1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Matrix Stiffness Triggers Lipid Metabolic Cross-talk between Tumor and Stromal Cells to Mediate Bevacizumab Resistance in Colorectal Cancer Liver Metastases

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Extracellular matrix stiffening drives bevacizumab resistance by stimulating hepatic stellate cells to provide fuel for mCRC cells in the liver, indicating a potential metabolism-based therapeutic strategy for overcoming resistance.

          Abstract

          Bevacizumab is an anti-VEGF monoclonal antibody that plays an important role in the combination treatment of advanced colorectal cancer. However, resistance remains a major hurdle limiting bevacizumab efficacy, highlighting the importance of identifying a mechanism of antiangiogenic therapy resistance. Here, we investigated biophysical properties of the extracellular matrix (ECM) related to metabolic processes and acquired resistance to bevacizumab. Evaluation of paired pre- and posttreatment samples of liver metastases from 20 colorectal cancer patients treated with combination bevacizumab therapy, including 10 responders and 10 nonresponders, indicated that ECM deposition in liver metastases and a highly activated fatty acid oxidation (FAO) pathway were elevated in nonresponders after antiangiogenic therapy compared with responders. In mouse models of liver metastatic colorectal cancer (mCRC), anti-VEGF increased ECM deposition and FAO in colorectal cancer cells, and treatment with the FAO inhibitor etomoxir enhanced the efficacy of antiangiogenic therapy. Hepatic stellate cells (HSC) were essential for matrix stiffness–mediated FAO in colon cancer cells. Matrix stiffness activated lipolysis in HSCs via the focal adhesion kinase (FAK)/yes-associated protein (YAP) pathway, and free fatty acids secreted by HSCs were absorbed as metabolic substrates and activated FAO in colon cancer cells. Suppressing HSC lipolysis using FAK and YAP inhibition enhanced the efficacy of anti-VEGF therapy. Together, these results indicate that bevacizumab-induced ECM remodeling triggers lipid metabolic cross-talk between colon cancer cells and HSCs. This metabolic mechanism of bevacizumab resistance mediated by the physical tumor microenvironment represents a potential therapeutic target for reversing drug resistance.

          Significance:

          Extracellular matrix stiffening drives bevacizumab resistance by stimulating hepatic stellate cells to provide fuel for mCRC cells in the liver, indicating a potential metabolism-based therapeutic strategy for overcoming resistance.

          Graphical Abstract

          Related collections

          Most cited references41

          • Record: found
          • Abstract: found
          • Article: not found

          The biology and function of fibroblasts in cancer.

          Among all cells, fibroblasts could be considered the cockroaches of the human body. They survive severe stress that is usually lethal to all other cells, and they are the only normal cell type that can be live-cultured from post-mortem and decaying tissue. Their resilient adaptation may reside in their intrinsic survival programmes and cellular plasticity. Cancer is associated with fibroblasts at all stages of disease progression, including metastasis, and they are a considerable component of the general host response to tissue damage caused by cancer cells. Cancer-associated fibroblasts (CAFs) become synthetic machines that produce many different tumour components. CAFs have a role in creating extracellular matrix (ECM) structure and metabolic and immune reprogramming of the tumour microenvironment with an impact on adaptive resistance to chemotherapy. The pleiotropic actions of CAFs on tumour cells are probably reflective of them being a heterogeneous and plastic population with context-dependent influence on cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Molecular mechanisms and clinical applications of angiogenesis.

            Blood vessels deliver oxygen and nutrients to every part of the body, but also nourish diseases such as cancer. Over the past decade, our understanding of the molecular mechanisms of angiogenesis (blood vessel growth) has increased at an explosive rate and has led to the approval of anti-angiogenic drugs for cancer and eye diseases. So far, hundreds of thousands of patients have benefited from blockers of the angiogenic protein vascular endothelial growth factor, but limited efficacy and resistance remain outstanding problems. Recent preclinical and clinical studies have shown new molecular targets and principles, which may provide avenues for improving the therapeutic benefit from anti-angiogenic strategies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mechanisms of hepatic stellate cell activation

              Activation of hepatic stellate cells (HSCs) in liver injury is the primary driver of hepatic fibrosis. In this Review, Tsuchida and Friedman detail the varied intracellular and extracellular signalling pathways leading to HSC activation, as well as the role of HSCs in liver fibrosis resolution and as therapeutic targets.
                Bookmark

                Author and article information

                Journal
                Cancer Res
                Cancer Res
                Cancer Research
                American Association for Cancer Research
                0008-5472
                1538-7445
                01 November 2023
                23 August 2023
                : 83
                : 21
                : 3577-3592
                Affiliations
                [1 ]Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
                [2 ]Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
                Author notes
                [#]

                Y. Zheng, R. Zhou, and J. Cai contributed equally to this article.

                [* ] Corresponding Author: Wangjun Liao, Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, China. E-mail: nfyyliaowj@ 123456163.com

                Cancer Res 2023;83:3577–92

                Author information
                https://orcid.org/0009-0007-6222-6303
                https://orcid.org/0000-0002-8250-9446
                https://orcid.org/0009-0008-7618-2951
                https://orcid.org/0000-0002-3565-988X
                https://orcid.org/0000-0002-5995-1295
                https://orcid.org/0009-0001-3542-2209
                https://orcid.org/0000-0002-1584-4949
                https://orcid.org/0009-0001-7777-910X
                https://orcid.org/0009-0000-5121-9246
                https://orcid.org/0000-0001-7961-0677
                https://orcid.org/0000-0003-0113-4089
                https://orcid.org/0000-0001-5961-390X
                https://orcid.org/0000-0003-4799-1502
                https://orcid.org/0000-0002-1364-8442
                Article
                CAN-23-0025
                10.1158/0008-5472.CAN-23-0025
                10618741
                37610655
                7eeee072-aaf5-4666-b724-1f435259932f
                ©2023 The Authors; Published by the American Association for Cancer Research

                This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.

                History
                : 03 January 2023
                : 06 July 2023
                : 21 August 2023
                Page count
                Pages: 16
                Funding
                Funded by: National Natural Science Foundation of China (NSFC), https://doi.org/10.13039/501100001809;
                Award ID: 82073303
                Award Recipient :
                Funded by: National Natural Science Foundation of China (NSFC), https://doi.org/10.13039/501100001809;
                Award ID: 82272635
                Award Recipient :
                Funded by: National Natural Science Foundation of China (NSFC), https://doi.org/10.13039/501100001809;
                Award ID: 82102731
                Award Recipient :
                Funded by: Nanfang Hospital, https://doi.org/10.13039/501100010112;
                Award ID: 2019Z025
                Award Recipient :
                Funded by: Natural Science Foundation of Guangdong Province (廣東省自然科學基金), https://doi.org/10.13039/501100003453;
                Award ID: 2022A1515012418
                Award Recipient :
                Categories
                Cancer Metabolism and Molecular Mechanisms

                Comments

                Comment on this article