and belonging to hot tumor. Patients CRC in the Cluster B group demonstrate a more favorable prognosis and exhibit better response to immunotherapy and chemotherapy. On the other hand, the Cluster A group resembles the high-risk group as it displays lower levels of immune cell infiltration, indicating a cold tumor phenotype. CRC patients in the Cluster A group have poorer prognoses and show less therapeutic efficacy towards immunotherapy and chemotherapy. Furthermore, we utilized a total of 36 pairs of tissue samples obtained from patients with CRC, along with two CRC cell lines for validation in vitro. This comprehensive approach further enhances the scientific validity and reliability of the identified subtypes and risk score in their ability to predict prognosis, response to immunotherapy, and response to chemotherapy among CRC patients. CONCLUSION We first established robust prognostic subtypes based on chemokines and chemokine receptors, which could potentially serve as a novel biomarker for guiding individualized treatment in patients with CRC undergoing immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Wenjie Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shimin Zhao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiufeng Cheng
- Department of Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Changlei Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China; Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
49
|
Yu Z, Xiong Z, Ma J, Du P, Wang S, Liu J, Cao Y, Yang Y. Prognostic and clinicopathological significance of systemic immune-inflammation index in upper tract urothelial carcinoma: a meta-analysis of 3911 patients. Front Oncol 2024; 14:1342996. [PMID: 38947894 PMCID: PMC11211359 DOI: 10.3389/fonc.2024.1342996] [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: 11/22/2023] [Accepted: 05/24/2024] [Indexed: 07/02/2024] Open
Abstract
Background Systemic immune-inflammation index (SII), a novel prognostic indicator, is being more commonly utilized in different types of cancer. This research project involved combining information from previously published studies to examine how pre-treatment SII can predict outcomes in individuals with upper tract urothelial carcinoma (UTUC). Further examination of the correlation between SII and clinical and pathological features in UTUC. Methods We thoroughly chose pertinent articles from various databases including PubMed, Embase, Cochrane Library, Web of Science, Chinese National Knowledge Infrastructure (CNKI), WanFang database, and Chinese Scientific Journal Database (VIP) until March 10, 2022.The data collected was analyzed using Stata 17.0 software (Stat Corp, College Station, TX). Subsequently, the impact of SII on the survival outcomes of UTUC patients was evaluated by combining HRs with 95% confidence intervals. Results Six included studies were finally confirmed, including 3911 UTUC patients in seven cohorts. The results showed that high SII before treatment predicted poor overall survival (HR =1.87, 95%CI 1.20-2.92, p=0.005), cancer specific survival (HR=2.70, 95%CI 1.47-4.96, P=0.001), and recurrence-free survival (HR =1.52, 95%CI 1.12-2.07, P=0.007). And the elevated SII may be related to LVI (present vs. absent) (OR=0.83, 95% CI=0.71-0.97, p=0.018), pT stage (pT ≥3 vs. < 3) (OR=1.82, 95% CI=1.21-2.72, p=0.004), and pN stage (N+ vs. N0) (OR=3.27, 95% CI=1.60-6.71, p=0.001). Conclusion A comprehensive analysis of all included articles in this study showed that higher pretreatment SII was related to poorer survival outcomes and adverse pathological features independently. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022316333.
Collapse
Affiliation(s)
- Ziyi Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhencheng Xiong
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Jinchao Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Peng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Shuo Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jia Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yudong Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Urological Department, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
50
|
Ksienski D, Truong PT, Bone JN, Egli S, Clarkson M, Patterson T, Lesperance M, Lakkunarajah S. Advanced cutaneous squamous cell carcinoma: Impact of age on the safety and efficacy of cemiplimab and the prognostic significance of blood biomarkers. J Geriatr Oncol 2024; 15:101789. [PMID: 38710153 DOI: 10.1016/j.jgo.2024.101789] [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: 11/13/2023] [Revised: 03/23/2024] [Accepted: 04/26/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Age-related differences in the safety profile of cemiplimab for patients with locally advanced or metastatic cutaneous squamous cell carcinoma (cSCC) have not been well described. We investigated the association of increasing age with immune related adverse events (irAE) from cemiplimab, efficacy outcomes, and the prognostic significance of pre-treatment blood biomarkers in contemporary practice. MATERIALS AND METHODS Patients starting first-line cemiplimab for locally advanced or metastatic cSCC at British Columbia Cancer between April 2019 and January 2023 were identified. Landmark four-month logistic regression analysis compared the odds of developing irAE or sequelae amongst patients aged <75 years to those aged 75-84 or ≥ 85. Objective responses were determined using Response Evaluation Criteria in Solid Tumors version 1.1. Univariable Cox proportional hazard (PH) regression modelling of factors associated with overall survival (OS) was performed. RESULTS Of 106 patients, the proportions aged <75, 75-84, and ≥ 85 years were 34%, 45%, and 21%, respectively. Overall, the proportion of patients with irAE ≥ grade 3, cemiplimab discontinuation, and hospitalization for immune toxicity was 27.4%, 31.1%, and 11.3%, respectively. There was no clear association between age and the odds of high grade irAE. However, increased odds of cemiplimab discontinuation was observed in patients aged 75-84 years (p = 0.05). Patients ≥85 years had increased hospitalizations due to irAE (OR = 5.00, 95% CI = 0.97-37.52) with two treatment-related deaths. Objective responses were similar across age cohorts (50.0%, 60.4%, and 54.5%) but progressive disease was higher in the age ≥ 85 group (22.2%, 18.8%, and 31.8%). On Cox PH regression analysis, age ≥ 85 years (vs. <75), Eastern Cooperative Oncology Group performance status 2-3 (vs. 0-1), and neutrophil to lymphocyte ratio (NLR) ≥7.80 (vs. <7.80) were associated with shorter survival. DISCUSSION While the odds of high grade irAE were similar across age groups, significant age-related differences in treatment discontinuation and hospitalization due to immune toxicity were observed. Despite a higher incidence of primary progression and shorter OS in the oldest cohort, cemiplimab yielded robust objective responses regardless of age. Higher pre-treatment NLR was associated with shorter survival and the cut-point identified requires further study.
Collapse
MESH Headings
- Humans
- Aged
- Aged, 80 and over
- Male
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Skin Neoplasms/drug therapy
- Skin Neoplasms/blood
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/blood
- Age Factors
- Prognosis
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Biomarkers, Tumor/blood
- British Columbia
- Retrospective Studies
- Middle Aged
Collapse
Affiliation(s)
- Doran Ksienski
- BC Cancer-Victoria, British Columbia, Canada; University of British Columbia, Victoria, British Columbia, Canada.
| | - Pauline T Truong
- BC Cancer-Victoria, British Columbia, Canada; University of British Columbia, Victoria, British Columbia, Canada
| | - Jeffrey N Bone
- Biostatistics, Clinical Research Support Unit, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Sarah Egli
- BC Cancer-Victoria, British Columbia, Canada
| | | | | | - Mary Lesperance
- University of Victoria, Department of Mathematics and Statistics, Victoria, British Columbia, Canada
| | | |
Collapse
|