1
|
Szukiewicz D. CX3CL1 (Fractalkine)-CX3CR1 Axis in Inflammation-Induced Angiogenesis and Tumorigenesis. Int J Mol Sci 2024; 25:4679. [PMID: 38731899 PMCID: PMC11083509 DOI: 10.3390/ijms25094679] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
2
|
Pourjamal N, Yazdi N, Halme A, Joncour VL, Laakkonen P, Saharinen P, Joensuu H, Barok M. Comparison of trastuzumab emtansine, trastuzumab deruxtecan, and disitamab vedotin in a multiresistant HER2-positive breast cancer lung metastasis model. Clin Exp Metastasis 2024; 41:91-102. [PMID: 38367127 PMCID: PMC10973002 DOI: 10.1007/s10585-024-10278-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
Human epidermal growth factor 2 (HER2)-positive breast cancer with lung metastases resistant to targeted agents is a common therapeutic challenge. Absence of preclinical lung metastasis models that are resistant to multiple anti-HER2 targeted drugs hampers the development of novel therapies. We established a novel HER2-positive breast cancer cell line (L-JIMT-1) with a high propensity to form lung metastases from the parenteral JIMT-1 cell line by injecting JIMT-1 cells into immunodeficient SCID mice. Lung metastases developed in all mice injected with L-JIMT-1 cells, and more rapidly and in greater numbers compared with the parental JIMT-1 cells. L-JIMT-1 cells expressed more epidermal growth factor receptor and HER2 than JIMT-1 cells. L-JIMT-1 cells were resistant to all five tyrosine kinase inhibitors tested in vitro (afatinib, erlotinib, lapatinib, sapitinib, and tucatinib). When we compared JIMT-1 and L-JIMT-1 sensitivity to three HER2-targeting antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), and disitamab vedotin (DV) in vitro, JIMT-1 cells were resistant T-DXd, partially sensitive to T-DM1, and sensitive to DV, while L-JIMT-1 cells were resistant to both T-DM1 and T-DXd, but moderately sensitive to DV. In a mouse model, all three ADCs inhibited the growth of L-JIMT-1 lung metastases compared to a vehicle, but DV and T-DXd more strongly than T-DM1, and DV treatment led to the smallest tumor burden. The L-JIMT breast cancer lung metastasis model developed may be useful in the evaluation of anti-cancer agents for multiresistant HER2-positive advanced breast cancer.
Collapse
Affiliation(s)
- Negar Pourjamal
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Laboratory of Molecular Oncology, University of Helsinki, Helsinki, Finland
| | - Narjes Yazdi
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Laboratory of Molecular Oncology, University of Helsinki, Helsinki, Finland
| | - Aleksi Halme
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vadim Le Joncour
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
- Laboratory Animal Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Pipsa Saharinen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, University of Helsinki, Helsinki, Finland
| | - Heikki Joensuu
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Laboratory of Molecular Oncology, University of Helsinki, Helsinki, Finland
- Department of Oncology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Mark Barok
- Helsinki University Hospital and University of Helsinki, Helsinki, Finland.
- Laboratory of Molecular Oncology, University of Helsinki, Helsinki, Finland.
- Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
3
|
Sivanesan E, Sanchez KR, Zhang C, He SQ, Linderoth B, Stephens KE, Raja SN, Guan Y. Spinal Cord Stimulation Increases Chemoefficacy and Prevents Paclitaxel-Induced Pain via CX3CL1. Neuromodulation 2023; 26:938-949. [PMID: 37045646 PMCID: PMC10330336 DOI: 10.1016/j.neurom.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/19/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Despite increasing utilization of spinal cord stimulation (SCS), its effects on chemoefficacy, cancer progression, and chemotherapy-induced peripheral neuropathy (CIPN) pain remain unclear. Up to 30% of adults who are cancer survivors may suffer from CIPN, and there are currently no effective preventative treatments. MATERIALS AND METHODS Through a combination of bioluminescent imaging, behavioral, biochemical, and immunohistochemical approaches, we investigated the role of SCS and paclitaxel (PTX) on tumor growth and PTX-induced peripheral neuropathy (PIPN) pain development in T-cell-deficient male rats (Crl:NIH-Foxn1rnu) with xenograft human non-small cell lung cancer. We hypothesized that SCS can prevent CIPN pain and enhance chemoefficacy partially by modulating macrophages, fractalkine (CX3CL1), and inflammatory cytokines. RESULTS We show that preemptive SCS enhanced the antitumor efficacy of PTX and prevented PIPN pain. Without SCS, rats with and without tumors developed robust PIPN pain-related mechanical hypersensitivity, but only those with tumors developed cold hypersensitivity, suggesting T-cell dependence for different PIPN pain modalities. SCS increased soluble CX3CL1 and macrophages and decreased neuronal and nonneuronal insoluble CX3CL1 expression and inflammation in dorsal root ganglia. CONCLUSION Collectively, our findings suggest that preemptive SCS is a promising strategy to increase chemoefficacy and prevent PIPN pain via CX3CL1-macrophage modulation.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Karla R Sanchez
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kimberly E Stephens
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Zhang W, Kong D, Li G, Yang Z, Cheng S, Li H, Feng L, Zhang K. Construction and validation of a chemokine family-based signature for the prediction of prognosis and therapeutic response in colon cancer. Heliyon 2023; 9:e16478. [PMID: 37484298 PMCID: PMC10360577 DOI: 10.1016/j.heliyon.2023.e16478] [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: 10/09/2022] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 07/25/2023] Open
Abstract
The role of chemokines in predicting the prognosis of colon cancer has not been mentioned. Chemokines have been shown to be associated with immune cell chemotaxis and activation, so the expression of chemokine genes in tumor tissue may be related to prognosis. We used a least absolute shrinkage and selection operator (LASSO) model based on chemokine gene families to construct a model that can predict the prognosis of colon cancer patients. We divided patients into high-risk groups and low-risk groups to study the prognosis. Then, we evaluated the relationship between the different risk groups in infiltration of immune cells. It was found that there was less immune cell infiltration in the high-risk group. We conducted a functional enrichment analysis based on model stratification, and explored the biological signal pathways enriched in the high and low-risk groups, which provided ideas for studying the mechanism behind its impact on prognosis. In addition, the chemokine-related gene signature could predict the response of patients to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Immunology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, College of Bioengineering, Hubei University of Technology, Wuhan 430068, China
| | - Defeng Kong
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Guoliang Li
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Zhenrong Yang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Hong Li
- Health Management Institute, The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, 28 Fuxing Road Beijing 100853, PR China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| |
Collapse
|
5
|
Bruss C, Kellner K, Ortmann O, Seitz S, Brockhoff G, Hutchinson JA, Wege AK. Advanced Immune Cell Profiling by Multiparameter Flow Cytometry in Humanized Patient-Derived Tumor Mice. Cancers (Basel) 2022; 14:2214. [PMID: 35565343 PMCID: PMC9103756 DOI: 10.3390/cancers14092214] [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: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/07/2022] Open
Abstract
"Humanized" mice have been widely used for the characterization of human cancer progression and as a powerful preclinical model. Standardization of multicolor phenotyping could help to identify immune cell patterns involved in checkpoint-related complications. Therefore, we applied established protocols for immune cell profiling to our humanized Patient-Derived Xenograft (hPDX) model. hPDX are characterized by the co-existence of a human immune system and a patient-derived tumor transplant. These mice possess a human-like immune system after CD34+ stem cell transplantation while the reconstitution level of the immune system was not related to the quantity of transplanted CD34+ cells. Contamination ≤ 1.2% by CD3+ cells in the hematopoietic stem cell (HSC) transplant did not trigger abnormal T cell maturation. Different B and T cell differentiation stages were identified, as well as regulatory T cells (Tregs) and exhausted T cells that expressed TIGIT, PD-1, or KLRG1. Overall, the application of standardized protocols for the characterization of immune cells using flow cytometry will contribute to a better understanding of immune-oncologic processes.
Collapse
Affiliation(s)
- Christina Bruss
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Kerstin Kellner
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Stephan Seitz
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| | - James A. Hutchinson
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Anja Kathrin Wege
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (C.B.); (K.K.); (O.O.); (S.S.); (G.B.)
| |
Collapse
|
6
|
Wege AK, Rom‐Jurek E, Jank P, Denkert C, Ugocsai P, Solbach C, Blohmer J, Sinn B, Mackelenbergh M, Möbus V, Trumpp A, Marangoni E, Pfarr N, Irlbeck C, Warfsmann J, Polzer B, Weber F, Ortmann O, Loibl S, Vladimirova V, Brockhoff G. mdm2
gene amplification is associated with luminal breast cancer progression in humanized
PDX
mice and a worse outcome of estrogen receptor positive disease. Int J Cancer 2021; 150:1357-1372. [DOI: 10.1002/ijc.33911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/25/2021] [Accepted: 12/07/2021] [Indexed: 01/02/2023]
Affiliation(s)
- Anja Kathrin Wege
- Department of Gynecology and Obstetrics University Medical Center Regensburg Regensburg Germany
| | - Eva‐Maria Rom‐Jurek
- Department of Gynecology and Obstetrics University Medical Center Regensburg Regensburg Germany
| | - Paul Jank
- Institute of Pathology, Philipps‐University Marburg UKGM University Hospital Marburg Marburg Germany
| | - Carsten Denkert
- Institute of Pathology, Philipps‐University Marburg UKGM University Hospital Marburg Marburg Germany
| | - Peter Ugocsai
- Department of Gynecology and Obstetrics University Medical Center Regensburg Regensburg Germany
| | - Christine Solbach
- Department of Gynecology and Obstetrics University Hospital Frankfurt Frankfurt Germany
| | - Jens‐Uwe Blohmer
- Breast Cancer Center Charité Universitätsmedizin Berlin Berlin Germany
| | - Bruno Sinn
- Breast Cancer Center Charité Universitätsmedizin Berlin Berlin Germany
| | - Marion Mackelenbergh
- Department of Gynecology and Obstetrics Schleswig‐Holstein University Hospital Kiel Germany
| | - Volker Möbus
- Department of Medicine II, Hematology and Oncology Goethe University of Frankfurt Frankfurt Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ‐ZMBH Alliance Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH), German Cancer Consortium (DKTK) Heidelberg Germany
| | - Elisabetta Marangoni
- Department of Translational Research, Institute Curie PSL Research University Paris France
| | - Nicole Pfarr
- Institute of Pathology Technical University Munich Munich Germany
| | - Christoph Irlbeck
- Division of Personalized Tumor Therapy Fraunhofer Institute for Toxicology and Experimental Medicine Regensburg Germany
- Department of Experimental Medicine University of Regensburg Regensburg Germany
| | - Jens Warfsmann
- Division of Personalized Tumor Therapy Fraunhofer Institute for Toxicology and Experimental Medicine Regensburg Germany
- Department of Experimental Medicine University of Regensburg Regensburg Germany
| | - Bernhard Polzer
- Division of Personalized Tumor Therapy Fraunhofer Institute for Toxicology and Experimental Medicine Regensburg Germany
- Department of Experimental Medicine University of Regensburg Regensburg Germany
| | | | - Olaf Ortmann
- Department of Gynecology and Obstetrics University Medical Center Regensburg Regensburg Germany
| | - Sibylle Loibl
- German Breast Group GBG Forschungs GmbH Neu‐Isenburg Germany
| | | | - Gero Brockhoff
- Department of Gynecology and Obstetrics University Medical Center Regensburg Regensburg Germany
| |
Collapse
|