1
|
Dou T, Li J, Zhang Y, Pei W, Zhang B, Wang B, Wang Y, Jia H. The cellular composition of the tumor microenvironment is an important marker for predicting therapeutic efficacy in breast cancer. Front Immunol 2024; 15:1368687. [PMID: 38487526 PMCID: PMC10937353 DOI: 10.3389/fimmu.2024.1368687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/19/2024] [Indexed: 03/17/2024] Open
Abstract
At present, the incidence rate of breast cancer ranks first among new-onset malignant tumors in women. The tumor microenvironment is a hot topic in tumor research. There are abundant cells in the tumor microenvironment that play a protumor or antitumor role in breast cancer. During the treatment of breast cancer, different cells have different influences on the therapeutic response. And after treatment, the cellular composition in the tumor microenvironment will change too. In this review, we summarize the interactions between different cell compositions (such as immune cells, fibroblasts, endothelial cells, and adipocytes) in the tumor microenvironment and the treatment mechanism of breast cancer. We believe that detecting the cellular composition of the tumor microenvironment is able to predict the therapeutic efficacy of treatments for breast cancer and benefit to combination administration of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, China
| | - Binyue Zhang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Wang
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Kim J, Koh DI, Lee M, Park YS, Hong SW, Shin JS, Lee MS, Kim MH, Lee JH, Jeong J, Bae S, Hong JK, Jeong HR, Ryu YS, Kim SM, Choi M, Kim H, Ryu H, Hur SC, Park J, Hur DY, Jin DH. Targeting isoforms of RON kinase (MST1R) drives antitumor efficacy. Cell Death Differ 2023; 30:2491-2507. [PMID: 37926711 PMCID: PMC10733321 DOI: 10.1038/s41418-023-01235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Recepteur d'origine nantais (RON, MST1R) is a single-span transmembrane receptor tyrosine kinase (RTK) aberrantly expressed in numerous cancers, including various solid tumors. How naturally occurring splicing isoforms of RON, especially those which are constitutively activated, affect tumorigenesis and therapeutic response, is largely unknown. Here, we identified that presence of activated RON could be a possible factor for the development of resistance against anti-EGFR (cetuximab) therapy in colorectal cancer patient tissues. Also, we elucidated the roles of three splicing variants of RON, RON Δ155, Δ160, and Δ165 as tumor drivers in cancer cell lines. Subsequently, we designed an inhibitor of RON, WM-S1-030, to suppress phosphorylation thereby inhibiting the activation of the three RON variants as well as the wild type. Specifically, WM-S1-030 treatment led to potent regression of tumor growth in solid tumors expressing the RON variants Δ155, Δ160, and Δ165. Two mechanisms for the RON oncogenic activity depending on KRAS genotype was evaluated in our study which include activation of EGFR and Src, in a trimeric complex, and stabilization of the beta-catenin. In terms of the immunotherapy, WM-S1-030 elicited notable antitumor immunity in anti-PD-1 resistant cell derived mouse model, likely via repression of M1/M2 polarization of macrophages. These findings suggest that WM-S1-030 could be developed as a new treatment option for cancer patients expressing these three RON variants.
Collapse
Affiliation(s)
- Joseph Kim
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Pharmacology, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-In Koh
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Minki Lee
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Yoon Sun Park
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
- Department of Pharmacology, AMIST, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Jae-Sik Shin
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Mi So Lee
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Min-Hwa Kim
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | | | | | | | - Jun Ki Hong
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | | | - Yea Seong Ryu
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
- Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea
| | - Seung-Mi Kim
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Mingee Choi
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Hyojin Kim
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Hyun Ryu
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Sun-Chul Hur
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Junho Park
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea
| | - Dae Young Hur
- Department of Anatomy and Tumor Immunology, Inje University College of Medicine, Busan, Republic of Korea
| | - Dong-Hoon Jin
- Wellmarkerbio Co., Ltd., Seoul, Republic of Korea.
- Department of Convergence Medicine, Asan Institute for Life Science, Asan Medical Center, Seoul, Republic of Korea.
- Department of Pharmacology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
4
|
Safaroghli-Azar A, Emadi F, Lenjisa J, Mekonnen L, Wang S. Kinase inhibitors: Opportunities for small molecule anticancer immunotherapies. Drug Discov Today 2023; 28:103525. [PMID: 36907320 DOI: 10.1016/j.drudis.2023.103525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/02/2023] [Accepted: 02/07/2023] [Indexed: 03/12/2023]
Abstract
As the fifth pillar of cancer treatment, immunotherapy has dramatically changed the paradigm of therapeutic strategies by focusing on the host's immune system. In the long road of immunotherapy development, the identification of immune-modulatory effects for kinase inhibitors opened a new chapter in this therapeutic approach. These small molecule inhibitors not only directly eradicate tumors by targeting essential proteins of cell survival and proliferation but can also drive immune responses against malignant cells. This review summarizes the current standings and challenges of kinase inhibitors in immunotherapy, either as a single agent or in a combined modality.
Collapse
Affiliation(s)
- Ava Safaroghli-Azar
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Fatemeh Emadi
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Jimma Lenjisa
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Laychiluh Mekonnen
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia
| | - Shudong Wang
- Drug Discovery and Development, University of South Australia, UniSA Clinical and Health Sciences, SA 5000, Australia.
| |
Collapse
|
5
|
Hunt BG, Fox LH, Davis JC, Jones A, Lu Z, Waltz SE. An Introduction and Overview of RON Receptor Tyrosine Kinase Signaling. Genes (Basel) 2023; 14:517. [PMID: 36833444 PMCID: PMC9956929 DOI: 10.3390/genes14020517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
RON is a receptor tyrosine kinase (RTK) of the MET receptor family that is canonically involved in mediating growth and inflammatory signaling. RON is expressed at low levels in a variety of tissues, but its overexpression and activation have been associated with malignancies in multiple tissue types and worse patient outcomes. RON and its ligand HGFL demonstrate cross-talk with other growth receptors and, consequentially, positions RON at the intersection of numerous tumorigenic signaling programs. For this reason, RON is an attractive therapeutic target in cancer research. A better understanding of homeostatic and oncogenic RON activity serves to enhance clinical insights in treating RON-expressing cancers.
Collapse
Affiliation(s)
- Brian G. Hunt
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Levi H. Fox
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - James C. Davis
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Angelle Jones
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | - Zhixin Lu
- 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 Hospital Medical Center, Cincinnati, OH 45220, USA
| |
Collapse
|
6
|
Attalla S, Taifour T, Muller W. Tailoring therapies to counter the divergent immune landscapes of breast cancer. Front Cell Dev Biol 2023; 11:1111796. [PMID: 36910138 PMCID: PMC9992199 DOI: 10.3389/fcell.2023.1111796] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/25/2023] [Indexed: 02/24/2023] Open
Abstract
Breast cancer remains a significant clinical concern affecting millions of women worldwide. Immunotherapy is a rapidly growing drug class that has revolutionized cancer treatment but remains marginally successful in breast cancer. The success of immunotherapy is dependent on the baseline immune responses as well as removing the brakes off pre-existing anti-tumor immunity. In this review, we summarize the different types of immune microenvironment observed in breast cancer as well as provide approaches to target these different immune subtypes. Such approaches have demonstrated pre-clinical success and are currently under clinical evaluation. The impact of combination of these approaches with already approved chemotherapies and immunotherapies may improve patient outcome and survival.
Collapse
Affiliation(s)
- Sherif Attalla
- Department Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tarek Taifour
- Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Department Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - William Muller
- Department Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Goodman Cancer Institute, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.,Department Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
Conformation specific antagonistic high affinity antibodies to the RON receptor kinase for imaging and therapy. Sci Rep 2022; 12:22564. [PMID: 36581692 PMCID: PMC9800565 DOI: 10.1038/s41598-022-26404-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022] Open
Abstract
The RON receptor tyrosine kinase is an exceptionally interesting target in oncology and immunology. It is not only overexpressed in a wide variety of tumors but also has been shown to be expressed on myeloid cells associated with tumor infiltration, where it serves to dampen tumour immune responses and reduce the efficacy of anti-CTLA4 therapy. Potent and selective inhibitory antibodies to RON might therefore both inhibit tumor cell growth and stimulate immune rejection of tumors. We derived cloned and sequenced a new panel of exceptionally avid anti-RON antibodies with picomolar binding affinities that inhibit MSP-induced RON signaling and show remarkable potency in antibody dependent cellular cytotoxicity. Antibody specificity was validated by cloning the antibody genes and creating recombinant antibodies and by the use of RON knock out cell lines. When radiolabeled with 89-Zirconium, the new antibodies 3F8 and 10G1 allow effective immuno-positron emission tomography (immunoPET) imaging of RON-expressing tumors and recognize universally exposed RON epitopes at the cell surface. The 10G1 was further developed into a novel bispecific T cell engager with a 15 pM EC50 in cytotoxic T cell killing assays.
Collapse
|
8
|
Hoover AR, Liu K, DeVette CI, Krawic JR, Medcalf AD, West CL, Hode T, Lam SSK, Welm AL, Sun XH, Hildebrand WH, Chen WR. Single-cell RNA sequencing reveals localized tumour ablation and intratumoural immunostimulant delivery potentiate T cell mediated tumour killing. Clin Transl Med 2022; 12:e937. [PMID: 35808806 PMCID: PMC9270578 DOI: 10.1002/ctm2.937] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/27/2022] [Accepted: 06/05/2022] [Indexed: 12/16/2022] Open
Abstract
Background Metastatic breast cancer poses great challenge in cancer treatment. N‐dihydrogalactochitosan (GC) is a novel immunoadjuvant that stimulates systemic immune responses when administered intratumourally following local tumour ablation. A combination of photothermal therapy (PTT) and GC, referred to as localized ablative immunotherapy (LAIT), extended animal survival and generates an activated B cell phenotype in MMTV‐PyMT mouse mammary tumour microenvironment (TME). However, how T cell populations respond to LAIT remains to be elucidated. Methods Using depletion antibodies, we studied the contributions of CD8+ and CD4+ T cells to the therapeutic effect of LAIT. Using single‐cell RNA‐sequencing (scRNAseq), we analysed tumour‐infiltrating T cell heterogeneity and dissected their transcriptomes upon treatments of PTT, GC, and LAIT (PTT+GC). Results Loss of CD8+ T cells after LAIT abrogated the therapeutic benefits of LAIT. Ten days after treatment, proportions of CD8+ and CD4+ T cells in untreated TME were 19.2% and 23.0%, respectively. Upon LAIT, both proportions were increased to 25.5% and 36.2%, respectively. In particular, LAIT increased the proportions of naïve and memory cells from a resting state to an activated state. LAIT consistently induced the expression of co‐stimulatory molecules, type I IFN responsive genes, and a series of antitumor cytokines, Ifng, Tnf, Il1, and Il17 in CD8+ and CD4+ T cells. LAIT also induced immune checkpoints Pdcd1, Ctla4, and Lag3 expression, consistent with T cell activation. Relevant to clinical translation, LAIT also upregulated genes in CD8+ and CD4+ T cells that positively correlated with extended survival of breast cancer patients. Conclusions Overall, our results reveal that LAIT prompts immunological remodelling of T cells by inducing broad proinflammatory responses and inhibiting suppressive signalling to drive antitumour immunity.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA.,Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| | - Christa I DeVette
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jason R Krawic
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Alexandra D Medcalf
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Connor L West
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| | - Tomas Hode
- Immunophotonics Inc., St. Louis, Missouri, USA
| | | | - Alana L Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Xiao-Hong Sun
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - William H Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
9
|
Xiao M, He J, Yin L, Chen X, Zu X, Shen Y. Tumor-Associated Macrophages: Critical Players in Drug Resistance of Breast Cancer. Front Immunol 2022; 12:799428. [PMID: 34992609 PMCID: PMC8724912 DOI: 10.3389/fimmu.2021.799428] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Drug resistance is one of the most critical challenges in breast cancer (BC) treatment. The occurrence and development of drug resistance are closely related to the tumor immune microenvironment (TIME). Tumor-associated macrophages (TAMs), the most important immune cells in TIME, are essential for drug resistance in BC treatment. In this article, we summarize the effects of TAMs on the resistance of various drugs in endocrine therapy, chemotherapy, targeted therapy, and immunotherapy, and their underlying mechanisms. Based on the current overview of the key role of TAMs in drug resistance, we discuss the potential possibility for targeting TAMs to reduce drug resistance in BC treatment, By inhibiting the recruitment of TAMs, depleting the number of TAMs, regulating the polarization of TAMs and enhancing the phagocytosis of TAMs. Evidences in our review support it is important to develop novel therapeutic strategies to target TAMs in BC to overcome the treatment of resistance.
Collapse
Affiliation(s)
- Maoyu Xiao
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguan Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
10
|
Lai SCA, Gundlapalli H, Ekiz HA, Jiang A, Fernandez E, Welm AL. Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression. Cancer Discov 2021; 11:3178-3197. [PMID: 34330779 PMCID: PMC8800951 DOI: 10.1158/2159-8290.cd-20-1172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/26/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapy has potential to prevent and treat metastatic breast cancer, but strategies to enhance immune-mediated killing of metastatic tumors are urgently needed. We report that a ligand-independent isoform of Ron kinase (SF-Ron) is a key target to enhance immune infiltration and eradicate metastatic tumors. Host-specific deletion of SF-Ron caused recruitment of lymphocytes to micrometastases, augmented tumor-specific T-cell responses, and nearly eliminated breast cancer metastasis in mice. Lack of host SF-Ron caused stem-like TCF1+ CD4+ T cells with type I differentiation potential to accumulate in metastases and prevent metastatic outgrowth. There was a corresponding increase in tumor-specific CD8+ T cells, which were also required to eliminate lung metastases. Treatment of mice with a Ron kinase inhibitor increased tumor-specific CD8+ T cells and protected from metastatic outgrowth. These data provide a strong preclinical rationale to pursue small-molecule Ron kinase inhibitors for the prevention and treatment of metastatic breast cancer. SIGNIFICANCE The discovery that SF-Ron promotes antitumor immune responses has significant clinical implications. Therapeutic antibodies targeting full-length Ron may not be effective for immunotherapy; poor efficacy of such antibodies in trials may be due to their inability to block SF-Ron. Our data warrant trials with inhibitors targeting SF-Ron in combination with immunotherapy. This article is highlighted in the In This Issue feature, p. 2945.
Collapse
Affiliation(s)
- Shu-Chin Alicia Lai
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Harika Gundlapalli
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - H. Atakan Ekiz
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Amanda Jiang
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Elvelyn Fernandez
- Genomics Summer Research for Minorities (GSRM) Program, University of Utah, Salt Lake City, Utah
| | - Alana L. Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
11
|
Li L, Wang J, Radford DC, Kopeček J, Yang J. Combination treatment with immunogenic and anti-PD-L1 polymer-drug conjugates of advanced tumors in a transgenic MMTV-PyMT mouse model of breast cancer. J Control Release 2021; 332:652-659. [PMID: 33607175 DOI: 10.1016/j.jconrel.2021.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 01/27/2023]
Abstract
Immune checkpoint blockade has revolutionized the treatment of tumors with immunogenic microenvironments. However, low response rate and acquired resistance are still major challenges. Herein we used a more clinically relevant model of transgenic MMTV-PyMT tumor that more closely mimics the development of human breast cancer in an immunocompetent background to investigate a polymer-based chemo-immunotherapy. We have found that tumors acquired an increased degree of immune suppression during progression, rendering them unresponsive to anti-PD-L1 therapy. To treat large tumors at their advanced stage, we applied a combination strategy consisting of two polymer-drug conjugates that could induce immunogenic cell death (ICD) and disrupt the PD-L1/PD-1 interaction, respectively. Although ICD-inducing conjugate remodeled tumor immune microenvironment by facilitating significant CD8+ T cell infiltration, advanced tumor adapted the immune suppressive mechanism of elevating PD-L1 expression on both cancer cells and myeloid cells thereafter to enable continued tumor growth. Concurrent treatment of PD-L1 blocking conjugate not only abrogated the PD-L1 expression from the two disparate cellular sources, but also considerably reduced the number of immunosuppressive myeloid cells, thereby leading to a significant shrinkage of advanced tumors. Our data provide evidence that combinatory strategy of ICD-inducing and PD-L-blocking modalities could reverse immune suppression and establish a basis for the rational design of cancer immunotherapy.
Collapse
Affiliation(s)
- Lian Li
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiawei Wang
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - D Christopher Radford
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
12
|
Attalla S, Taifour T, Bui T, Muller W. Insights from transgenic mouse models of PyMT-induced breast cancer: recapitulating human breast cancer progression in vivo. Oncogene 2021; 40:475-491. [PMID: 33235291 PMCID: PMC7819848 DOI: 10.1038/s41388-020-01560-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/27/2020] [Accepted: 11/06/2020] [Indexed: 01/05/2023]
Abstract
Breast cancer is associated with the second highest cancer-associated deaths worldwide. Therefore, understanding the key events that determine breast cancer progression, modulation of the tumor-microenvironment and metastasis, which is the main cause of cancer-associated death, are of great importance. The mammary specific polyomavirus middle T antigen overexpression mouse model (MMTV-PyMT), first published in 1992, is the most commonly used genetically engineered mouse model (GEMM) for cancer research. Mammary lesions arising in MMTV-PyMT mice follow similar molecular and histological progression as human breast tumors, making it an invaluable tool for cancer researchers and instrumental in understanding tumor biology. In this review, we will highlight key studies that demonstrate the utility of PyMT derived GEMMs in understanding the molecular basis of breast cancer progression, metastasis and highlight its use as a pre-clinical tool for therapeutic discovery.
Collapse
Affiliation(s)
- Sherif Attalla
- Department of Biochemistry, McGill University, Montreal, QC, H3A 1A3, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Tarek Taifour
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
- Faculty of Medicine, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Tung Bui
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - William Muller
- Department of Biochemistry, McGill University, Montreal, QC, H3A 1A3, Canada.
- Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada.
- Faculty of Medicine, McGill University, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|
13
|
Chen Y, Jin H, Song Y, Huang T, Cao J, Tang Q, Zou Z. Targeting tumor-associated macrophages: A potential treatment for solid tumors. J Cell Physiol 2020; 236:3445-3465. [PMID: 33200401 DOI: 10.1002/jcp.30139] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/20/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAMs) in solid tumors exert protumor activities by releasing cytokines or growth factors into the tumor microenvironment. Increasing studies have also shown that TAMs play a key role in tumor progression, such as tumor angiogenesis, immunosuppression, cell proliferation, migration, invasion, and metastasis. A large body of evidence shows that the abundance of TAMs in solid tumors is correlated with poor disease prognosis and resistance to therapies. Therefore, targeting TAMs in solid tumors is considered to be a promising immunotherapeutic strategy. At present, the therapeutic strategies of targeting macrophages mainly include limiting monocyte recruitment, depletion strategies, promoting macrophage phagocytic activity, and induction of macrophage reprogramming. Additionally, targeting TAMs in combination with conventional therapies has been demonstrated to be a promising therapeutic strategy in solid tumors. In the present review, we summarized various TAMs-targeting therapeutic strategies for treating solid tumors. This review also discusses the challenges for targeting TAMs as tumor treatments, the obstacles in clinical trials, and the perspective for the future development of TAMs-targeting therapies for various cancers.
Collapse
Affiliation(s)
- Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Yucen Song
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ting Huang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jun Cao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Qing Tang
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhengzhi Zou
- MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
14
|
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: 22] [Impact Index Per Article: 5.5] [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
|
15
|
Gonda TA, Fang J, Salas M, Do C, Hsu E, Zhukovskaya A, Siegel A, Takahashi R, Lopez-Bujanda ZA, Drake CG, Manji GA, Wang TC, Olive KP, Tycko B. A DNA Hypomethylating Drug Alters the Tumor Microenvironment and Improves the Effectiveness of Immune Checkpoint Inhibitors in a Mouse Model of Pancreatic Cancer. Cancer Res 2020; 80:4754-4767. [PMID: 32816859 DOI: 10.1158/0008-5472.can-20-0285] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 06/26/2020] [Accepted: 07/30/2020] [Indexed: 11/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer that has proven refractory to immunotherapy. Previously, treatment with the DNA hypomethylating drug decitabine (5-aza-dC; DAC) extended survival in the KPC-Brca1 mouse model of PDAC. Here we investigated the effects of DAC in the original KPC model and tested combination therapy with DAC followed by immune checkpoint inhibitors (ICI). Four protocols were tested: PBS vehicle, DAC, ICI (anti-PD-1 or anti-VISTA), and DAC followed by ICI. For each single-agent and combination treatment, tumor growth was measured by serial ultrasound, tumor-infiltrating lymphoid and myeloid cells were characterized, and overall survival was assessed. Single-agent DAC led to increased CD4+ and CD8+ tumor-infiltrating lymphocytes (TIL), PD1 expression, and tumor necrosis while slowing tumor growth and modestly increasing mouse survival without systemic toxicity. RNA-sequencing of DAC-treated tumors revealed increased expression of Chi3l3 (Ym1), reflecting an increase in a subset of tumor-infiltrating M2-polarized macrophages. While ICI alone had modest effects, DAC followed by either of ICI therapies additively inhibited tumor growth and prolonged mouse survival. The best results were obtained using DAC followed by anti-PD-1, which extended mean survival from 26 to 54 days (P < 0.0001). In summary, low-dose DAC inhibits tumor growth and increases both TILs and a subset of tumor-infiltrating M2-polarized macrophages in the KPC model of PDAC, and DAC followed by anti-PD-1 substantially prolongs survival. Because M2-polarized macrophages are predicted to antagonize antitumor effects, targeting these cells may be important to enhance the efficacy of combination therapy with DAC plus ICI. SIGNIFICANCE: In a pancreatic cancer model, a DNA hypomethylating drug increases tumor-infiltrating effector T cells, increases a subset of M2 macrophages, and significantly prolongs survival in combination with immune checkpoint inhibitors.See related commentary by Nephew, p. 4610.
Collapse
Affiliation(s)
- Tamas A Gonda
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York. .,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Division of Gastroenterology and Hepatology, Department of Medicine, New York University, New York, New York
| | - Jarwei Fang
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York
| | - Martha Salas
- Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, New Jersey
| | - Catherine Do
- Division of Genetics & Epigenetics, Hackensack-Meridian Health Center for Discovery and Innovation, Nutley, New Jersey
| | - Emily Hsu
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Anna Zhukovskaya
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York
| | - Ariel Siegel
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York
| | - Ryota Takahashi
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Zoila A Lopez-Bujanda
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York.,Graduate Program in Pathobiology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles G Drake
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Gulam A Manji
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Timothy C Wang
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Kenneth P Olive
- Department of Medicine, Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Benjamin Tycko
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University, New York, New York. .,John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, New Jersey.,Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C
| |
Collapse
|
16
|
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: 10] [Impact Index Per Article: 2.5] [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
|
17
|
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: 0.8] [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
|
18
|
Lenting K, van den Heuvel CNAM, van Ewijk A, ElMelik D, de Boer R, Tindall E, Wei G, Kusters B, te Dorsthorst M, ter Laan M, Huynen MA, Leenders WP. Mapping actionable pathways and mutations in brain tumours using targeted RNA next generation sequencing. Acta Neuropathol Commun 2019; 7:185. [PMID: 31747973 PMCID: PMC6865071 DOI: 10.1186/s40478-019-0826-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/02/2019] [Indexed: 01/28/2023] Open
Abstract
Many biology-based precision drugs are available that neutralize aberrant molecular pathways in cancer. Molecular heterogeneity and the lack of reliable companion diagnostic biomarkers for many drugs makes targeted treatment of cancer inaccurate for many individuals. Identifying actionable hyperactive biological pathways in individual cancers may improve this situation. To achieve this we applied a novel targeted RNA next generation sequencing (t/RNA-NGS) technique to surgically obtained glioma tissues. The test combines mutation detection with analysis of biological pathway activities that are involved in tumour behavior in many cancer types (e.g. tyrosine kinase signaling, angiogenesis signaling, immune response, metabolism), via quantitative measurement of transcript levels and splice variants of hundreds of genes. We here present proof of concept that the technique, which uses molecular inversion probes, generates a histology-independent molecular diagnosis and identifies classifiers that are strongly associated with conventional histopathology diagnoses and even with patient prognosis. The test not only confirmed known glioma-associated molecular aberrations but also identified aberrant expression levels of actionable genes and mutations that have so far been considered not to be associated with glioma, opening up the possibility of drug repurposing for individual patients. Its cost-effectiveness makes t/RNA-NGS to an attractive instrument to aid oncologists in therapy decision making.
Collapse
|
19
|
Kim SA, Lee KH, Lee DH, Lee JK, Lim SC, Joo YE, Chung IJ, Noh MG, Yoon TM. Receptor tyrosine kinase, RON, promotes tumor progression by regulating EMT and the MAPK signaling pathway in human oral squamous cell carcinoma. Int J Oncol 2019; 55:513-526. [PMID: 31268163 DOI: 10.3892/ijo.2019.4836] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/18/2019] [Indexed: 11/05/2022] Open
Abstract
The receptor tyrosine kinase, recepteur d'origine nantais (RON), is known to be associated with the progression, metastasis, and prognosis of various types of cancers. Nevertheless, the role of RON in human oral squamous cell carcinoma (OSCC) is unclear. This study evaluated whether RON affects oncogenic behavior, oncogenic signaling pathways, and clinical outcomes, including survival, in human OSCC. Reverse transcription‑PCR, quantitative PCR, western blotting and immunohistochemical staining were used to determine mRNA and protein expression levels of RON. Cell invasion, migration and apoptosis assays were used to assess the functional effects of small interfering RNA‑mediated knockdown of RON or snail family transcriptional repressor 2 (SLUG). RON knockdown suppressed tumor cell invasion and migration and enhanced apoptosis in human OSCC cells. RON knockdown also decreased the phosphorylation of MAPK signaling proteins, such as ERK1/2, JNK and p38. In addition, RON knockdown suppressed the expression of the epithelial mesenchymal transition (EMT)‑related transcription factor, SLUG. SLUG knockdown blocked the enhancement of cell invasion and migration induced by macrophage‑stimulation protein (MSP)‑mediated RON activation in OSCC cells. The cell morphology was changed to spindle‑like shape under MSP‑mediated RON activation in OSCC cells. RON was overexpressed in both fresh and paraffin‑embedded human OSCC tissues. Taken together, these results indicate that RON contributed to tumor progression by regulating the EMT‑related factor, SLUG, and the MAPK pathway in OSCC. This study may provide a theoretical basis for the application of RON‑targeting agents, currently being studied in various cancer fields, for the treatment of OSCC.
Collapse
Affiliation(s)
- Sun-Ae Kim
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Dong Hoon Lee
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Joon Kyoo Lee
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Sang Chul Lim
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Ik-Joo Chung
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Myung-Giun Noh
- Department of Genomic Medicine, Gwangju Institute of Science and Technology, Gwangju, Gwangju 61005, Republic of Korea
| | - Tae Mi Yoon
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| |
Collapse
|
20
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
21
|
Yao HP, Feng L, Suthe SR, Chen LH, Weng TH, Hu CY, Jun ES, Wu ZG, Wang WL, Kim SC, Tong XM, Wang MH. Therapeutic efficacy, pharmacokinetic profiles, and toxicological activities of humanized antibody-drug conjugate Zt/g4-MMAE targeting RON receptor tyrosine kinase for cancer therapy. J Immunother Cancer 2019; 7:75. [PMID: 30871619 PMCID: PMC6419354 DOI: 10.1186/s40425-019-0525-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/31/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Aberrant expression of the RON receptor tyrosine kinase is a pathogenic feature and a validated drug target in various types of cancers. Currently, therapeutic antibodies targeting RON for cancer therapy are under intensive evaluation. Here we report the development and validation of a novel humanized anti-RON antibody-drug conjugate for cancer therapy. METHODS Antibody humanization was achieved by grafting sequences of complementarity-determining regions from mouse monoclonal antibody Zt/g4 into human IgG1/κ acceptor frameworks. The selected humanized Zt/g4 subclone H1L3 was conjugated with monomethyl auristatin E using a dipeptide linker to form H-Zt/g4-MMAE. Pharmacokinetic analysis of H-Zt/g4-MMAE was determined using hydrophobic interaction chromatography and a MMAE ADC ELISA kit. Biochemical and biological assays were used for measuring RON expression, internalization, cell viability and death. Therapeutic efficacies of H-Zt/g4-MMAE were validated in vivo using three pancreatic cancer xenograft models. Toxicological activities of H-Zt/g4-MMAE were determined in mouse and cynomolgus monkey. RESULTS H-Zt/g4-MMAE had a drug to antibody ratio of 3.77:1 and was highly stable in human plasma with a dissociation rate less than 5% within a 20 day period. H-Zt/g4-MMAE displayed a favorable pharmacokinetic profile in both mouse and cynomolgus monkey. In vitro, H-Zt/g4-MMAE induced RON internalization, which results in killing of pancreatic cancer cells with IC50 values at 10-20 nM. In vivo, H-Zt/g4-MMAE inhibited pancreatic cancer xenograft growth with tumoristatic concentrations at 1~3 mg/kg bodyweight. Significantly, H-Zt/g4-MMAE eradicated tumors across multiple xenograft models regardless their chemoresistant and metastatic statuses. Moreover, H-Zt/g4-MMAE inhibited and eradicated xenografts mediated by pancreatic cancer stem-like cells and by primary cells from patient-derived tumors. Toxicologically, H-Zt/g4-MMAE is well tolerated in mice up to 60 mg/kg. In cynomolgus monkey, H-Zt/g4-MMAE up to 30 mg/kg had a manageable and reversible toxicity profile. CONCLUSIONS H-Zt/g4-MMAE is superior in eradication of pancreatic cancer xenografts with favorable pharmacokinetic profiles and manageable toxicological activities. These findings warrant the transition of H-Zt/g4-MMAE into clinical trials in the future.
Collapse
Affiliation(s)
- Hang-Ping Yao
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Feng
- Cancer Biology Research Center, Hangzhou, China.,Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, USA
| | - Sreedhar Reddy Suthe
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, USA
| | - Ling-Hui Chen
- Zhejiang Provincial Key Laboratory for Precision Diagnosis & Treatment of Hepatic & Pancreatic Oncology, Zhejiang Province, China.,Division of Hepatobiliary & Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Hao Weng
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen-Yu Hu
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Eun Sung Jun
- Department of Biomedical Sciences, Kowloon Tong, Hong Kong
| | - Zhi-Gang Wu
- Zhejiang Provincial Key Laboratory for Precision Diagnosis & Treatment of Hepatic & Pancreatic Oncology, Zhejiang Province, China
| | - Wei-Lin Wang
- Zhejiang Provincial Key Laboratory for Precision Diagnosis & Treatment of Hepatic & Pancreatic Oncology, Zhejiang Province, China. .,Division of Hepatobiliary & Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Song Cheol Kim
- Biliary and Pancreatic Cancer Center at Department of Surgery, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Xiang-Min Tong
- Department of Laboratory Medicine, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China.
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis & Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center School of Pharmacy, Amarillo, TX, USA. .,Zhejiang Provincial Key Laboratory for Precision Diagnosis & Treatment of Hepatic & Pancreatic Oncology, Zhejiang Province, China.
| |
Collapse
|
22
|
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
|