1
|
Brown NE, Jones A, Hunt BG, Waltz SE. Prostate tumor RON receptor signaling mediates macrophage recruitment to drive androgen deprivation therapy resistance through Gas6-mediated Axl and RON signaling. Prostate 2022; 82:1422-1437. [PMID: 35860905 PMCID: PMC9492645 DOI: 10.1002/pros.24416] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/25/2022] [Accepted: 07/05/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Androgen deprivation therapy (ADT), or chemical castration, is the first-line therapy for prostate cancer; however, resistance leaves few treatment options. Prostatic tumor-associated macrophages (TAMs) have been shown to promote prostate cancer growth and are abundant in castration-resistant prostate cancer (CRPC), suggesting a role in promoting CRPC. We recently showed a tumor cell-intrinsic mechanism by which RON promotes CRPC. Given previous reports that RON alters prostate cancer cell chemokine production and RON-overexpressing tumors alter macrophage function, we hypothesized that a macrophage-dependent mechanism regulated by tumor cell intrinsic RON also promotes CRPC. METHODS Using RON-modulated genetically engineered mouse models (GEMMs) and GEMM-derived cell lines and co-cultures with bone marrow-derived macrophages, we show functional and molecular characteristics of signaling pathways in supporting CRPC. Further, we used an unbiased phosphokinase array to identify pathway interactions regulated by RON. Finally, using human prostate cancer cell lines and prostate cancer patient data sets, we show the relevance of our findings to human prostate cancer. RESULTS Studies herein show that macrophages recruited into the prostate tumor microenvironment (TME) serve as a source for Gas6 secretion which serves to further enhance RON and Axl receptor activation in prostate tumor cells thereby driving CRPC. Further, we show targeting RON and macrophages in a murine model promotes CRPC sensitization to ADT. CONCLUSIONS We discovered a novel role for the RON receptor in prostate cancer cells in promoting CRPC through the recruitment of macrophages into the prostate TME. Macrophage-targeting agents in combination with RON/Axl inhibition are likely to provide clinical benefits for patients with CRPC.
Collapse
Affiliation(s)
- Nicholas E. Brown
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Angelle Jones
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Brian G. Hunt
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Susan E. Waltz
- Department of Cancer BiologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Research ServiceCincinnati Veterans Affairs Medical CenterCincinnatiOhioUSA
| |
Collapse
|
2
|
Hunt BG, Jones A, Lester C, Davis JC, Benight NM, Waltz SE. RON ( MST1R) and HGFL ( MST1) Co-Overexpression Supports Breast Tumorigenesis through Autocrine and Paracrine Cellular Crosstalk. Cancers (Basel) 2022; 14:2493. [PMID: 35626096 PMCID: PMC9140067 DOI: 10.3390/cancers14102493] [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: 04/05/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Aberrant RON signaling is present in numerous cancers including breast cancer. Evidence suggests that the ligand, hepatocyte growth factor-like (HGFL), is also overexpressed in breast cancer. RON (MST1R) and HGFL (MST1) genes are located on human chromosome 3 and mouse chromosome 9 respectively and are found near each other in both species. Based on co-expression patterns, we posited that RON and HGFL are co-regulated and that coordinate upregulation drives aggressive tumorigenesis. METHODS Mouse models were used to establish the functional significance of RON and HGFL co-overexpression on the activation of tumor cells and tumor-associated macrophages in breast cancer. TCGA and METABRIC gene expression and alteration data were used to query the relationships between MST1R and MST1 in breast cancer. RESULTS In tumor models, physiologic sources of HGFL modestly improve Arginase-1+ (M2) macrophage recruitment to the tumor proper. Tumor-cell produced HGFL functions in autocrine to sustain tumor cell RON activation and MAPK-dependent secretion of chemotactic factors and in paracrine to activate RON on macrophages and to promote breast cancer stem cell self-renewal. In silico analyses support that RON and HGFL are co-expressed across virtually all cancer types including breast cancer and that common genomic alterations do not appear to be drivers of RON/HGFL co-overexpression. CONCLUSIONS Co-overexpression of RON and HGFL in breast cancer cells (augmented by physiologic sources of HGFL) promotes tumorigenesis through autocrine-mediated RON activation/RON-dependent secretome changes and paracrine activation of macrophage RON to promote breast cancer stem cell self-renewal.
Collapse
Affiliation(s)
- Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
| | - Angelle Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
| | - James C. Davis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
| | - Nancy M. Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (B.G.H.); (A.J.); (C.L.); (J.C.D.); (N.M.B.)
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
3
|
Cazes A, Childers BG, Esparza E, Lowy AM. The MST1R/RON Tyrosine Kinase in Cancer: Oncogenic Functions and Therapeutic Strategies. Cancers (Basel) 2022; 14:cancers14082037. [PMID: 35454943 PMCID: PMC9027306 DOI: 10.3390/cancers14082037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary MST1R/RON receptor tyrosine kinase is a highly conserved transmembrane protein present on epithelial cells, macrophages, and recently identified in a T-cell subset. RON activation attenuates inflammation in healthy tissue. Interestingly, it is overexpressed in several epithelial neoplasms with increasing levels of expression associated with worse outcomes. Though the mechanisms involved are still under investigation, RON is involved in carcinogenesis via immune modulation of the immune tumor microenvironment, activation of numerous oncogenic pathways, and is protective under cellular stress. Alternatively, inhibition of RON abrogates tumor progression in both animal and human tissue models. Given this, RON is a targetable protein of great interest for cancer treatment. Here, we review RON’s function in tissue inflammation and cancer progression, and review cancer clinical trials to date that have used agents targeting RON signaling. Abstract The MST1R/RON receptor tyrosine kinase is a homologue of the more well-known MET receptor. Like MET, RON orchestrates cell signaling pathways that promote oncogenesis and enable cancer cell survival; however, it has a more unique role in the regulation of inflammation. RON was originally described as a transmembrane receptor expressed on tissue resident macrophages and various epithelial cells. RON is overexpressed in a variety of cancers and its activation modifies multiple signaling pathways with resultant changes in epithelial and immune cells which together modulate oncogenic phenotypes. While several RON isoforms have been identified with differences in structure, activation, and pathway regulation, increased RON expression and/or activation is consistently associated with worse outcomes. Tyrosine kinase inhibitors targeting RON have been developed, making RON an actionable therapeutic target.
Collapse
|
4
|
Ruiz-Torres SJ, Bourn JR, Benight NM, Hunt BG, Lester C, Waltz SE. Macrophage-mediated RON signaling supports breast cancer growth and progression through modulation of IL-35. Oncogene 2022; 41:321-333. [PMID: 34743208 PMCID: PMC8758553 DOI: 10.1038/s41388-021-02091-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 01/20/2023]
Abstract
Tumor associated macrophages (TAMs) play a major role in regulating mammary tumor growth and in directing the responses of tumor infiltrating leukocytes in the microenvironment. However, macrophage-specific mechanisms regulating the interactions of macrophages with tumor cells and other leukocytes that support tumor progression have not been extensively studied. In this study, we show that the activation of the RON receptor tyrosine kinase signaling pathway specifically in macrophages supports breast cancer growth and metastasis. Using clinically relevant murine models of breast cancer, we demonstrate that loss of macrophage RON expression results in decreases in mammary tumor cell proliferation, survival, cancer stem cell self-renewal, and metastasis. Macrophage RON signaling modulates these phenotypes via direct effects on the tumor proper and indirectly by regulating leukocyte recruitment including macrophages, T-cells, and B-cells in the mammary tumor microenvironment. We further show that macrophage RON expression regulates the macrophage secretome including IL-35 and other immunosuppressive factors. Overall, our studies implicate activation of RON signaling in macrophages as a key player in supporting a thriving mammary pro-tumor microenvironment through novel mechanisms including the augmentation of tumor cell properties through IL-35.
Collapse
Affiliation(s)
- Sasha J. Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Jennifer R. Bourn
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Nancy M. Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA,Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA,Address correspondence to: Susan E. Waltz, PhD, Department of Cancer Biology, Vontz Center for Molecular Studies, University of Cincinnati College of Medicine, 3125 Eden Ave, Cincinnati, OH 45267-0521, Tel: 513.558.8675,
| |
Collapse
|
5
|
Bourn JR, Ruiz-Torres SJ, Hunt BG, Benight NM, Waltz SE. Tumor cell intrinsic RON signaling suppresses innate immune responses in breast cancer through inhibition of IRAK4 signaling. Cancer Lett 2021; 503:75-90. [PMID: 33508385 PMCID: PMC7981256 DOI: 10.1016/j.canlet.2021.01.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/11/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
Increasing evidence suggests that cancer cells require both alterations in intrinsic cellular processes and the tumor microenvironment for tumor establishment, growth, and progression to metastatic disease. Despite this, knowledge of tumor-cell intrinsic molecular mechanisms controlling both tumor cell processes as well as the tumor microenvironment is limited. In this study, we provide evidence demonstrating the novel role of RON signaling in regulating breast cancer initiation, progression, and metastasis through modulation of tumor cell intrinsic processes and the tumor microenvironment. Using clinically relevant models of breast cancer, we show that RON signaling in the mammary epithelial tumor cells promotes tumor cell survival and proliferation as well as an immunopermissive microenvironment associated with decreased M1 macrophage, natural killer (NK) cell, and CD8+ T cell recruitment. Moreover, we demonstrate that RON signaling supports these phenotypes through novel mechanisms involving suppression of IRAK4 signaling and inhibition of type I Interferons. Our studies indicate that activation of RON signaling within breast cancer cells promotes tumor cell intrinsic growth and immune evasion which support breast cancer progression and highlight the role of targeting RON signaling as a potential therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Jennifer R Bourn
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Sasha J Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Brian G Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Nancy M Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267-0521, USA; Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH, 45267, USA.
| |
Collapse
|
6
|
Huang L, Fang X, Shi D, Yao S, Wu W, Fang Q, Yao H. MSP-RON Pathway: Potential Regulator of Inflammation and Innate Immunity. Front Immunol 2020; 11:569082. [PMID: 33117355 PMCID: PMC7577085 DOI: 10.3389/fimmu.2020.569082] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Macrophage-stimulating protein (MSP), a soluble protein mainly synthesized by the liver, is the only known ligand for recepteur d'origine nantais (RON), which is a member of the MET proto-oncogene family. Recent studies show that the MSP-RON signaling pathway not only was important in tumor behavior but also participates in the occurrence or development of many immune system diseases. Activation of RON in macrophages results in the inhibition of nitric oxide synthesis as well as lipopolysaccharide (LPS)-induced inflammatory response. MSP-RON is also associated with chronic inflammatory responses, especially chronic liver inflammation, and might serve as a novel regulator of inflammation, which may affect the metabolism in the body. Another study provided evidence of the relationship between MSP-RON and autoimmune diseases, suggesting a potential role for MSP-RON in the development of drugs for autoimmune diseases. Moreover, MSP-RON plays an important role in maintaining the stability of the tissue microenvironment and contributes to immune escape in the tumor immune microenvironment. Here, we summarize the role of MSP-RON in immunity, based on recent findings, and lay the foundation for further research.
Collapse
Affiliation(s)
- Lingtong Huang
- Department of Critical Care Units, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueling Fang
- Department of Critical Care Units, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danrong Shi
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuhao Yao
- Department of Stormotologry, Wenzhou Medical University Renji College, Wenzhou, China
| | - Weifang Wu
- Department of Critical Care Units, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Fang
- Department of Critical Care Units, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hangping Yao
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Millar R, Kilbey A, Remak S, Severson TM, Dhayade S, Sandilands E, Foster K, Bryant DM, Blyth K, Coffelt SB. The MSP-RON axis stimulates cancer cell growth in models of triple negative breast cancer. Mol Oncol 2020; 14:1868-1880. [PMID: 32484599 PMCID: PMC7400785 DOI: 10.1002/1878-0261.12734] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/30/2020] [Accepted: 05/28/2020] [Indexed: 11/09/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis and high rates of relapse. The lack of actionable targets for TNBC has contributed to the high mortality rates of this disease, and new candidate molecules for potential manipulation are urgently required. Here, we show that macrophage-stimulating protein (MSP) and its tyrosine kinase receptor, Recepteur d'origine nantais (RON), are potent drivers of cancer cell growth and tumor progression in a mouse model of TNBC driven by the loss of Trp53 and Brca1. After comparison of two genetically engineered mouse models of TNBC, we found that mammary tumors from K14-Cre;Brca1F/F ;Trp53F/F (KB1P) mice exhibit high endogenous levels of MSP and RON expression. We show that MSP stimulates serine/threonine kinase 1 and extracellular regulated MAPK activation as well as cancer cell growth in cell lines derived from the two mouse models, while genetic and pharmacological inhibition of RON prevents these effects. Similarly, KB1P tumor progression in mice was robustly attenuated by treatment with a RON inhibitor with accompanied reduction in the proliferation marker, Ki-67. Analysis of human gene expression data confirmed that the genes encoding MSP and RON are robustly expressed in human TNBC as well as other subsets of breast cancer. Our findings uncover a mouse model where MSP expression and RON expression are naturally increased, and they provide evidence that this receptor and its ligand are viable candidate molecules for targeted treatment of breast cancer.
Collapse
Affiliation(s)
- Rhona Millar
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Anna Kilbey
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Sarah‐Jane Remak
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Tesa M. Severson
- Division of OncogenesisNetherlands Cancer InstituteAmsterdamThe Netherlands
| | | | - Emma Sandilands
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Kyla Foster
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - David M. Bryant
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Karen Blyth
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| | - Seth B. Coffelt
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
| |
Collapse
|
8
|
Antibody-drug conjugates targeting RON receptor tyrosine kinase as a novel strategy for treatment of triple-negative breast cancer. Drug Discov Today 2020; 25:1160-1173. [PMID: 32479905 DOI: 10.1016/j.drudis.2020.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/05/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Treatment of triple-negative breast cancer (TNBC) is a challenge to oncologists. Currently, the lack of effective therapy has fostered a major effort to discover new targets and therapeutics to combat this disease. The recepteur d'origine nantais (RON) receptor has been implicated in the pathogenesis of TNBC. Clinical studies have revealed that aberrant RON expression is crucial in regulating TNBC malignant phenotypes. Increased RON expression also has prognostic value for breast cancer progress. These features provide the rationale to target RON for TNBC treatment. In this review, we discuss the importance of RON in TNBC tumorigenesis and the development of anti-RON antibody-drug conjugates (ADCs) for clinical application. The findings from preclinical studies lay the foundation for clinical trials of this novel biotherapeutic for TNBC therapy.
Collapse
|
9
|
Sullivan C, Brown NE, Vasiliauskas J, Pathrose P, Starnes SL, Waltz SE. Prostate Epithelial RON Signaling Promotes M2 Macrophage Activation to Drive Prostate Tumor Growth and Progression. Mol Cancer Res 2020; 18:1244-1254. [PMID: 32439702 DOI: 10.1158/1541-7786.mcr-20-0060] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/11/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
Abstract
Effective treatment of advanced prostate cancer persists as a significant clinical need as only 30% of patients with distant disease survive to 5 years after diagnosis. Targeting signaling and tumor cell-immune cell interactions in the tumor microenvironment has led to the development of powerful immunotherapeutic agents, however, the prostate tumor milieu remains impermeable to these strategies highlighting the need for novel therapeutic targets. In this study, we provide compelling evidence to support the role of the RON receptor tyrosine kinase as a major regulator of macrophages in the prostate tumor microenvironment. We show that loss of RON selectively in prostate epithelial cells leads to significantly reduced prostate tumor growth and metastasis and is associated with increased intratumor infiltration of macrophages. We further demonstrate that prostate epithelial RON loss induces transcriptional reprogramming of macrophages to support expression of classical M1 markers and suppress expression of alternative M2 markers. Interestingly, our results show epithelial RON activation drives upregulation of RON expression in macrophages as a positive feed-forward mechanism to support prostate tumor growth. Using 3D coculture assays, we provide additional evidence that epithelial RON expression coordinates interactions between prostate tumor cells and macrophages to promote macrophage-mediated tumor cell growth. Taken together, our results suggest that RON receptor signaling in prostate tumor cells directs the functions of macrophages in the prostate tumor microenvironment to promote prostate cancer. IMPLICATIONS: Epithelial RON is a novel immunotherapeutic target that is responsible for directing the macrophage antitumor immune response to support prostate tumor growth and progression.
Collapse
Affiliation(s)
- Camille Sullivan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Nicholas E Brown
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Juozas Vasiliauskas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Peterson Pathrose
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Sandra L Starnes
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio
| |
Collapse
|
10
|
MST1R (RON) expression is a novel prognostic biomarker for metastatic progression in breast cancer patients. Breast Cancer Res Treat 2020; 181:529-540. [PMID: 32342233 DOI: 10.1007/s10549-020-05653-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE This study evaluates the prognostic significance of MST1R (RON) expression in breast cancer with respect to disease progression, long-term survival, subtype, and association with conventional prognostic factors. METHODS The approach includes interrogation of survival and tumor staging with paired MST1R RNA expression from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets. Protein expression evaluation was performed using immunohistochemistry (IHC) staining of MST1R on breast cancer tissue samples from the Cancer Diagnosis Program Breast Cancer Progression tissue microarray and locally obtained breast tumor tissue samples analyzed with paired survival, metastasis, and subtype. RESULTS Data from TCGA (n = 774) show poorer relapse-free survival (RFS) in patients with high MST1R expression (P = 0.32) and no difference in MST1R expression based on tumor stage (P = 0.77) or nodal status (P = 0.94). Patients in the GEO-derived Kaplan-Meier Plotter microarray dataset demonstrate the association of MST1R and poorer overall survival (n = 1402, P = 0.018) and RFS in patients receiving chemotherapy (n = 798, P = 0.041). Patients with high MST1R expression display worse overall survival (P = 0.01) and receiver operator characteristic (ROC) analysis demonstrate the predictive capacity of increased MST1R with early death (P = 0.0017) in IHC-stained samples. Paired IHC-stained breast tumor samples from the primary versus metastatic site show MST1R expression is associated with metastatic progression (P = 0.032), and ROC analysis supports the predictive capacity of MST1R in metastatic progression (P = 0.031). No associations of MST1R with estrogen receptor (ER), progesterone receptor (PR), both ER and PR, HER2 positivity, or triple-negativity were found (P = 0.386, P = 0.766, P = 0.746, P = 0.457, P = 0.947, respectively). CONCLUSIONS MST1R expression has prognostic value in breast cancer with respect to survival and metastatic progression. MST1R expression is not associated with tumor stage, nodal status, or subtype.
Collapse
|
11
|
Babicky ML, Harper MM, Chakedis J, Cazes A, Mose ES, Jaquish DV, French RP, Childers B, Alakus H, Schmid MC, Foubert P, Miyamoto J, Holman PJ, Walterscheid ZJ, Tang CM, Varki N, Sicklick JK, Messer K, Varner JA, Waltz SE, Lowy AM. MST1R kinase accelerates pancreatic cancer progression via effects on both epithelial cells and macrophages. Oncogene 2019; 38:5599-5611. [PMID: 30967626 PMCID: PMC6625868 DOI: 10.1038/s41388-019-0811-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022]
Abstract
The MST1R (RON) kinase is overexpressed in >80% of human pancreatic cancers, but its role in pancreatic carcinogenesis is unknown. In this study, we examined the relevance of Mst1r kinase to Kras driven pancreatic carcinogenesis using genetically engineered mouse models. In the setting of mutant Kras, Mst1r overexpression increased acinar-ductal metaplasia (ADM), accelerated progression of pancreatic intraepithelial neoplasia (PanIN), and resulted in the accumulation of (mannose receptor C type 1) MRC1+, (arginase 1) Arg+ macrophages in the tumor microenvironment. Conversely, absence of a functional Mst1r kinase slowed PanIN initiation, resulted in smaller tumors, prolonged survival and a reduced tumor associated macrophage content. Mst1r expression was associated with increased production of its ligand Mst1, and in orthotopic models, suppression of Mst1 expression resulted in reduced tumor size, changes in macrophage polarization and enhanced T cell infiltration. This study demonstrates the functional significance of Mst1r during pancreatic cancer initiation and progression. Further, it provides proof of concept that targeting Mst1r can modulate pancreatic cancer growth and the microenvironment. This study provides further rationale for targeting Mst1r as a therapeutic strategy.
Collapse
Affiliation(s)
- Michele L Babicky
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Megan M Harper
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jeffery Chakedis
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alex Cazes
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Evangeline S Mose
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dawn V Jaquish
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Randall P French
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Betzaira Childers
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hakan Alakus
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael C Schmid
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Phillippe Foubert
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jaclyn Miyamoto
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Patrick J Holman
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Zakkary J Walterscheid
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Chih-Min Tang
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nissi Varki
- Department of Family Medicine and Epidemiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jason K Sicklick
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Karen Messer
- Department of Family Medicine and Epidemiology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Judith A Varner
- Department of Pathology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, and Research Service, Cincinnati Veteran's Administration Medical Center, Cincinnati, OH, 45267, USA
| | - Andrew M Lowy
- Division of Surgical Oncology, Department of Surgery, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
12
|
Brown NE, Sullivan C, Waltz SE. Therapeutic Considerations for Ron Receptor Expression in Prostate Cancer. EMS CANCER SCIENCE JOURNAL 2018; 1:003. [PMID: 30775725 PMCID: PMC6377156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
INTRODUCTION The Ron receptor tyrosine kinase was initially discovered as a protein which played a critical role in regulating inflammatory responses. This effect was primarily determined through studies in various macrophage populations. Since its initial discovery, a role has emerged for Ron as a driver of cancer within epithelial cells. After numerous publications have detailed a role for Ron in promoting tumor initiation, growth, and metastasis, Ron has been designated as an emerging therapeutic option in a variety of cancers. AREAS COVERED This review discusses the current literature regarding the role of Ron in prostate cancer and places special emphasis on the role of Ron in both epithelial cells and macrophages. Whole body loss of Ron signaling initially exposed a variety of prostate cancer growth mechanisms regulated by Ron. With the knowledge that Ron plays an integral part in regulating the function of epithelial cells and macrophages, studies commenced to discern the cell type specific functions for Ron in prostate cancer. A novel role for Ron in promoting Castration Resistant Prostate Cancer has recently been uncovered, and the results of these studies are summarized herein. Furthermore, this review gives a summary of several currently available compounds which show promise at targeting Ron in both epithelial and macrophage populations. OUTLOOK Sufficient evidence has been provided for the initiation of clinical trials focused on targeting Ron in both macrophage and epithelial compartments for the treatment of prostate cancer. A number of therapeutic avenues for targeting Ron in prostate cancer are currently available; however, special consideration will need to take place knowing that Ron signaling impacts multiple cell types. Further understanding of the cell type specific functions of Ron in prostate cancer will help inform and shape future clinical research and therapeutic strategies.
Collapse
Affiliation(s)
- Nicholas E. Brown
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Camille Sullivan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA
| |
Collapse
|
13
|
Ekiz HA, Lai SCA, Gundlapalli H, Haroun F, Williams MA, Welm AL. Inhibition of RON kinase potentiates anti-CTLA-4 immunotherapy to shrink breast tumors and prevent metastatic outgrowth. Oncoimmunology 2018; 7:e1480286. [PMID: 30228950 PMCID: PMC6140584 DOI: 10.1080/2162402x.2018.1480286] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/17/2018] [Accepted: 05/20/2018] [Indexed: 12/16/2022] Open
Abstract
The advent of immune checkpoint blockade as a new strategy for immunotherapy has changed the outlook for many aggressive cancers. Although complete tumor eradication is attainable in some cases, durable clinical responses are observed only in a small fraction of patients, underlining urgent need for improvement. We previously showed that RON, a receptor tyrosine kinase expressed in macrophages, suppresses antitumor immune responses, and facilitates progression and metastasis of breast cancer. Here, we investigated the molecular changes that occur downstream of RON activation in macrophages, and whether inhibition of RON can cooperate with checkpoint immunotherapy to eradicate tumors. Activation of RON by its ligand, MSP, altered the gene expression profile of macrophages drastically and upregulated surface levels of CD80 and PD-L1, ligands for T-cell checkpoint receptors CTLA-4 and PD-1. Genetic deletion or pharmacological inhibition of RON in combination with anti-CTLA-4, but not with anti-PD-1, resulted in improved clinical responses against orthotopically transplanted tumors compared to single-agent treatment groups, resulting in complete tumor eradication in 46% of the animals. Positive responses to therapy were associated with higher levels of T-cell activation markers and tumor-infiltrating lymphocytes. Importantly, co-inhibition of RON and anti-CTLA-4 was also effective in clearing metastatic breast cancer cells in lungs, resulting in clinical responses in nearly 60% of the mice. These findings suggest that RON inhibition can be a novel approach to potentiate responses to checkpoint immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Huseyin Atakan Ekiz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Shu-Chin Alicia Lai
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Harika Gundlapalli
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Fadi Haroun
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Matthew A Williams
- Department of Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Ruiz-Torres SJ, Benight NM, Karns RA, Lower EE, Guan JL, Waltz SE. HGFL-mediated RON signaling supports breast cancer stem cell phenotypes via activation of non-canonical β-catenin signaling. Oncotarget 2017; 8:58918-58933. [PMID: 28938607 PMCID: PMC5601703 DOI: 10.18632/oncotarget.19441] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/11/2017] [Indexed: 12/16/2022] Open
Abstract
Breast cancer stem cells (BCSCs), which drive tumor progression, recurrence, and metastasis, are considered a major challenge for breast cancer treatments, thus the discovery of novel pathways regulating BCSC maintenance remains essential to develop new strategies to effectively target this population and combat disease mortality. The HGFL-RON signaling is overexpressed in human breast cancers and is associated with increased breast cancer progression, metastasis, and poor prognosis. Here, we report that overexpression of RON/MST1R and HGFL/MST1 in cell lines and primary tumors increases BCSC self-renewal, numbers, and tumorigenic potential after syngeneic transplantation. Transcriptome analyses also reveal that the HGFL-RON signaling pathway regulates additional BCSC functions and supports an immunosuppressive microenvironment to stimulate tumor formation and progression. Moreover, we show that genetic and chemical downregulation of HGFL-RON signaling disrupts BCSC phenotypes and tumor growth by suppressing the RON-mediated phosphorylation/activation of β-CATENIN/CTNNB1 and its effector NF-κB/RELA. These studies indicate that HGFL-RON signaling regulates BCSC phenotypes and the tumor microenvironment to drive tumorigenesis and present HGFL/RON as novel therapeutic targets to effectively eradicate BCSCs in patients.
Collapse
Affiliation(s)
- Sasha J Ruiz-Torres
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Nancy M Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Rebekah A Karns
- Division of Bioinformatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.,Research Service, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45267, USA
| |
Collapse
|