1
|
VandenHeuvel SN, Nash LL, Raghavan SA. Dormancy in Metastatic Colorectal Cancer: Tissue Engineering Opportunities for In Vitro Modeling. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40195931 DOI: 10.1089/ten.teb.2025.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Colorectal cancer (CRC) recurs at a striking rate, specifically in patients with liver metastasis. Dormant CRC cells disseminated following initial primary tumor resection or treatment often resurface years later to form aggressive, therapy-resistant tumors that result in high patient mortality. Routine imaging-based screenings often fail to detect dormant cancer cell clusters, and there are no overt symptomatic presentations, making dormant CRC a major clinical challenge to diagnose and treat. Tissue engineering approaches are ideally suited to model dormant cancer cells and enable the discovery of therapeutic vulnerabilities or unique mechanistic dependencies of dormant CRC. Emerging evidence suggests that tissue-engineered approaches have been successfully used to model dormant breast and lung cancer. With CRC responsible for the second most cancer-related deaths worldwide and CRC patients commonly experiencing recurrence, it is essential to expand dormancy models to understand this phenomenon in the context of CRC. Most published in vitro models of CRC dormancy simplify the complex tumor microenvironment with two-dimensional culture systems to elucidate dormancy-driving mechanisms. Building on this foundation, future research should apply tissue engineering methods to this growing field to generate competent three-dimensional models and increase mechanistic knowledge. This review summarizes the current state of in vitro CRC dormancy models, highlighting the techniques utilized to give rise to dormant CRC cells: nutrient depletion, anticancer drugs, physical extracellular matrix interactions, and genetic manipulation. The metrics used to validate dormancy within each model are also consolidated to demonstrate the lack of established standards and the ambiguity around comparing studies that have been validated differently. The methods of these studies are organized in this review to increase comprehensibility and identify needs and opportunities for future bioengineered in vitro models to address dormancy-driven mortality in patients with CRC liver metastasis. Impact Statement Dormant cancer drives high patient mortality, especially in metastatic colorectal cancer, owing to the clinical inability to identify dormant cells prior to their overt recurrence. Lacking clinical insights, in vitro modeling for mechanistic and therapeutic discovery is hindered. Here, we review models and methods of inducing colorectal cancer dormancy with the goal of consolidating findings for reference. We also highlight the need for advanced, tissue-engineered models to better mimic the organ-specific 3D microenvironment of metastatic colorectal cancer. New models would enable breakthroughs in understanding mechanisms driving dormancy progression and reversal, thereby providing context for therapeutic advances to improve patient survival.
Collapse
Affiliation(s)
| | - Lucia L Nash
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Regional Excellence Center in Cancer, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
2
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [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: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia 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, 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, 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, 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, China.
| |
Collapse
|
3
|
Huang HY, Chen YZ, Zhao C, Zheng XN, Yu K, Yue JX, Ju HQ, Shi YX, Tian L. Alternations in inflammatory macrophage niche drive phenotypic and functional plasticity of Kupffer cells. Nat Commun 2024; 15:9337. [PMID: 39472435 PMCID: PMC11522483 DOI: 10.1038/s41467-024-53659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Inflammatory signals lead to recruitment of circulating monocytes and induce their differentiation into pro-inflammatory macrophages. Therefore, whether blocking inflammatory monocytes can mitigate disease progression is being actively evaluated. Here, we employ multiple lineage-tracing models and show that monocyte-derived macrophages (mo-mac) are the major population of immunosuppressive, liver metastasis-associated macrophages (LMAM), while the proportion of Kupffer cells (KC) as liver-resident macrophages is diminished in metastatic nodules. Paradoxically, genetic ablation of mo-macs results in only a marginal decrease in LMAMs. Using a proliferation-recording system and a KC-tracing model in a monocyte-deficient background, we find that LMAMs can be replenished either via increased local macrophage proliferation or by promoting KC infiltration. In the latter regard, KCs undergo transient proliferation and exhibit substantial phenotypic and functional alterations through epigenetic reprogramming following the vacating of macrophage niches by monocyte depletion. Our data thus suggest that a simultaneous blockade of monocyte recruitment and macrophage proliferation may effectively target immunosuppressive myelopoiesis and reprogram the microenvironment towards an immunostimulatory state.
Collapse
Affiliation(s)
- Han-Ying Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan-Zhou Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chuang Zhao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xin-Nan Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kai Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jia-Xing Yue
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan-Xia Shi
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Lin Tian
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
4
|
Deng Z, Loyher PL, Lazarov T, Li L, Shen Z, Bhinder B, Yang H, Zhong Y, Alberdi A, Massague J, Sun JC, Benezra R, Glass CK, Elemento O, Iacobuzio-Donahue CA, Geissmann F. The nuclear factor ID3 endows macrophages with a potent anti-tumour activity. Nature 2024; 626:864-873. [PMID: 38326607 PMCID: PMC10881399 DOI: 10.1038/s41586-023-06950-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 12/07/2023] [Indexed: 02/09/2024]
Abstract
Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.
Collapse
Affiliation(s)
- Zihou Deng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Li Li
- Graduate Center, City University of New York, New York, NY, USA
| | - Zeyang Shen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY, USA
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yi Zhong
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joan Massague
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph C Sun
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher K Glass
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY, USA
| | | | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
5
|
Epidermal Growth Factor Receptor as Target for Perioperative Elimination of Circulating Colorectal Cancer Cells. JOURNAL OF ONCOLOGY 2022; 2022:3577928. [PMID: 35035479 PMCID: PMC8759909 DOI: 10.1155/2022/3577928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022]
Abstract
Surgical resection of the tumor is the primary treatment of colorectal cancer patients. However, we previously demonstrated that abdominal surgery promotes the adherence of circulating tumor cells (CTC) in the liver and subsequent liver metastasis development. Importantly, preoperative treatment with specific tumor-targeting monoclonal antibodies (mAb) prevented surgery-induced liver metastasis development in rats. This study investigated whether the epidermal growth factor receptor (EGFR) represents a suitable target for preoperative antibody treatment of colorectal cancer patients undergoing surgery. The majority of patients with resectable colorectal liver metastases were shown to have EGFR + CTCs. Three different anti-EGFR mAbs (cetuximab, zalutumumab, and panitumumab) were equally efficient in the opsonization of tumor cell lines. Additionally, all three mAbs induced antibody-dependent cellular phagocytosis (ADCP) of tumor cells by macrophages at low antibody concentrations in vitro, independent of mutations in EGFR signaling pathways. The plasma of cetuximab-treated patients efficiently opsonized tumor cells ex vivo and induced phagocytosis. Furthermore, neither proliferation nor migration of epithelial cells was affected in vitro, supporting that wound healing will not be hampered by treatment with low anti-EGFR mAb concentrations. These data support the use of a low dose of anti-EGFR mAbs prior to resection of the tumor to eliminate CTCs without interfering with the healing of the anastomosis. Ultimately, this may reduce the risk of metastasis development, consequently improving long-term patient outcome significantly.
Collapse
|
6
|
Miarka L, Hauser C, Helm O, Holdhof D, Beckinger S, Egberts JH, Gundlach JP, Lenk L, Rahn S, Mikulits W, Trauzold A, Sebens S. The Hepatic Microenvironment and TRAIL-R2 Impact Outgrowth of Liver Metastases in Pancreatic Cancer after Surgical Resection. Cancers (Basel) 2019; 11:cancers11060745. [PMID: 31146405 PMCID: PMC6627672 DOI: 10.3390/cancers11060745] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/26/2019] [Indexed: 12/12/2022] Open
Abstract
Most patients with pancreatic ductal adenocarcinoma (PDAC) undergoing curative resection relapse within months, often with liver metastases. The hepatic microenvironment determines induction and reversal of dormancy during metastasis. Both tumor growth and metastasis depend on the Tumor necrosis factor (TNF)-related apoptosis-inducing ligand-receptor 2 (TRAIL-R2). This study investigated the interplay of TRAIL-R2 and the hepatic microenvironment in liver metastases formation and the impact of surgical resection. Although TRAIL-R2-knockdown (PancTu-I shTR2) decreased local relapses and number of macroscopic liver metastases after primary tumor resection in an orthotopic PDAC model, the number of micrometastases was increased. Moreover, abdominal surgery induced liver inflammation involving activation of hepatic stellate cells (HSCs) into hepatic myofibroblasts (HMFs). In coculture with HSCs, proliferation of PancTu-I shTR2 cells was significantly lower compared to PancTu-I shCtrl cells, an effect still observed after switching coculture from HSC to HMF, mimicking surgery-mediated liver inflammation and enhancing cell proliferation. CXCL-8/IL-8 blockade diminished HSC-mediated growth inhibition in PancTu-I shTR2 cells, while Vascular Endothelial Growth Factor (VEGF) neutralization decreased HMF-mediated proliferation. Overall, this study points to an important role of TRAIL-R2 in PDAC cells in the interplay with the hepatic microenvironment during metastasis. Resection of primary PDAC seems to induce liver inflammation, which might contribute to outgrowth of liver metastases.
Collapse
Affiliation(s)
- Lauritz Miarka
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
| | - Charlotte Hauser
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany.
| | - Ole Helm
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
| | - Dörthe Holdhof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
- Department, Research Institute Children's Cancer Center Hamburg, 20251 Hamburg, Germany.
| | - Silje Beckinger
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
| | - Jan-Hendrik Egberts
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany.
| | - Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany.
| | - Lennart Lenk
- Department of Pediatrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein, Schwanenweg 20, 24105 Kiel, Germany.
| | - Sascha Rahn
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Anna Trauzold
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, 24105 Kiel, Germany.
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel, Arnold-Heller-Str. 3, Building 17, 24105 Kiel, Germany.
| |
Collapse
|
7
|
Keirsse J, Van Damme H, Geeraerts X, Beschin A, Raes G, Van Ginderachter JA. The role of hepatic macrophages in liver metastasis. Cell Immunol 2018; 330:202-215. [PMID: 29661474 DOI: 10.1016/j.cellimm.2018.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 12/21/2022]
Abstract
The liver is a major target organ for metastasis of both gastrointestinal and extra-gastrointestinal cancers. Due to its frequently inoperable nature, liver metastasis represents a leading cause of cancer-associated death worldwide. In the past years, the pivotal role of the immune system in this process is being increasingly recognised. In particular, the role of the hepatic macrophages, both recruited monocyte-derived macrophages (Mo-Mfs) and tissue-resident Kupffer cells (KCs), has been shown to be more versatile than initially imagined. However, the lack of tools to easily distinguish between these two macrophage populations has hampered the assignment of particular functionalities to specific hepatic macrophage subsets. In this Review, we highlight the most remarkable findings regarding the origin and functions of hepatic macrophage populations, and we provide a detailed description of their distinct roles in the different phases of the liver metastatic process.
Collapse
Affiliation(s)
- Jiri Keirsse
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Helena Van Damme
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Xenia Geeraerts
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Alain Beschin
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Raes
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium; Lab of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
Milette S, Sicklick JK, Lowy AM, Brodt P. Molecular Pathways: Targeting the Microenvironment of Liver Metastases. Clin Cancer Res 2017; 23:6390-6399. [PMID: 28615370 PMCID: PMC5668192 DOI: 10.1158/1078-0432.ccr-15-1636] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/27/2017] [Accepted: 06/09/2017] [Indexed: 12/31/2022]
Abstract
Curative treatment for metastatic solid cancers remains elusive. The liver, which is nourished by a rich blood supply from both the arterial and portal venous systems, is the most common site of visceral metastases, particularly from cancers arising in the gastrointestinal tract, with colorectal cancer being the predominant primary site in Western countries. A mounting body of evidence suggests that the liver microenvironment (LME) provides autocrine and paracrine signals originating from both parenchymal and nonparenchymal cells that collectively create both pre- and prometastatic niches for the development of hepatic metastases. These resident cells and their molecular mediators represent potential therapeutic targets for the prevention and/or treatment of liver metastases (LM). This review summarizes: (i) the current therapeutic options for treating LM, with a particular focus on colorectal cancer LM; (ii) the role of the LME in LM at each of its phases; (iii) potential targets in the LME identified through preclinical and clinical investigations; and (iv) potential therapeutic approaches for targeting elements of the LME before and/or after the onset of LM as the basis for future clinical trials. Clin Cancer Res; 23(21); 6390-9. ©2017 AACR.
Collapse
Affiliation(s)
- Simon Milette
- Departments of Surgery, Medicine, and Oncology McGill University and the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jason K. Sicklick
- Division of Surgical Oncology, Department of Surgery, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Andrew M. Lowy
- Division of Surgical Oncology, Department of Surgery, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Pnina Brodt
- Departments of Surgery, Medicine, and Oncology McGill University and the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Molla NW, Hassanain MM, Fadel Z, Boucher LM, Madkhali A, Altahan RM, Alrijraji EA, Simoneau EB, Alamri H, Salman A, Gao Z, Metrakos PP. Effect of non-alcoholic liver disease on recurrence rate and liver regeneration after liver resection for colorectal liver metastases. ACTA ACUST UNITED AC 2017; 24:e233-e243. [PMID: 28680292 DOI: 10.3747/co.24.3133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Resection of metastases is the only potential cure for patients with liver metastasis from colorectal cancer (crc-lm). But despite an improved overall 5-year survival, the recurrence rate is still as high as 60%. Non-alcoholic fatty liver disease (nafld) can decrease the liver's capacity to regenerate after resection and might also affect cancer recurrence, potentially by elevating transforming growth factor β, levels of specific metalloproteinases, and oxidative stress. The objective of the present work was to determine the effect of the histologic features of nafld on cancer recurrence and liver regeneration. METHODS This retrospective analysis considered 60 patients who underwent an R0 hepatectomy for crc-lm. Volumetric analysis of the liver was calculated using axial view, portovenous phase, 2.5 mm thickness, multiphasic computed tomography images taken before and after surgery. The histologic features of nafld (steatosis, inflammation, and ballooning) were scored using the nafld activity score, and the degree of fibrosis was determined. RESULTS The hepatic recurrence rate was 38.33%. Median overall survival duration was 56 months. Median disease-free survival duration was 14 months, and median hepatic disease-free survival duration was 56 months. Multivariate analysis revealed significant correlations of hepatic disease-free survival with hepatocyte ballooning (p = 0.0009), lesion diameter (p = 0.014), and synchronous disease (p = 0.006). Univariate and multivariate analyses did not reveal any correlation with degree of steatosis or recurrence rate. CONCLUSIONS This study reveals an important potential negative effect of hepatocyte ballooning on hepatic disease-free survival.
Collapse
Affiliation(s)
- N W Molla
- Department of Surgery, Section of Hepatopancreatobiliary, and.,Department of Radiology, McGill University Health Centre, Montreal, QC.,Department of Radiology and
| | - M M Hassanain
- Department of Surgery, Section of Hepatopancreatobiliary, and.,Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - Z Fadel
- Department of Surgery, Section of Hepatopancreatobiliary, and
| | - L M Boucher
- Department of Radiology, McGill University Health Centre, Montreal, QC
| | - A Madkhali
- Department of Surgery, Section of Hepatopancreatobiliary, and.,Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - R M Altahan
- Department of Surgery, Section of Hepatopancreatobiliary, and
| | - E A Alrijraji
- Department of Surgery, Section of Hepatopancreatobiliary, and
| | - E B Simoneau
- Department of Surgery, Section of Hepatopancreatobiliary, and
| | - H Alamri
- Department of Surgery, Section of Hepatopancreatobiliary, and.,Department of Surgery, King Saud University, Riyadh, Saudi Arabia
| | - A Salman
- Department of Surgery, Section of Hepatopancreatobiliary, and
| | - Z Gao
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - P P Metrakos
- Department of Surgery, Section of Hepatopancreatobiliary, and.,Department of Pathology, McGill University Health Centre, Montreal, QC
| |
Collapse
|
10
|
Brodt P. Role of the Microenvironment in Liver Metastasis: From Pre- to Prometastatic Niches. Clin Cancer Res 2016; 22:5971-5982. [PMID: 27797969 DOI: 10.1158/1078-0432.ccr-16-0460] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 01/14/2023]
Abstract
Liver metastases remain a major barrier to successful management of malignant disease, particularly for cancers of the gastrointestinal tract but also for other malignancies, such as breast carcinoma and melanoma. The ability of metastatic cells to survive and proliferate in the liver is determined by the outcome of complex, reciprocal interactions between tumor cells and different local resident subpopulations, including the sinusoidal endothelium, stellate, Kupffer, and inflammatory cells that are mediated through cell-cell and cell-extracellular matrix adhesion and the release of soluble factors. Cross-communication between different hepatic resident cells in response to local tissue damage and inflammation and the recruitment of bone marrow cells further enhance this intercellular communication network. Both resident and recruited cells can play opposing roles in the progression of metastasis, and the balance of these divergent effects determines whether the tumor cells will die, proliferate, and colonize the new site or enter a state of dormancy. Moreover, this delicate balance can be tilted in favor of metastasis, if factors produced by the primary tumor precondition the microenvironment to form niches of activated resident cells that promote tumor expansion. This review aims to summarize current knowledge on these diverse interactions and the impact they can have on the clinical management of hepatic metastases. Clin Cancer Res; 22(24); 5971-82. ©2016 AACR.
Collapse
Affiliation(s)
- Pnina Brodt
- Departments of Surgery, Medicine, and Oncology, McGill University and the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
11
|
Braster R, Bögels M, Beelen RHJ, van Egmond M. The delicate balance of macrophages in colorectal cancer; their role in tumour development and therapeutic potential. Immunobiology 2015; 222:21-30. [PMID: 26358365 DOI: 10.1016/j.imbio.2015.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/28/2015] [Accepted: 08/29/2015] [Indexed: 02/07/2023]
Abstract
Most tumours are heavily infiltrated by immune cells. This has been correlated with either a good or a bad patient prognosis, depending on the (sub) type of immune cells. Macrophages represent one of the most prominent leukocyte populations in the majority of tumours. Functions of macrophages range from cytotoxicity, to stimulation of tumour growth by secretion of cytokines, growth and angiogenic factors, or suppressing immune responses. In most tumours macrophages are described as cells with immune suppressing, and wound healing properties, which aids tumour development. Yet, increasing evidence shows that macrophages are potent inhibitors of tumour growth in colorectal cancer. Macrophages in this respect show high plasticity. The presence of high macrophage numbers in the tumour may therefore become advantageous, if cells can be reprogrammed from tumour promoting macrophages into potent effector cells. Enhancing cytotoxic properties of macrophages by microbial products, pro-inflammatory cytokines or monoclonal antibody therapy are promising possibilities, and are currently tested in clinical trials.
Collapse
Affiliation(s)
- R Braster
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - M Bögels
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - R H J Beelen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - M van Egmond
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; Department of Surgery, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Paschos KA, Majeed AW, Bird NC. Natural history of hepatic metastases from colorectal cancer - pathobiological pathways with clinical significance. World J Gastroenterol 2014; 20:3719-3737. [PMID: 24744570 PMCID: PMC3983432 DOI: 10.3748/wjg.v20.i14.3719] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 11/12/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer hepatic metastases represent the final stage of a multi-step biological process. This process starts with a series of mutations in colonic epithelial cells, continues with their detachment from the large intestine, dissemination through the blood and/or lymphatic circulation, attachment to the hepatic sinusoids and interactions with the sinusoidal cells, such as sinusoidal endothelial cells, Kupffer cells, stellate cells and pit cells. The metastatic sequence terminates with colorectal cancer cell invasion, adaptation and colonisation of the hepatic parenchyma. All these events, termed the colorectal cancer invasion-metastasis cascade, include multiple molecular pathways, intercellular interactions and expression of a plethora of chemokines and growth factors, and adhesion molecules, such as the selectins, the integrins or the cadherins, as well as enzymes including matrix metalloproteinases. This review aims to present recent advances that provide insights into these cell-biological events and emphasizes those that may be amenable to therapeutic targeting.
Collapse
|
13
|
Yang Q, Liu Y, Shi Y, Zheng M, He J, Chen Z. The role of intracellular high-mobility group box 1 in the early activation of Kupffer cells and the development of Con A-induced acute liver failure. Immunobiology 2013; 218:1284-92. [DOI: 10.1016/j.imbio.2013.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/21/2013] [Indexed: 01/26/2023]
|
14
|
Wen SW, Ager EI, Neo J, Christophi C. The renin angiotensin system regulates Kupffer cells in colorectal liver metastases. Cancer Biol Ther 2013; 14:720-7. [PMID: 23792575 DOI: 10.4161/cbt.25092] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Blockade of the renin angiotensin system (RAS) can inhibit tumor growth and this may be mediated via undefined immunomodulatory actions. This study investigated the effects of RAS blockade on liver macrophages (Kupffer cells; KCs) in an orthotopic murine model of colorectal cancer (CRC) liver metastases. Here we showed that pharmacological targeting of the RAS [ANG II (31.25 µg/kg/h i.p.), ANG-(1-7) (24 µg/kg/h i.p.) or the ACE inhibitor; captopril (750 mg/kg/d i.p.)] altered endogenous KC numbers in the tumor-bearing liver throughout metastatic growth. Captopril, and to a lesser extent ANG-(1-7), increased KC numbers in the liver but not tumor. KCs were found to express the key RAS components: ACE and AT1R. Treatment with captopril and ANG II increased the number of AT1R-expressing KCs, although total KC numbers were not affected by ANG II. Captopril (0.1 µM) also increased macrophage invasion in vitro. Additionally, captopril was administered with KC depletion before tumor induction (day 0) or at established metastatic growth (day 18) using gadolinium chloride (GdCl 3; 20 mg/kg). Livers were collected at day 21 and quantitative stereology used as a measure of tumor burden. Captopril reduced growth of CRC liver metastases. However, when captopril was combined with early KC depletion (day 0) tumor growth was significantly increased compared with captopril alone. In contrast, late KC depletion (day 18) failed to influence the anti-tumor effects of captopril. The result of these studies suggests that manipulation of the RAS can alter KC numbers and may subsequently influence progression of CRC liver metastases.
Collapse
Affiliation(s)
- Shu Wen Wen
- Department of Surgery, The University of Melbourne, Austin Health, Heidelberg, Victoria, Australia.
| | | | | | | |
Collapse
|
15
|
Wen SW, Ager EI, Christophi C. Bimodal role of Kupffer cells during colorectal cancer liver metastasis. Cancer Biol Ther 2013; 14:606-13. [PMID: 23792646 DOI: 10.4161/cbt.24593] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Kupffer cells (KCs) are resident liver macrophages that play a crucial role in liver homeostasis and in the pathogenesis of liver disease. Evidence suggests KCs have both stimulatory and inhibitory functions during tumor development but the extent of these functions remains to be defined. Using KC depletion studies in an orthotopic murine model of colorectal cancer (CRC) liver metastases we demonstrated the bimodal role of KCs in determining tumor growth. KC depletion with gadolinium chloride before tumor induction was associated with an increased tumor burden during the exponential growth phase. In contrast, KC depletion at the late stage of tumor growth (day 18) decreased liver tumor load compared with non-depleted animals. This suggests KCs exhibit an early inhibitory and a later stimulatory effect. These two opposing functions were associated with changes in iNOS and VEGF expression as well as T-cell infiltration. KC depletion at day 18 increased numbers of CD3 (+) T cells and iNOS-expressing infiltrating cells in the tumor, but decreased the number of VEGF-expressing infiltrating cells. These alterations may be responsible for the observed reduction in tumor burden following depletion of pro-tumor KCs at the late stage of metastatic growth. Taken together, our results indicate that the bimodal role of KC activity in liver tumors may provide the key to timing immunomodulatory intervention for the treatment of CRC liver metastases.
Collapse
Affiliation(s)
- Shu Wen Wen
- Department of Surgery, The University of Melbourne, Austin Health, Heidelberg, Australia.
| | | | | |
Collapse
|
16
|
Chen P, Bonaldo P. Role of macrophage polarization in tumor angiogenesis and vessel normalization: implications for new anticancer therapies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 301:1-35. [PMID: 23317816 DOI: 10.1016/b978-0-12-407704-1.00001-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiogenesis, the formation of new capillary blood vessels from preexisting vasculature, is one of the hallmarks of cancer that is pivotal for tumor growth and metastasis. Tumor vessels are known to be abnormal, with typically aberrant, leaky and disordered vessels. Thus, the combination of angiogenesis inhibition and vessel normalization is a potential strategy for anticancer therapy. The solid tumor is composed of not only cancer cells, but also the nonmalignant resident stromal cells, such as bone-marrow-derived cells (BMDCs) and cancer-associated fibroblasts (CAFs). Tumor-associated macrophages (TAMs) are the most abundant cell components of BMDCs, which play a significant role in promoting tumor progression. Accumulating evidences from both patient biopsies and experimental animal models have shown that TAMs function in tumor angiogenesis and vessel abnormalization in a density- and phenotype-dependent manner. This chapter will discuss the evidence for the factors and signaling pathways that are involved in macrophage recruitment and polarization in the tumor microenvironment, and it summarizes the role and underlying molecular mechanisms of macrophage polarization in tumor angiogenesis and vessel normalization. In addition, an overview of the potential of targeting TAM polarization for anticancer therapy will be provided.
Collapse
Affiliation(s)
- Peiwen Chen
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | | |
Collapse
|
17
|
|
18
|
Koh SL, Ager EI, Christophi C. Liver regeneration and tumour stimulation: implications of the renin-angiotensin system. Liver Int 2010; 30:1414-26. [PMID: 20633100 DOI: 10.1111/j.1478-3231.2010.02306.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver resection is the most effective treatment for primary liver tumours and metastasis to the liver, and remains the only potentially long-term curative therapy for patients with colorectal cancer (CRC) liver metastases. Nevertheless, there is a significant incidence of tumour recurrence following liver resection. Cellular and molecular changes resulting from resection and the subsequent liver regeneration process may influence the kinetics of tumour growth, contributing to recurrence. Although commonly associated with the systemic homeostasis of blood pressure, fluid and electrolyte, the renin-angiotensin system (RAS) has recently been shown to play a role in regulating cell proliferation, apoptosis and angiogenesis in local organs as well as in malignancies. An electronic search of the English literature on the role of the RAS in liver regeneration and tumourigenesis was performed using PubMed, with additional relevant articles sourced from reference lists. Studies have shown that the blockade of the RAS pathway stimulates liver regeneration and inhibits tumour progression. An understanding of the role of RAS in liver regeneration and tumourigenesis may enable alternative strategies to improve patient outcome and survival after liver resection. This review will discuss the role of the RAS in liver regeneration and in tumour recurrence post-liver resection. The potential of the RAS as a novel therapeutic target for CRC liver metastases patients undergoing liver resection will be outlined.
Collapse
Affiliation(s)
- Shir Lin Koh
- Austin Health, Department of Surgery, The University of Melbourne, Heidelberg, Vic., Australia.
| | | | | |
Collapse
|
19
|
Li Y, Cao H, Jiao Z, Pakala SB, Sirigiri DNR, Li W, Kumar R, Mishra L. Carcinoembryonic antigen interacts with TGF-{beta} receptor and inhibits TGF-{beta} signaling in colorectal cancers. Cancer Res 2010; 70:8159-68. [PMID: 20889724 PMCID: PMC3001246 DOI: 10.1158/0008-5472.can-10-1073] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
As a tumor marker for colorectal cancers, carcinoembryonic antigen (CEA) enhances the metastatic potential of cancer cells. CEA functions as an intercellular adhesion molecule and is upregulated in a wide variety of human cancers. However, the molecular mechanisms by which CEA mediates metastasis remain to be understood. Transforming growth factor-β (TGF-β) signaling regulates both tumor suppression and metastasis, and also contributes to the stimulation of CEA transcription and secretion in colorectal cancer cells. However, it remains unknown whether CEA, in turn, influences TGF-β functions and if a regulatory cross-talk exists between CEA and the TGF-β signaling pathway. Here, we report that CEA directly interacts with TGF-β receptor and inhibits TGF-β signaling. Targeting CEA with either CEA-specific antibody or siRNA rescues TGF-β response in colorectal cancer cell lines with elevated CEA, thereby restoring the inhibitory effects of TGF-β signaling on proliferation. CEA also enhances the survival of colorectal cancer cells in both local colonization and liver metastasis in animal study. Our study provides novel insights into the interaction between CEA and TGF-β signaling pathway and establishes a negative feedback loop in amplifying the progression of colon cancer cells to more invasive phenotypes. These findings offer new therapeutic opportunities to inhibit colorectal cancer cell proliferation by cotargeting CEA in promoting tumor-inhibitory action of the TGF-β pathway.
Collapse
Affiliation(s)
- Ying Li
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, the University of Texas
| | - Hong Cao
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, the University of Texas
| | - Zhongxian Jiao
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, the University of Texas
| | - Suresh B. Pakala
- Department of Biochemistry and Molecular Biology, the George Washington University Medical Center, Washington, DC 22237
| | | | - Wenpin Li
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, the University of Texas
| | - Rakesh Kumar
- Department of Biochemistry and Molecular Biology, the George Washington University Medical Center, Washington, DC 22237
| | - Lopa Mishra
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, the University of Texas
| |
Collapse
|
20
|
Abstract
Colorectal cancer is one of the commonest malignancies in the "developed" world. The liver constitutes the main host organ for its distant metastases which, when present, augur a bad prognosis for the disease. Kupffer cells (KCs) are macrophages that constantly reside within the liver and form an effective first line defence against multiple harmful agents which reach the hepatic sinusoids via the portal circulation. KCs remove chemical compounds and dead or damaged cells, eliminate bacteria and protect against invading tumour cells. They may play a crucial tumouricidal role, exerting cytotoxic and cytostatic functions through the release of multiple cytokines and chemokines. Subsequently, colorectal metastasising cells are destroyed either by KC-performed phagocytosis or via the stimulation of other immune cells which migrate into the sinusoids and act accordingly. On the contrary, KC products, including cytokines, growth factors and matrix-degrading enzymes may promote liver metastasis, supporting tumour cell extravasation, motility and invasion. Current research aims to exploit the antineoplastic properties of KCs in new therapeutic approaches of colorectal cancer liver metastasis. Numerous agents, such as the granulocyte macrophage-colony stimulating factor, interferon gamma, muramyl peptide analogues and various antibody based treatments, have been tested in experimental models with promising results. Future trials may investigate their use in everyday clinical practice and compare their therapeutic value with current treatment of the disease.
Collapse
Affiliation(s)
- Konstantinos A Paschos
- Liver Research Group, Section of Oncology, School of Medicine, Royal Hallamshire Hospital, The University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
21
|
Nanni P, Nicoletti G, Landuzzi L, Croci S, Murgo A, Palladini A, Antognoli A, Ianzano ML, Stivani V, Grosso V, Maira SM, García-Echeverría C, Scotlandi K, De Giovanni C, Lollini PL. High metastatic efficiency of human sarcoma cells in Rag2/gammac double knockout mice provides a powerful test system for antimetastatic targeted therapy. Eur J Cancer 2009; 46:659-68. [PMID: 20031388 DOI: 10.1016/j.ejca.2009.11.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 11/13/2009] [Accepted: 11/19/2009] [Indexed: 11/20/2022]
Abstract
Immunodeficient animal models are invaluable tools to investigate the metastatic propensity of human tumours. However residual immune responses, in particular natural killer (NK) cells, severely hamper the traffic and growth of human tumour cells. We studied whether a genetically modified mouse host lacking T, B and NK immunity allowed an improved expression of the metastatic phenotype of malignant human tumours. Metastatic spread of a panel of human sarcoma cell lines was studied in double knockout Rag2(-/-);gammac(-/-) mice in comparison with NK-depleted nude mice. Rag2(-/-);gammac(-/-) mice receiving intravenous (i.v.) or subcutaneous (s.c.) human sarcoma cell lines developed extensive multiorgan metastases. Metastatic efficiency in Rag2(-/-);gammac(-/-) was superior than in nude mice in terms of both metastatic sites and metastasis number. Metastatic growth in Rag2(-/-);gammac(-/-) mice was faster than that in nude mice, thus allowing an earlier metastasis evaluation. Most human sarcomas metastasised in the liver of Rag2(-/-);gammac(-/-) mice, a kind of organ preference undetectable in nude mice and specific of sarcomas, as several carcinoma cell lines failed to colonise the liver of Rag2(-/-);gammac(-/-) mice, independently of their metastatic spread to other sites. In vitro analysis of the molecular mechanisms of liver metastasis of sarcomas implicated liver-produced growth and motility factors, in particular the insulin-like growth factor (IGF) axis. NVP-BEZ235, a specific inhibitor of downstream signal transduction targeting PI3K and mTOR, strongly inhibited liver metastasis of human sarcoma cells. In conclusion, the Rag2(-/-);gammac(-/-) mouse model allowed the expression of human metastatic phenotypes inapparent in conventional immunodeficient mice and the preclinical testing of appropriate targeted therapies.
Collapse
Affiliation(s)
- Patrizia Nanni
- Cancer Research Section, Department of Experimental Pathology, Viale Filopanti 22, University of Bologna, 40126 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Luo Y, Ohmori H, Fujii K, Moriwaka Y, Sasahira T, Kurihara M, Tatsumoto N, Sasaki T, Yamashita Y, Kuniyasu H. HMGB1 attenuates anti-metastatic defence of the liver in colorectal cancer. Eur J Cancer 2009; 46:791-9. [PMID: 20018503 DOI: 10.1016/j.ejca.2009.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2009] [Revised: 11/12/2009] [Accepted: 11/19/2009] [Indexed: 12/15/2022]
Abstract
High mobility group box (HMGB) 1 induces apoptosis of monocyte-lineage cells. We examined the effect of HMGB1 on Kupffer cells (KCs). In 50 Dukes C and 12 liver-metastasised Dukes D colorectal cancers (CRCs), higher HMGB1 concentration in the primary tumours and metastatic foci, and fewer KCs were found in Dukes D cases than in Dukes C cases. The portal blood HMGB1 concentration was higher in Dukes D cases than in Dukes C cases. HMGB1 induced growth inhibition and apoptosis in mouse KCs in a dose-dependent manner, which was associated with the phosphorylation of c-Jun N-terminal kinase (JNK). JNK inhibition and knockdown of HMGB1 receptor abrogated growth inhibition and apoptosis. In a nude mouse liver metastasis model, the caecal administration of HMGB1 decreased the number of KCs and increased the embedment of Colo320 CRC cells in a dose-dependent manner. HMGB1 transfection increased the liver metastasis of Colo320 cells, and the metastasis was inhibited by anti-HMGB1 antibody administration. These results suggest that HMGB1 secreted from primary tumours decreases the number of KCs and attenuates the anti-metastatic defence of the liver in patients with CRCs.
Collapse
Affiliation(s)
- Yi Luo
- Department of Molecular Pathology, Nara Medical University, Kashihara, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Otten MA, van der Bij GJ, Verbeek SJ, Nimmerjahn F, Ravetch JV, Beelen RHJ, van de Winkel JGJ, van Egmond M. Experimental Antibody Therapy of Liver Metastases Reveals Functional Redundancy between FcγRI and FcγRIV. THE JOURNAL OF IMMUNOLOGY 2008; 181:6829-36. [DOI: 10.4049/jimmunol.181.10.6829] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
24
|
Abstract
The renin-angiotensin system (RAS) is usually associated with its systemic action on cardiovascular homoeostasis. However, recent studies suggest that at a local tissue level, the RAS influences tumour growth. The potential of the RAS as a target for cancer treatment and the suggested underlying mechanisms of its paracrine effects are reviewed here. These include modulation of angiogenesis, cellular proliferation, immune responses and extracellular matrix formation. Knowledge of the RAS has increased dramatically in recent years with the discovery of new enzymes, peptides and feedback mechanisms. The local RAS appears to influence tumour growth and metastases and there is evidence of tissue- and tumour-specific differences. Recent experimental studies provide strong evidence that drugs that inhibit the RAS have the potential to reduce cancer risk or retard tumour growth and metastases. Manipulation of the RAS may, therefore, provide a safe and inexpensive anticancer strategy.
Collapse
Affiliation(s)
- Eleanor I Ager
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria 3084, Australia.
| | | | | |
Collapse
|
25
|
Nitric oxide and MCP-1 regulation in LPS activated rat Kupffer cells. Mol Cell Biochem 2008; 319:91-8. [DOI: 10.1007/s11010-008-9881-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Accepted: 07/03/2008] [Indexed: 01/16/2023]
|
26
|
Lee Y, Lee JS, Kim CM, Jeong JY, Choi JI, Kim MJ. Area of paradoxical signal drop after the administration of superparamagnetic iron oxide on the T2-weighted image of a patient with lymphangitic metastasis of the liver. Magn Reson Imaging 2008; 26:577-82. [PMID: 18093780 DOI: 10.1016/j.mri.2007.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 09/25/2007] [Accepted: 10/25/2007] [Indexed: 10/22/2022]
Abstract
We report a geographic area of prominent hypointensity in T2-weighted images vs. normal adjacent liver parenchyma in a patient with cholangiocarcinoma and lymphangitic metastasis of the liver after superparamagnetic iron oxide (SPIO) administration. The area showing this prominent signal drop showed Kupffer cell proliferation and lymphangitic metastasis during a pathologic examination.
Collapse
Affiliation(s)
- Youkyung Lee
- Department of Radiology and Institute of Radiation Medicine, Seoul National University Hospital, Chongno-gu, Seoul 110-774, Korea
| | | | | | | | | | | |
Collapse
|
27
|
Anselmi K, Stolz DB, Nalesnik M, Watkins SC, Kamath R, Gandhi CR. Gliotoxin causes apoptosis and necrosis of rat Kupffer cells in vitro and in vivo in the absence of oxidative stress: exacerbation by caspase and serine protease inhibition. J Hepatol 2007; 47:103-13. [PMID: 17466404 PMCID: PMC2764960 DOI: 10.1016/j.jhep.2007.02.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 02/12/2007] [Accepted: 02/22/2007] [Indexed: 12/18/2022]
Abstract
BACKGROUND/AIMS A potential application of gliotoxin therapy for liver fibrosis was suggested by its apoptotic effect on fibrogenic activated stellate cells. We investigated if gliotoxin exerts similar effects on hepatic macrophage Kupffer cells. METHODS Effects of gliotoxin on Kupffer cells isolated from the normal liver and in vivo following its administration to CCl(4)-induced cirrhotic rats were studied. RESULTS Gliotoxin caused apoptosis of cultured Kupffer cells, the effect being apparent at 0.3 microM concentration within 1h; longer incubation caused necrosis. This effect was associated with mitochondrial cytochrome c release, caspase-3 activation and ATP depletion. Interestingly, inhibition of caspase-3 and serine proteases accelerated and augmented gliotoxin-induced cell death via necrosis. Gliotoxin stimulated nuclear translocation of NFkappaB, and phosphorylation of p38, ERK1/2 and JNK MAP kinases, but these signaling molecules were not involved in gliotoxin-induced death of Kupffer cells. In vivo administration of gliotoxin to cirrhotic rats caused apoptosis of Kupffer cells, stellate cells and hepatocytes. In control rats, the effect was minimal on the nonparenchymal cells and not apparent on hepatocytes. CONCLUSIONS In the fibrotic liver, gliotoxin nonspecifically causes death of hepatic cell types. Modification of gliotoxin molecule may be necessary for selective targeting and elimination of activated stellate cells.
Collapse
Affiliation(s)
- Kristin Anselmi
- Thomas E. Starzl Transplantation Institute, Department of Surgery, and VA medical Center, University of Pittsburgh, E-1518 BST, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Kupffer cells (KC) constitute 80-90% of the tissue macrophages present in the body. They reside within the lumen of the liver sinusoids, and are therefore constantly exposed to gut-derived bacteria, microbial debris and bacterial endotoxins, known to activate macrophages. Upon activation KC release various products, including cytokines, prostanoides, nitric oxide and reactive oxygen species. These factors regulate the phenotype of KC themselves, and the phenotypes of neighboring cells, such as hepatocytes, stellate cells, endothelial cells and other immune cells that traffic through the liver. Therefore, KC are intimately involved in the liver's response to infection, toxins, ischemia, resection and other stresses. This review summarizes established basic concepts of KC function as well as their role in the pathogenesis of various liver diseases.
Collapse
Affiliation(s)
- Manfred Bilzer
- Department of Medicine II, University of Munich, Klinikum Grosshadern, Munich, Germany.
| | | | | |
Collapse
|