1
|
Cheng TC, Hung MC, Wang LH, Tu SH, Wu CH, Yen Y, Chen CL, Whang-Peng J, Lee WJ, Liao YC, Lee YC, Pan MH, Lin HK, Tzeng HE, Guo P, Chu CY, Chen LC, Ho YS. Histamine N-methyltransferase (HNMT) as a potential auxiliary biomarker for predicting adaptability to anti-HER2 drug treatment in breast cancer patients. Biomark Res 2025; 13:7. [PMID: 39789599 PMCID: PMC11720525 DOI: 10.1186/s40364-024-00715-5] [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: 04/21/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Up to 23% of breast cancer patients recurred within a decade after trastuzumab treatment. Conversely, one trial found that patients with low HER2 expression and metastatic breast cancer had a positive response to trastuzumab-deruxtecan (T-Dxd). This indicates that relying solely on HER2 as a single diagnostic marker to predict the efficacy of anti-HER2 drugs is insufficient. This study highlights the interaction between histamine N-methyltransferase (HNMT) and HER2 as an adjunct predictor for trastuzumab response. Furthermore, modulation of HER2 expression by HNMT may explain why those with low HER2 expression still respond to T-Dxd. METHODS We investigated the impact of HNMT protein expression on the efficacy of anti-HER2 therapy in both in vivo and ex vivo models of patient-derived xenografts and cell line-derived xenografts. Our analysis included Förster resonance energy transfer (FRET) to assess the interaction strength between HNMT and HER2 proteins in trastuzumab-resistant and sensitive tumor tissues. Additionally, we used fluorescence lifetime imaging microscopy (FLIM), cleaved luciferase, and immunoprecipitation to study the interaction dynamics of HNMT and HER2. Furthermore, we evaluated the influence of HNMT activity on the binding of anti-HER2 antibodies to their targets through flow cytometry. We also observed the nuclear translocation of HNMT/HER2-ICD cells using fluorescent double staining and DeltaVision microscopy. Finally, ChIP sequencing was employed to identify target genes affected by the HNMT/HER2-ICD complex. RESULTS This study highlights HNMT as a potential auxiliary biomarker for diagnosing HER2 + breast cancer. FRET analysis demonstrated a significant interaction between HNMT and HER2 protein in trastuzumab-sensitive tumor tissue (n = 50), suggesting the potential of HNMT as a predictor of treatment response. Mechanistic studies revealed that the interaction between HNMT and HER2 contributes to increased HER2 protein expression at the transcriptional level, thereby impacting the efficacy of anti-HER2 therapy. Furthermore, a subset of triple-negative breast cancers characterized by HNMT overexpression was found to be sensitive to HER2 antibody-drug conjugates such as T-Dxd. CONCLUSIONS These findings offer crucial insights for clinicians evaluating candidates for anti-HER2 therapy, especially for HER2-low breast cancer patients who could gain from T-Dxd treatment. Identifying HNMT expression could help clinicians pinpoint patients who would benefit from anti-HER2 therapy.
Collapse
Affiliation(s)
- Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Lu-Hai Wang
- Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Shih-Hsin Tu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University; & Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chih-Hsiung Wu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University; & Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jacqueline Whang-Peng
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Jui Lee
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan
| | - You-Cheng Liao
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ching Lee
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Sciences and Technology, National Taiwan University, Taipei, Taiwan
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Huey-En Tzeng
- Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung-Hsing University, Taichung, Taiwan
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, and James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Cheng-Ying Chu
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- CRISPR Gene Targeting Core, Taipei Medical University, Taipei, 110, Taiwan
| | - Li-Ching Chen
- Department of Biological Science & Technology, College of Life Sciences, China Medical University, Taichung, Taiwan.
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
2
|
Gurrea-Rubio M, Lin F, Wicha MS, Mao-Draayer Y, Fox DA. Ligands of CD6: roles in the pathogenesis and treatment of cancer. Front Immunol 2025; 15:1528478. [PMID: 39840036 PMCID: PMC11747410 DOI: 10.3389/fimmu.2024.1528478] [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: 11/14/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
Cluster of Differentiation 6 (CD6), an established marker of T cells, has multiple and complex functions in regulation of T cell activation and proliferation, and in adhesion of T cells to antigen-presenting cells and epithelial cells in various organs and tissues. Early studies on CD6 demonstrated its role in mediating cell-cell interactions through its first ligand to be identified, CD166/ALCAM. The observation of CD6-dependent functions of T cells that could not be explained by interactions with CD166/ALCAM led to discovery of a second ligand, CD318/CDCP1. An additional cell surface molecule (CD44) is being studied as a potential third ligand of CD6. CD166, CD318, and CD44 are widely expressed by both differentiated cancer cells and cancer stem-like cells, and the level of their expression generally correlates with poor prognosis and increased metastatic potential. Therefore, there has been an increased focus on understanding how CD6 interacts with its ligands in the context of cancer biology and cancer immunotherapy. In this review, we assess the roles of these CD6 ligands in both the pathogenesis and treatment of cancer.
Collapse
Affiliation(s)
- Mikel Gurrea-Rubio
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Feng Lin
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Max S. Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Multiple Sclerosis Center of Excellence, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - David A. Fox
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
3
|
Yang M, Li Y, Kong L, Huang S, He L, Liu P, Mo S, Lu X, Lin X, Xiao Y, Shi D, Huang X, Chen B, Chen X, Ouyang Y, Li J, Lin C, Song L. Inhibition of DPAGT1 suppresses HER2 shedding and trastuzumab resistance in human breast cancer. J Clin Invest 2023; 133:e164428. [PMID: 37463446 DOI: 10.1172/jci164428] [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: 08/12/2022] [Accepted: 05/23/2023] [Indexed: 07/20/2023] Open
Abstract
Human epidermal growth factor receptor 2-targeted (HER2-targeted) therapy is the mainstay of treatment for HER2+ breast cancer. However, the proteolytic cleavage of HER2, or HER2 shedding, induces the release of the target epitope at the ectodomain (ECD) and the generation of a constitutively active intracellular fragment (p95HER2), impeding the effectiveness of anti-HER2 therapy. Therefore, identifying key regulators in HER2 shedding might provide promising targetable vulnerabilities against resistance. In the current study, we found that upregulation of dolichyl-phosphate N-acetylglucosaminyltransferase (DPAGT1) sustained high-level HER2 shedding to confer trastuzumab resistance, which was associated with poor clinical outcomes. Upon trastuzumab treatment, the membrane-bound DPAGT1 protein was endocytosed via the caveolae pathway and retrogradely transported to the ER, where DPAGT1 induced N-glycosylation of the sheddase - ADAM metallopeptidase domain 10 (ADAM10) - to ensure its expression, maturation, and activation. N-glycosylation of ADAM10 at N267 protected itself from ER-associated protein degradation and was essential for DPAGT1-mediated HER2 shedding and trastuzumab resistance. Importantly, inhibition of DPAGT1 with tunicamycin acted synergistically with trastuzumab treatment to block HER2 signaling and reverse resistance. These findings reveal a prominent mechanism for HER2 shedding and suggest that targeting DPAGT1 might be a promising strategy against trastuzumab-resistant breast cancer.
Collapse
Affiliation(s)
- Muwen Yang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Yue Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Lingzhi Kong
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong China
| | - Lixin He
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Mo
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong China
| | - Xiuqing Lu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Xi Lin
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Yunyun Xiao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Xinjian Huang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Boyu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Ying Ouyang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Jun Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong China
| | - Chuyong Lin
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
| | - Libing Song
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine and
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Qi X, Li Z, Zhang J, Li H, Zhang G, Li M, Li B, Fu Y, Cai M, Wang H, Tong T, Gao J. Mechanistic insights into CDCP1 clustering on non-small-cell lung cancer membranes revealed by super-resolution fluorescent imaging. iScience 2023; 26:106103. [PMID: 36866248 PMCID: PMC9972570 DOI: 10.1016/j.isci.2023.106103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/30/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
CDCP1 is a transmembrane protein that is involved in a variety of important biological processes and upregulated in a variety of human solid malignancies; however, its spatial distribution and variation at the molecular level remain unclear. To solve this problem, we first analyzed its expression level and prognostic implications in lung cancer. Then, we used super-resolution microscopy to reveal the spatial organization of CDCP1 at different levels, and found that cancer cells generated more and larger CDCP1 clusters than normal cells. Furthermore, we found that CDCP1 can be integrated into larger and denser clusters as functional domains upon activation. Our findings elucidated the significant differences of CDCP1 clustering characteristics between cancer and normal cells, and revealed the relationship between its distribution and function, which will contribute to a comprehensive understanding of its oncogenic mechanism, and will be of great help for the development of CDCP1-targeted drugs for lung cancer.
Collapse
Affiliation(s)
- Xiao Qi
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Zihao Li
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Jinrui Zhang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongru Li
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Guangxin Zhang
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Meng Li
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Baofeng Li
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Yilin Fu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
- Laboratory for Marine Biology and Biotechnology, Qing dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong 266237, China
| | - Ti Tong
- Department of Thoracic Surgery, the Second Hospital of Jilin University, Changchun, Jilin 130041, China
- Corresponding author
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- Corresponding author
| |
Collapse
|
5
|
Bartkowiak K, Mossahebi Mohammadi P, Gärtner S, Kwiatkowski M, Andreas A, Geffken M, Peine S, Verpoort K, Scholz U, Deutsch TM, Michel LL, Schneeweiss A, Thewes V, Trumpp A, Müller V, Riethdorf S, Schlüter H, Pantel K. Detection and Isolation of Circulating Tumor Cells from Breast Cancer Patients Using CUB Domain-Containing Protein 1. J Proteome Res 2023; 22:1213-1230. [PMID: 36926972 DOI: 10.1021/acs.jproteome.2c00739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
In cancer metastasis, single circulating tumor cells (CTCs) in the blood and disseminated tumor cells (DTCs) in the bone marrow mediate cancer metastasis. Because suitable biomarker proteins are lacking, CTCs and DTCs with mesenchymal attributes are difficult to isolate from the bulk of normal blood cells. To establish a procedure allowing the isolation of such cells, we analyzed the cell line BC-M1 established from DTCs in the bone marrow of a breast cancer patient by stable isotope labeling by amino acids in cell culture (SILAC) and mass spectrometry. We found high levels of the transmembrane protein CUB domain-containing protein 1 (CDCP1) in breast cancer cell lines with mesenchymal attributes. Peripheral blood mononuclear cells were virtually negative for CDCP1. Confirmation in vivo by CellSearch revealed CDCP1-positive CTCs in 8 of 30 analyzed breast cancer patients. Only EpCam-positive CTCs were enriched by CellSearch. Using the extracellular domain of CDCP1, we established a magnetic-activated cell sorting (MACS) approach enabling also the enrichment of EpCam-negative CTCs. Thus, our approach is particularly suited for the isolation of mesenchymal CTCs with downregulated epithelial cancer that occur, for example, in triple-negative breast cancer patients who are prone to therapy failure.
Collapse
Affiliation(s)
- Kai Bartkowiak
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Parinaz Mossahebi Mohammadi
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Sebastian Gärtner
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Marcel Kwiatkowski
- Laboratory for Metabolic Signaling, Institute of Biochemistry, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Antje Andreas
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Maria Geffken
- Department of Transfusion Medicine, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Sven Peine
- Department of Transfusion Medicine, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Karl Verpoort
- Practice for Haematology and Oncology, Hohe Weide 17b, 20295 Hamburg, Germany
| | - Ursula Scholz
- Gynecological Oncology, Asklepios Klinik Hamburg-Barmbek, Rübenkamp 220, 22307 Hamburg, Germany
| | - Thomas M Deutsch
- Department of Obstetrics and Gynecology, University of Heidelberg, Im Neuenheimer Feld 440, 69120 Heidelberg, Germany
| | - Laura L Michel
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Verena Thewes
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Volkmar Müller
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Sabine Riethdorf
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Department of Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Klaus Pantel
- Department of Tumour Biology, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| |
Collapse
|
6
|
Bonham C, Mandati V, Singh R, Pappin D, Tonks N. Coupling substrate-trapping with proximity-labeling to identify protein tyrosine phosphatase PTP1B signaling networks. J Biol Chem 2023; 299:104582. [PMID: 36871762 PMCID: PMC10148153 DOI: 10.1016/j.jbc.2023.104582] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
The ability to define functional interactions between enzymes and their substrates is crucial for understanding biological control mechanisms; however, such methods face challenges in the transient nature and low stoichiometry of enzyme-substrate interactions. Now, we have developed an optimized strategy that couples substrate-trapping mutagenesis to proximity-labeling mass spectrometry for quantitative analysis of protein complexes involving the protein tyrosine phosphatase PTP1B. This methodology represents a significant shift from classical schemes; it is capable of being performed at near-endogenous expression levels and increasing stoichiometry of target enrichment without a requirement for stimulation of supraphysiological tyrosine phosphorylation levels or maintenance of substrate complexes during lysis and enrichment procedures. Advantages of this new approach are illustrated through application to PTP1B interaction networks in models of HER2-positive and Herceptin-resistant breast cancer. We have demonstrated that inhibitors of PTP1B significantly reduced proliferation and viability in cell-based models of acquired and de novo Herceptin resistance in HER2-positive breast cancer. Using differential analysis, comparing substrate-trapping to wild-type PTP1B, we have identified multiple unreported protein targets of PTP1B with established links to HER2-induced signaling and provided internal validation of method specificity through overlap with previously identified substrate candidates. Overall, this versatile approach can be readily integrated with evolving proximity-labeling platforms (TurboID, BioID2, etc.), and is broadly applicable across all PTP family members for the identification of conditional substrate specificities and signaling nodes in models of human disease.
Collapse
Affiliation(s)
- ChristopherA Bonham
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - Vinay Mandati
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - RakeshK Singh
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - DarrylJ Pappin
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - NicholasK Tonks
- Cold Spring Harbor Laboratory, One Bungtown Road, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
7
|
CDCP1 expression is frequently increased in aggressive urothelial carcinoma and promotes urothelial tumor progression. Sci Rep 2023; 13:73. [PMID: 36593286 PMCID: PMC9807563 DOI: 10.1038/s41598-022-26579-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023] Open
Abstract
The prognosis of patients with advanced urothelial carcinoma (UC) remains poor and improving treatment continues to be a major medical need. CUB domain containing protein 1 (CDCP1) is a known oncogene in various types of solid cancers and its overexpression is associated with impaired prognosis. However, its role in UC remains undetermined. Here we assessed the clinical relevance of CDCP1 in two cohorts of UC at different stages of the disease. Immunohistochemistry showed that CDCP1 is highly expressed in advanced UC, which significantly correlates with shorter overall survival. Importantly, the basal/squamous UC subtype showed significantly enriched CDCP1 at the mRNA and protein levels. The functional role of CDCP1 overexpression was assessed taking advantage of ex vivo organoids derived from the CDCP1pcLSL/+ transgenic mouse model. Furthermore, CDCP1 knockout UC cell lines were generated using CRISPR/Cas9 technology. Interestingly, CDCP1 overexpression significantly induced the activation of MAPK/ERK pathways in ex vivo organoids and increased their proliferation. Similarly, CDCP1 knockout in UC cell lines reduced their proliferation and migration, concomitant with MAPK/ERK pathway activity reduction. Our results highlight the relevance of CDCP1 in advanced UC and demonstrate its oncogenic role, suggesting that targeting CDCP1 could be a rational therapeutic strategy for the treatment of advanced UC.
Collapse
|
8
|
Gurrea-Rubio M, Fox DA. The dual role of CD6 as a therapeutic target in cancer and autoimmune disease. Front Med (Lausanne) 2022; 9:1026521. [PMID: 36275816 PMCID: PMC9579686 DOI: 10.3389/fmed.2022.1026521] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Autoimmune disease involves loss of tolerance to self-antigen, while progression of cancer reflects insufficient recognition and response of the immune system to malignant cells. Patients with immune compromised conditions tend to be more susceptible to cancer development. On the other hand, cancer treatments, especially checkpoint inhibitor therapies, can induce severe autoimmune syndromes. There is recent evidence that autoimmunity and cancer share molecular targets and pathways that may be dysregulated in both types of diseases. Therefore, there has been an increased focus on understanding these biological pathways that link cancer and its treatment with the appearance of autoimmunity. In this review, we hope to consolidate our understanding of current and emerging molecular targets used to treat both cancer and autoimmunity, with a special focus on Cluster of Differentiation (CD) 6.
Collapse
|
9
|
Overexpression of CDCP1 is Associated with Poor Prognosis and Enhanced Immune Checkpoints Expressions in Breast Cancer. JOURNAL OF ONCOLOGY 2022; 2022:1469354. [PMID: 36090897 PMCID: PMC9452972 DOI: 10.1155/2022/1469354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022]
Abstract
CUB-domain containing protein 1 (CDCP1) is a transmembrane protein acting as an effector of SRC family kinases, which play an oncogenic role in multiple human cancers. However, its clinical and immune correlations in breast cancer (BrCa) have not been explored. To define the expression, prognostic value, and potential molecular role of CDCP1 in BrCa, multiple public datasets, and an in-house cohort were used. Compared with paratumor tissue, CDCP1 was remarkably upregulated in the tumor tissues at both mRNA and protein levels. In the in-house cohort, CDCP1 protein expression was related to several clinicopathological parameters, including age, ER status, PR status, molecular type, and survival status. Kaplan–Meier analysis and Cox regression analysis exhibited that CDCP1 was an important prognostic biomarker in BrCa. In addition, enrichment analysis uncovered that CDCP1 was not only involved in multiple oncogenic pathways, but correlated with overexpression of immune checkpoints. Overall, we reported that increased expression of CDCP1 is a favorable prognostic factor in patients with BrCa. In addition, the correlations between CDCP1 and immune checkpoints provide a novel insight into the adjuvant treatment for immune checkpoint blockade via targeting CDCP1.
Collapse
|
10
|
Murakami Y, Kusakabe D, Watari K, Kawahara A, Azuma K, Akiba J, Taniguchi M, Kuwano M, Ono M. AXL/CDCP1/SRC axis confers acquired resistance to osimertinib in lung cancer. Sci Rep 2022; 12:8983. [PMID: 35643725 PMCID: PMC9148303 DOI: 10.1038/s41598-022-12995-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 05/17/2022] [Indexed: 11/10/2022] Open
Abstract
Osimertinib, a third-generation EGFR-TKI, has nowadays been applied to non-small cell lung cancer harboring activated EGFR mutation with or without T790M, but ultimately develop resistance to this drug. Here we report a novel mechanism of acquired resistance to osimertinib and the reversal of which could improve the clinical outcomes. In osimertinib-resistant lung cancer cell lines harboring T790M mutation that we established, expression of multiple EGFR family proteins and MET was markedly reduced, whereas expression of AXL, CDCP1 and SRC was augmented along with activation of AKT. Surprisingly, AXL or CDCP1 expression was induced by osimertinib in a time-dependent manner up to 3 months. Silencing of CDCP1 or AXL restored the sensitivity to osimertinib with reduced activation of SRC and AKT. Furthermore, silencing of both CDCP1 and AXL increased the sensitivity to osimertinib. Either silencing of SRC or dasatinib, a SRC family kinase (SFK) inhibitor, suppressed AKT phosphorylation and cell growth. Increased expression of AXL and CDCP1 was observed in refractory tumor samples from patients with lung cancer treated with osimertinib. Together, this study suggests that AXL/SFK/AKT and CDCP1/SFK/AKT signaling pathways play some roles in acquired osimertinib resistance of non-small cell lung cancer.
Collapse
Affiliation(s)
- Yuichi Murakami
- Cancer Translational Research Center, St. Mary's Institute of Health Sciences, Kurume, Fukuoka, Japan.,Department of Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Kusakabe
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Watari
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Koichi Azuma
- Division of Respirology, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Jun Akiba
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka, Japan
| | | | - Michihiko Kuwano
- Cancer Translational Research Center, St. Mary's Institute of Health Sciences, Kurume, Fukuoka, Japan
| | - Mayumi Ono
- Cancer Translational Research Center, St. Mary's Institute of Health Sciences, Kurume, Fukuoka, Japan. .,Department of Pharmaceutical Oncology, Graduate School of Nursing, St. Mary's College, 422 Tsubukuhonmachi, Kurume, Fukuoka, 830-8558, Japan.
| |
Collapse
|
11
|
Loo SK, Yates ME, Yang S, Oesterreich S, Lee AV, Wang X. Fusion-associated carcinomas of the breast: Diagnostic, prognostic, and therapeutic significance. Genes Chromosomes Cancer 2022; 61:261-273. [PMID: 35106856 PMCID: PMC8930468 DOI: 10.1002/gcc.23029] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 11/11/2022] Open
Abstract
Recurrent gene fusions comprise a class of viable genetic targets in solid tumors that have culminated several recent breakthrough cancer therapies. Their role in breast cancer, however, remains largely underappreciated due to the complexity of genomic rearrangements in breast malignancy. Just recently, we and others have identified several recurrent gene fusions in breast cancer with important clinical and biological implications. Examples of the most significant recurrent gene fusions to date include (1) ESR1::CCDC170 gene fusions in luminal B and endocrine-resistant breast cancer that exert oncogenic function via modulating the HER2/HER3/SRC Proto-Oncogene (SRC) complex, (2) ESR1 exon 6 fusions in metastatic disease that drive estrogen-independent estrogen-receptor transcriptional activity, (3) BCL2L14::ETV6 fusions in a more aggressive form of the triple-negative subtype that prime epithelial-mesenchymal transition and endow paclitaxel resistance, (4) the ETV6::NTRK3 fusion in secretory breast carcinoma that constitutively activates NTRK3 kinase, (5) the oncogenic MYB-NFIB fusion as a genetic driver underpinning adenoid cystic carcinomas of the breast that activates MYB Proto-Oncogene (MYB) pathway, and (6) the NOTCH/microtubule-associated serine-threonine (MAST) kinase gene fusions that activate NOTCH and MAST signaling. Importantly, these fusions are enriched in more aggressive and lethal breast cancer presentations and appear to confer therapeutic resistance. Thus, these gene fusions could be utilized as genetic biomarkers to identify patients who require more intensive treatment and surveillance. In addition, kinase fusions are currently being evaluated in breast cancer clinical trials and ongoing mechanistic investigation is exposing therapeutic vulnerabilities in patients with fusion-positive disease.
Collapse
Affiliation(s)
- Suet Kee Loo
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Megan E. Yates
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15232, USA
| | - Sichun Yang
- Center for Proteomics and Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steffi Oesterreich
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Adrian V. Lee
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Xiaosong Wang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15232, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| |
Collapse
|
12
|
CDCP1: A promising diagnostic biomarker and therapeutic target for human cancer. Life Sci 2022; 301:120600. [DOI: 10.1016/j.lfs.2022.120600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
|
13
|
Lin Z, Zhang Z, Zheng H, Xu H, Wang Y, Chen C, Liu J, Yi G, Li Z, Wang X, Huang G. Molecular mechanism by which CDCP1 promotes proneural-mesenchymal transformation in primary glioblastoma. Cancer Cell Int 2022; 22:151. [PMID: 35410293 PMCID: PMC9003964 DOI: 10.1186/s12935-021-02373-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/28/2021] [Indexed: 11/10/2022] Open
Abstract
Background Compared with the proneural (PN) subtype of glioblastoma (GBM), the mesenchymal (MES) subtype is more invasive and immune evasive and is closely related to poor prognosis. Here, we used transcriptome data and experimental evidence to indicate that CUB domain-containing protein 1 (CDCP1) is a novel regulator that facilitates the transformation of PN-GBM to MES-GBM. Methods The mRNA expression data of CDCP1 in glioma were collected from the TCGA, CGGA and GEO databases, and in vitro experiments verified CDCP1 expression in glioma tissue samples. Independent prognostic analysis revealed the correlation of the CDCP1 expression level and patient survival. Bioinformatics analysis and experiments verified the biological function of CDCP1. Multivariate proportional hazards models and a PPI network were used to select key genes. A prognostic risk model for predicting the survival of glioma patients was constructed based on the selected genes. Results The results showed that the expression of CDCP1 increased with increasing tumor grade and that the overexpression of CDCP1 correlated with a poor prognosis. CDCP1 was highly expressed in MES-GBM but weakly expressed in PN-GBM. The risk model (considering CDCP1 combined with CD44 and ITGAM expression) could represent a tool for predicting survival and prognosis in glioma patients. Conclusions Our study indicates that CDCP1 plays an important role in facilitating the transformation of PN-GBM to MES-GBM. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02373-1.
Collapse
Affiliation(s)
- Zhiying Lin
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi, China.,Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China
| | - Zhu Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haojie Zheng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haiyan Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yajuan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Chao Chen
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junlu Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Guozhong Yi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China.,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xiaoyan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China. .,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Guanglong Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou, 510515, Guangdong, China. .,The Laboratory for Precision Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
14
|
Kawase N, Sugihara A, Kajiwara K, Hiroshima M, Akamatsu K, Nada S, Matsumoto K, Ueda M, Okada M. SRC kinase activator CDCP1 promotes hepatocyte growth factor-induced cell migration/invasion of a subset of breast cancer cells. J Biol Chem 2022; 298:101630. [PMID: 35085554 PMCID: PMC8867115 DOI: 10.1016/j.jbc.2022.101630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer invasion and metastasis are the major causes of cancer patient mortality. Various growth factors, including hepatocyte growth factor (HGF), are known to promote cancer invasion and metastasis, but the regulatory mechanisms involved are not fully understood. Here, we show that HGF-promoted migration and invasion of breast cancer cells are regulated by CUB domain–containing protein 1 (CDCP1), a transmembrane activator of SRC kinase. In metastatic human breast cancer cell line MDA-MB-231, which highly expresses the HGF receptor MET and CDCP1, we show that CDCP1 knockdown attenuated HGF-induced MET activation, followed by suppression of lamellipodia formation and cell migration/invasion. In contrast, in the low invasive/nonmetastatic breast cancer cell line T47D, which had no detectable MET and CDCP1 expression, ectopic MET expression stimulated the HGF-dependent activation of invasive activity, and concomitant CDCP1 expression activated SRC and further promoted invasive activity. In these cells, CDCP1 expression dramatically activated HGF-induced membrane remodeling, which was accompanied by activation of the small GTPase Rac1. Analysis of guanine nucleotide exchange factors revealed that ARHGEF7 was specifically required for CDCP1-dependent induction of HGF-induced invasive ability. Furthermore, immunofluorescence staining demonstrated that CDCP1 coaccumulated with ARHGEF7. Finally, we confirmed that the CDCP1-SRC axis was also crucial for HGF and ARHGEF7-RAC1 signaling in MDA-MB-231 cells. Altogether, these results demonstrate that the CDCP1-SRC-ARHGEF7-RAC1 pathway plays an important role in the HGF-induced invasion of a subset of breast cancer cells.
Collapse
Affiliation(s)
- Naoyuki Kawase
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Atsuya Sugihara
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kentaro Kajiwara
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Michio Hiroshima
- Laboratory for Cell Signaling Dynamics, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Kanako Akamatsu
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Oncogene Research, World Premier International Immunology Frontier Research Centre, Osaka University, Suita, Osaka, Japan
| | - Shigeyuki Nada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masahiro Ueda
- Laboratory for Cell Signaling Dynamics, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan; Laboratory of Single Molecule Biology, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Masato Okada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan; Laboratory of Oncogene Research, World Premier International Immunology Frontier Research Centre, Osaka University, Suita, Osaka, Japan; Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
15
|
Iwabuchi E, Miki Y, Sasano H. The Visualization of Protein-Protein Interactions in Breast Cancer: Deployment Study in Pathological Examination. Acta Histochem Cytochem 2021; 54:177-183. [PMID: 35023880 PMCID: PMC8727844 DOI: 10.1267/ahc.21-00084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/14/2021] [Indexed: 11/22/2022] Open
Abstract
The therapeutic strategy is determined by protein expression using immunohistochemistry of estrogen receptor (ER), progesterone receptor, and human epidermal growth factor receptor 2 (HER2) in formalin-fixed paraffin-embedded (FFPE) breast cancer tissues. However, few proteins function independently, and many of them functions due to protein-protein interactions (PPIs) with other proteins. Therefore, it is important to focus on PPIs. This review summarizes the PPIs of ER and HER2 in breast cancer, especially those using a proximity ligation assay that can visualize PPIs in FFPE tissues. In particular, assessing the interaction of CEACAM6 with HER2 may serve as a surrogate marker for the efficacy of trastuzumab in patients with breast cancer. Therefore, in this review, the technique used to detect the interaction of CEACAM6 and HER2 in routinely processed pathological specimens will be applied to the clinical practice of drug selection. We showed the possibility as a novel pathological examination method using PPIs.
Collapse
Affiliation(s)
- Erina Iwabuchi
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Yasuhiro Miki
- Department of Disaster Obstetrics and Gynecology, International Research Institute of Disaster Science (IRIDes), Tohoku University
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine
| |
Collapse
|
16
|
Ebian HF, Issa DR, Al-Karamany AS, Etewa RL, El Maghraby HM, Hussein S. Evaluation of CDCP1 (CD318) and endoglin (CD105) expression as prognostic markers in acute myeloid leukemia. Cancer Biomark 2021; 34:285-296. [DOI: 10.3233/cbm-210346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The most commonly used prognostic factors in acute myeloid leukemia (AML) are cytogenetic, molecular, and morphological markers. However, AML prognosis is still unfavorable particularly in adults. So, further reliable markers are urgently needed to improve the risk stratification and treatment decisions. CUB domain-containing protein 1 (CDCP1; CD318) and endoglin (CD105) are new markers correlated with poor prognosis in different solid tumors, but their role in AML prognosis is not fully evaluated. OBJECTIVES: This work aimed to evaluate the prognostic role of CD318 and CD105 in AML and their impact on the outcomes. METHODS: Sixty-five newly diagnosed AML patients were included in this study. CD318 and CD105 expression was assessed by quantitative real-time polymerase chain reaction. Patients were followed up for ∼ 2 years to evaluate the prognostic impact of gene expression on the outcomes. RESULTS: Patients with high CD318 and CD105 showed higher white blood cell (WBC) count, M2 subtype, poor cytogenetic risk, reduced complete remission, and a greater number of deaths compared to low CD318 and CD105. CD318 was correlated with CD105, and both were correlated with WBC count, bone marrow blasts, and peripheral blood blasts. After a follow-up period of up to 24 months, relapse-free survival for high CD318 and CD105 was significantly different (42.1% and 52.6% vs. 64.5% and 58.1% for low CD318 and CD105, respectively). Survival was worse in patients with high CD318 and CD105, as the mean survival time was 13.9 and 13.3 months compared to 24 and 22.7 months in low CD318 and CD105, respectively. CONCLUSIONS: CD318 and CD105 are upregulated in AML patients. Their overexpression was associated with poor response to treatment and poor outcomes. Therefore, CD318 and CD105 can be useful prognostic markers in AML.
Collapse
Affiliation(s)
- Huda F. Ebian
- Clinical Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Dina R. Issa
- Internal Medicine Department, Faculty of Medicine, Helwan University, Egypt
| | - Amira S. Al-Karamany
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rasha L. Etewa
- Pathology Department College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Hanaa M. El Maghraby
- Medical Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Samia Hussein
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
17
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
18
|
WEE1 inhibition reverses trastuzumab resistance in HER2-positive cancers. Gastric Cancer 2021; 24:1003-1020. [PMID: 33723720 DOI: 10.1007/s10120-021-01176-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND To date, many efforts have been made to understand the resistance mechanism of trastuzumab in human epidermal growth factor receptor 2 (HER2)-positive breast and gastric cancer. However, there is still a huge unmet medical need for patients with trastuzumab resistance. METHODS In our study, we generated four trastuzumab-resistant (HR) cancer cell lines from ERBB2-amplified gastric and biliary tract cancer cell lines (SNU-216, NCI-N87, SNU-2670, and SNU-2773). RESULTS Here, we found higher PD-L1 expression in trastuzumab-resistant (HR) HER2-positive cancer cells than in parental cells, and blocking PD-L1 reversed the resistance to trastuzumab in HR cells. Trastuzumab upregulated PD-L1 expression via NF-κB activation in both parental and HR cells, however, led to DNA damage only in parental cells. The WEE1 inhibitor adavosertib, which downregulates PD-L1 expression, enhanced trastuzumab efficacy by blocking BRCA1-CMTM6-PD-L1 signals and the HER2-CDCP-1-SRC axis. Additionally, the levels of galectin-9, CD163, FoxP3, and CTLA-4 were diminished by blocking WEE1 in the presence of human PBMCs in vitro. CONCLUSION Taken together, the strategy of co-targeting HER2 and WEE1 could overcome resistance to trastuzumab in HER2-positive cancers, supporting further clinical development in HER2-positive cancer patients.
Collapse
|
19
|
Li D, San M, Zhang J, Yang A, Xie W, Chen Y, Lu X, Zhang Y, Zhao M, Feng X, Zheng Y. Oxytocin receptor induces mammary tumorigenesis through prolactin/p-STAT5 pathway. Cell Death Dis 2021; 12:588. [PMID: 34099636 PMCID: PMC8184747 DOI: 10.1038/s41419-021-03849-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022]
Abstract
Oxytocin receptor (OXTR) is involved in social behaviors, thermoregulation, and milk ejection, yet little is known about its role in breast cancer. To investigate the role of OXTR in mammary gland development and tumorigenesis, a transgenic mouse model of OXTR overexpression (++Oxtr) was used. Overexpression of OXTR-induced progressive mammary hyperplasia, unexpected milk production, and tumorigenesis in females. OXTR-induced mammary tumors showed ERBB2 upregulation and mixed histological subtypes with predomination of papillary and medullary carcinomas. OXTR overexpression led to an activation of prolactin (PRL)/p-STAT5 pathway and created a microenvironment that promotes mammary-specific tumorigenesis. PRL inhibitor bromocriptine (Br) could mitigate OXTR-driven mammary tumor growth. The study demonstrates Oxtr is an oncogene and a potential drug target for HER2-type breast cancer.
Collapse
Affiliation(s)
- Dan Li
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China.,The Precise Medicine Center, Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China
| | - Mingjun San
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Jing Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Anlan Yang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Wanhua Xie
- The Precise Medicine Center, Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China
| | - Yang Chen
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China.,School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xiaodan Lu
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Yuntao Zhang
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Mingyue Zhao
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Xuechao Feng
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Yaowu Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China. .,Institute of Biomedical Sciences, Shanxi University, Taiyuan, Shanxi, 030006, China.
| |
Collapse
|
20
|
Kajiwara K, Yamano S, Aoki K, Okuzaki D, Matsumoto K, Okada M. CDCP1 promotes compensatory renal growth by integrating Src and Met signaling. Life Sci Alliance 2021; 4:4/4/e202000832. [PMID: 33574034 PMCID: PMC7893822 DOI: 10.26508/lsa.202000832] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022] Open
Abstract
CDCP1 promotes HGF-induced compensatory renal growth by focally and temporally integrating Src and Met-STAT3 signaling in lipid rafts. Compensatory growth of organs after loss of their mass and/or function is controlled by hepatocyte growth factor (HGF), but the underlying regulatory mechanisms remain elusive. Here, we show that CUB domain-containing protein 1 (CDCP1) promotes HGF-induced compensatory renal growth. Using canine kidney cells as a model of renal tubules, we found that HGF-induced temporal up-regulation of Src activity and its scaffold protein, CDCP1, and that the ablation of CDCP1 robustly abrogated HGF-induced phenotypic changes, such as morphological changes and cell growth/proliferation. Mechanistic analyses revealed that up-regulated CDCP1 recruits Src into lipid rafts to activate STAT3 associated with the HGF receptor Met, and activated STAT3 induces the expression of matrix metalloproteinases and mitogenic factors. After unilateral nephrectomy in mice, the Met-STAT3 signaling is transiently up-regulated in the renal tubules of the remaining kidney, whereas CDCP1 ablation attenuates regenerative signaling and significantly suppresses compensatory growth. These findings demonstrate that CDCP1 plays a crucial role in controlling compensatory renal growth by focally and temporally integrating Src and Met signaling.
Collapse
Affiliation(s)
- Kentaro Kajiwara
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shotaro Yamano
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Kanagawa, Japan
| | - Kazuhiro Aoki
- Division of Quantitative Biology, Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, Aichi, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kunio Matsumoto
- Division of Tumor Dynamics and Regulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Masato Okada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
21
|
Khan T, Kryza T, Lyons NJ, He Y, Hooper JD. The CDCP1 Signaling Hub: A Target for Cancer Detection and Therapeutic Intervention. Cancer Res 2021; 81:2259-2269. [PMID: 33509939 DOI: 10.1158/0008-5472.can-20-2978] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/22/2020] [Accepted: 01/22/2021] [Indexed: 11/16/2022]
Abstract
CUB-domain containing protein 1 (CDCP1) is a type I transmembrane glycoprotein that is upregulated in malignancies of the breast, lung, colorectum, ovary, kidney, liver, pancreas, and hematopoietic system. Here, we discuss CDCP1 as an important hub for oncogenic signaling and its key roles in malignant transformation and summarize approaches focused on exploiting it for cancer diagnosis and therapy. Elevated levels of CDCP1 are associated with progressive disease and markedly poorer survival. Predominantly located on the cell surface, CDCP1 lies at the nexus of key tumorigenic and metastatic signaling cascades, including the SRC/PKCδ, PI3K/AKT, WNT, and RAS/ERK axes, the oxidative pentose phosphate pathway, and fatty acid oxidation, making important functional contributions to cancer cell survival and growth, metastasis, and treatment resistance. These findings have stimulated the development of agents that target CDCP1 for detection and treatment of a range of cancers, and results from preclinical models suggest that these approaches could be efficacious and have manageable toxicity profiles.
Collapse
Affiliation(s)
- Tashbib Khan
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Nicholas J Lyons
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia.
| |
Collapse
|
22
|
Li L, Lin L, Veeraraghavan J, Hu Y, Wang X, Lee S, Tan Y, Schiff R, Wang XS. Therapeutic role of recurrent ESR1-CCDC170 gene fusions in breast cancer endocrine resistance. Breast Cancer Res 2020; 22:84. [PMID: 32771039 PMCID: PMC7414578 DOI: 10.1186/s13058-020-01325-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 07/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Endocrine therapy is the most common treatment for estrogen receptor (ER)-positive breast cancer, but its effectiveness is limited by high rates of primary and acquired resistance. There are likely many genetic causes, and recent studies suggest the important role of ESR1 mutations and fusions in endocrine resistance. Previously, we reported a recurrent ESR1 fusion called ESR1-CCDC170 in 6–8% of the luminal B breast cancers that has a worse clinical outcome after endocrine therapy. Despite being the most frequent ESR1 fusion, its functional role in endocrine resistance has not been studied in vivo, and the engaged mechanism and therapeutic relevance remain uncharacterized. Methods The endocrine sensitivities of HCC1428 or T47D breast cancer cells following genetic perturbations of ESR1-CCDC170 were assessed using clonogenic assays and/or xenograft mouse models. The underlying mechanisms were investigated by reverse phase protein array, western blotting, immunoprecipitation, and bimolecular fluorescence complementation assays. The sensitivity of ESR1-CCDC170 expressing breast cancer cells to concomitant treatments of tamoxifen and HER/SRC inhibitors was assessed by clonogenic assays. Results Our results suggested that different ESR1-CCDC170 fusions endow different levels of reduced endocrine sensitivity in vivo, resulting in significant survival disadvantages. Further investigation revealed a novel mechanism that ESR1-CCDC170 binds to HER2/HER3/SRC and activates SRC/PI3K/AKT signaling. Silencing of ESR1-CCDC170 in the fusion-positive cell line, HCC1428, downregulates HER2/HER3, represses pSRC/pAKT, and improves endocrine sensitivity. More important, breast cancer cells expressing ectopic or endogenous ESR1-CCDC170 are highly sensitive to treatment regimens combining endocrine agents with the HER2 inhibitor lapatinib and/or the SRC inhibitor dasatinib. Conclusion ESR1-CCDC170 may endow breast cancer cell survival under endocrine therapy via maintaining/activating HER2/HER3/SRC/AKT signaling which implies a potential therapeutic strategy for managing these fusion positive tumors.
Collapse
Affiliation(s)
- Li Li
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ling Lin
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiheng Hu
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xian Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sanghoon Lee
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiao-Song Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA. .,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA. .,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
23
|
Kryza T, Khan T, Puttick S, Li C, Sokolowski KA, Tse BWC, Cuda T, Lyons N, Gough M, Yin J, Parkin A, Deryugina EI, Quigley JP, Law RHP, Whisstock JC, Riddell AD, Barbour AP, Wyld DK, Thomas PA, Rose S, Snell CE, Pajic M, He Y, Hooper JD. Effective targeting of intact and proteolysed CDCP1 for imaging and treatment of pancreatic ductal adenocarcinoma. Theranostics 2020; 10:4116-4133. [PMID: 32226543 PMCID: PMC7086361 DOI: 10.7150/thno.43589] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background: CUB domain-containing protein 1 (CDCP1) is a cell surface receptor regulating key signalling pathways in malignant cells. CDCP1 has been proposed as a molecular target to abrogate oncogenic signalling pathways and specifically deliver anti-cancer agents to tumors. However, the development of CDCP1-targeting agents has been questioned by its frequent proteolytic processing which was thought to result in shedding of the CDCP1 extracellular domain limiting its targetability. In this study, we investigated the relevance of targeting CDCP1 in the context of pancreatic ductal adenocarcinoma (PDAC) and assess the impact of CDCP1 proteolysis on the effectiveness of CDCP1 targeting agents. Methods: The involvement of CDCP1 in PDAC progression was assessed by association analysis in several PDAC cohorts and the proteolytic processing of CDCP1 was evaluated in PDAC cell lines and patient-derived cells. The consequences of CDCP1 proteolysis on its targetability in PDAC cells was assessed using immunoprecipitation, immunostaining and biochemical assays. The involvement of CDCP1 in PDAC progression was examined by loss-of-function in vitro and in vivo experiments employing PDAC cells expressing intact or cleaved CDCP1. Finally, we generated antibody-based imaging and therapeutic agents targeting CDCP1 to demonstrate the feasibility of targeting this receptor for detection and treatment of PDAC tumors. Results: High CDCP1 expression in PDAC is significantly associated with poorer patient survival. In PDAC cells proteolysis of CDCP1 does not always result in the shedding of CDCP1-extracellular domain which can interact with membrane-bound CDCP1 allowing signal transduction between the different CDCP1-fragments. Targeting CDCP1 impairs PDAC cell functions and PDAC tumor growth independently of CDCP1 cleavage status. A CDCP1-targeting antibody is highly effective at delivering imaging radionuclides and cytotoxins to PDAC cells allowing specific detection of tumors by PET/CT imaging and superior anti-tumor effects compared to gemcitabine in in vivo models. Conclusion: Independent of its cleavage status, CDCP1 exerts oncogenic functions in PDAC and has significant potential to be targeted for improved radiological staging and treatment of this cancer. Its elevated expression by most PDAC tumors and lack of expression by normal pancreas and other major organs, suggest that targeting CDCP1 could benefit a significant proportion of PDAC patients. These data support the further development of CDCP1-targeting agents as personalizable tools for effective imaging and treatment of PDAC.
Collapse
|
24
|
Identification of CD318 (CDCP1) as novel prognostic marker in AML. Ann Hematol 2020; 99:477-486. [PMID: 31965270 PMCID: PMC7060168 DOI: 10.1007/s00277-020-03907-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
Abstract
Genetic and morphological markers are well-established prognostic factors in acute myeloid leukemia (AML). However, further reliable markers are urgently needed to improve risk stratification in AML. CD318 (CDCP1) is a transmembrane protein which in solid tumors promotes formation of metastasis and correlates with poor survival. Despite its broad expression on hematological precursor cells, its prognostic significance in hematological malignancies so far remains unclear. Here, we evaluated the role of CD318 as novel prognostic marker in AML by immunophenotyping of leukemic blasts. Flow cytometric evaluation of CD318 on leukemic cells in 70 AML patients revealed a substantial expression in 40/70 (57%) of all cases. CD318 surface levels were significantly correlated with overall survival in patients receiving anthracycline-based induction therapy or best available alternative therapy. Using receiver-operating characteristics, we established a cut-off value to define CD318lo and CD318hi expression in both cohorts. Notably, high CD318 expression correlated inversely as prognostic marker in both treatment cohorts: as poor prognostic marker in patients receiving intense therapy, whereas upon palliative care it correlated with better outcome. In conclusion, FACS-based determination of CD318 expression may serve as novel prognostic factor depending on implemented therapy in AML patients.
Collapse
|
25
|
Casciano JC, Perry C, Cohen-Nowak AJ, Miller KD, Vande Voorde J, Zhang Q, Chalmers S, Sandison ME, Liu Q, Hedley A, McBryan T, Tang HY, Gorman N, Beer T, Speicher DW, Adams PD, Liu X, Schlegel R, McCarron JG, Wakelam MJO, Gottlieb E, Kossenkov AV, Schug ZT. MYC regulates fatty acid metabolism through a multigenic program in claudin-low triple negative breast cancer. Br J Cancer 2020; 122:868-884. [PMID: 31942031 PMCID: PMC7078291 DOI: 10.1038/s41416-019-0711-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 11/22/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background Recent studies have suggested that fatty acid oxidation (FAO) is a key metabolic pathway for the growth of triple negative breast cancers (TNBCs), particularly those that have high expression of MYC. However, the underlying mechanism by which MYC promotes FAO remains poorly understood. Methods We used a combination of metabolomics, transcriptomics, bioinformatics, and microscopy to elucidate a potential mechanism by which MYC regulates FAO in TNBC. Results We propose that MYC induces a multigenic program that involves changes in intracellular calcium signalling and fatty acid metabolism. We determined key roles for fatty acid transporters (CD36), lipases (LPL), and kinases (PDGFRB, CAMKK2, and AMPK) that each contribute to promoting FAO in human mammary epithelial cells that express oncogenic levels of MYC. Bioinformatic analysis further showed that this multigenic program is highly expressed and predicts poor survival in the claudin-low molecular subtype of TNBC, but not other subtypes of TNBCs, suggesting that efforts to target FAO in the clinic may best serve claudin-low TNBC patients. Conclusion We identified critical pieces of the FAO machinery that have the potential to be targeted for improved treatment of patients with TNBC, especially the claudin-low molecular subtype.
Collapse
Affiliation(s)
- Jessica C Casciano
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Caroline Perry
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Adam J Cohen-Nowak
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Katelyn D Miller
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Johan Vande Voorde
- The Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Qifeng Zhang
- The Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Susan Chalmers
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - Mairi E Sandison
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK.,Department of Biomedical Engineering, University of Strathclyde, Wolfson Centre, 106 Rottenrow, Glasgow, G4 0NW, UK
| | - Qin Liu
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Ann Hedley
- The Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Tony McBryan
- The Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.,Institute of Cancer Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, G61 1BD, UK
| | - Hsin-Yao Tang
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Nicole Gorman
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Thomas Beer
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - David W Speicher
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Xuefeng Liu
- Center for Cell Reprogramming, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3900 Reservoir Road, Washington D.C., 20057, USA
| | - Richard Schlegel
- Center for Cell Reprogramming, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3900 Reservoir Road, Washington D.C., 20057, USA
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | | | - Eyal Gottlieb
- The Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, 1 Efron St. Bat Galim, 3525433, Haifa, Israel
| | - Andrew V Kossenkov
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA
| | - Zachary T Schug
- The Wistar Institute, Molecular and Cellular Oncogenesis, 3601 Spruce Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
26
|
Harrington BS, He Y, Khan T, Puttick S, Conroy PJ, Kryza T, Cuda T, Sokolowski KA, Tse BWC, Robbins KK, Arachchige BJ, Stehbens SJ, Pollock PM, Reed S, Weroha SJ, Haluska P, Salomon C, Lourie R, Perrin LC, Law RHP, Whisstock JC, Hooper JD. Anti-CDCP1 immuno-conjugates for detection and inhibition of ovarian cancer. Am J Cancer Res 2020; 10:2095-2114. [PMID: 32104500 PMCID: PMC7019151 DOI: 10.7150/thno.30736] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
CUB-domain containing protein 1 (CDCP1) is a cancer associated cell surface protein that amplifies pro-tumorigenic signalling by other receptors including EGFR and HER2. Its potential as a cancer target is supported by studies showing that anti-CDCP1 antibodies inhibit cell migration and survival in vitro, and tumor growth and metastasis in vivo. Here we characterize two anti-CDCP1 antibodies, focusing on immuno-conjugates of one of these as a tool to detect and inhibit ovarian cancer. Methods: A panel of ovarian cancer cell lines was examined for cell surface expression of CDCP1 and loss of expression induced by anti-CDCP1 antibodies 10D7 and 41-2 using flow cytometry and Western blot analysis. Surface plasmon resonance analysis and examination of truncation mutants was used to analyse the binding properties of the antibodies for CDCP1. Live-cell spinning-disk confocal microscopy of GFP-tagged CDCP1 was used to track internalization and intracellular trafficking of CDCP1/antibody complexes. In vivo, zirconium 89-labelled 10D7 was detected by positron-emission tomography imaging, of an ovarian cancer patient-derived xenograft grown intraperitoneally in mice. The efficacy of cytotoxin-conjugated 10D7 was examined against ovarian cancer cells in vitro and in vivo. Results: Our data indicate that each antibody binds with high affinity to the extracellular domain of CDCP1 causing rapid internalization of the receptor/antibody complex and degradation of CDCP1 via processes mediated by the kinase Src. Highlighting the potential clinical utility of CDCP1, positron-emission tomography imaging, using zirconium 89-labelled 10D7, was able to detect subcutaneous and intraperitoneal xenograft ovarian cancers in mice, including small (diameter <3 mm) tumor deposits of an ovarian cancer patient-derived xenograft grown intraperitoneally in mice. Furthermore, cytotoxin-conjugated 10D7 was effective at inhibiting growth of CDCP1-expressing ovarian cancer cells in vitro and in vivo. Conclusions: These data demonstrate that CDCP1 internalizing antibodies have potential for killing and detection of CDCP1 expressing ovarian cancer cells.
Collapse
|
27
|
CDCP1 enhances Wnt signaling in colorectal cancer promoting nuclear localization of β-catenin and E-cadherin. Oncogene 2019; 39:219-233. [DOI: 10.1038/s41388-019-0983-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 11/08/2022]
|
28
|
Gao W, Xu Y, Chen T, Du Z, Liu X, Hu Z, Wei D, Gao C, Zhang W, Li Q. Targeting oxidative pentose phosphate pathway prevents recurrence in mutant Kras colorectal carcinomas. PLoS Biol 2019; 17:e3000425. [PMID: 31461438 PMCID: PMC6736310 DOI: 10.1371/journal.pbio.3000425] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/10/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recurrent tumors originate from cancer stem cells (CSCs) that survive conventional treatments. CSCs consist of heterogeneous subpopulations that display distinct sensitivity to anticancer drugs. Such a heterogeneity presents a significant challenge in preventing tumor recurrence. In the current study, we observed that quiescent CUB-domain-containing protein 1 (CDCP1)+ CSCs are enriched after chemotherapy in mutant Kirsten rat sarcoma viral oncogene homolog (Kras) colorectal carcinomas (CRCs) and serve as a reservoir for recurrence. Mechanistically, glucose catabolism in CDCP1+ CSCs is routed to the oxidative pentose phosphate pathway (PPP); multiple cycling of carbon backbones in the oxidative PPP potentially maximizes NADPH reduction to counteract chemotherapy-induced reactive oxygen species (ROS) formation, thereby allowing CDCP1+ CSCs to survive chemotherapeutic attack. This is dependent on silent mating type information regulation 2 homolog 5 (Sirt5)-mediated inhibition of the glycolytic enzyme triosephosphate isomerase (TPI) through demalonylation of Lys56. Blocking demalonylation of TPI at Lys56 increases chemosensitivity of CDCP1+ CSCSs and delays recurrence of mutant Kras CRCs in vivo. These findings pinpoint a new therapeutic approach for combating mutant Kras CRCs.
Collapse
Affiliation(s)
- WenChao Gao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of General Surgery, ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - YuTing Xu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tao Chen
- Endoscopy Center, ZhongShan Hospital, Fudan University, Shanghai, China
| | - ZunGuo Du
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Pathology, HuaShan Hospital, Fudan University, Shanghai, China
| | - XiuJuan Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Pathology, HuaShan Hospital, Fudan University, Shanghai, China
| | - ZhiQian Hu
- Department of General Surgery, ChangZheng Hospital, Second Military Medical University, Shanghai, China
| | - Dong Wei
- Department of Anus and Intestine Surgery, PLA Central Hospital 150, Luoyang, China
| | - ChunFang Gao
- Department of Anus and Intestine Surgery, PLA Central Hospital 150, Luoyang, China
| | - Wei Zhang
- Department of Cancer Biology, Cancer Center of Wake Forest Baptist Medical Center, Winston-Salem, North Carolina, United States of America
| | - QingQuan Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Abstract
OBJECTIVE The incidence of pancreatic adenocarcinoma (PA) approximates its prevalence, as the malignancy is almost consistently fatal within a year. Although the currently available adjuvant therapy seems to provide survival benefit, it is only moderate, and the standard regimen has not yet been established. Therefore, more biological resources to investigate the PA are needed. METHODS Here, we established and characterized 10 human pancreatic cancer cell lines derived from primary tumor mass. Whole exome sequencing technique was used to identify driver mutations and aberrant pathways in each cell line. RESULTS Five anticancer drugs were treated to find half maximal effective concentration (EC50), and the response was analyzed in reference to mutational status. Frame shift mutations in ARID1A gene and HER2 amplification were mutually related to better response to the anticancer drugs. In contrast, frame shift mutation in MSH6 gene was associated with resistance to anticancer drugs. CONCLUSIONS In summary, we established 10 pancreatic cancer cell lines and integrated various molecular aberrations and features of pancreatic cancer cells. Our biological resources are expected to contribute to facilitating research on PA.
Collapse
|
30
|
Evidence that cell surface localization of serine protease activity facilitates cleavage of the protease activated receptor CDCP1. Biol Chem 2018; 399:1091-1097. [DOI: 10.1515/hsz-2017-0308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/07/2018] [Indexed: 12/13/2022]
Abstract
Abstract
The cellular receptor CUB domain containing protein 1 (CDCP1) is commonly elevated and functionally important in a range of cancers. CDCP1 is cleaved by serine proteases at adjacent sites, arginine 368 (R368) and lysine 369 (K369), which induces cell migration in vitro and metastasis in vivo. We demonstrate that membrane localization of serine protease activity increases efficacy of cleavage of CDCP1, and that both secreted and membrane anchored serine proteases can have distinct preferences for cleaving at CDCP1-R368 and CDCP1-K369. Approaches that disrupt membrane localization of CDCP1 cleaving serine proteases may interfere with the cancer promoting effects of CDCP1 proteolysis.
Collapse
|
31
|
Lee J, Kim H, Lee JE, Shin SJ, Oh S, Kwon G, Kim H, Choi YY, White MA, Paik S, Cheong JH, Kim HS. Selective Cytotoxicity of the NAMPT Inhibitor FK866 Toward Gastric Cancer Cells With Markers of the Epithelial-Mesenchymal Transition, Due to Loss of NAPRT. Gastroenterology 2018; 155:799-814.e13. [PMID: 29775598 DOI: 10.1053/j.gastro.2018.05.024] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Markers of the epithelial-to-mesenchymal transition (EMT) in gastric tumor tissues are associated with poor patient outcomes. We performed a screen to identify pharmacologic compounds that kill gastric cancer cells with EMT-associated gene expression patterns and investigate their mechanisms. METHODS We identified 29 gastric cancer cell lines with a gene expression signature previously associated with an EMT subtype, based on data from RNA sequence analyses, and confirmed the mesenchymal phenotypes of 7 lines (Hs746T, SNU1750, MKN1, SK4, SNU484, SNU668, and YCC11), based on invasive activity and protein markers. We screened 1,345 compounds for their ability to kill cells with the EMT signature compared with cell lines without this pattern. We tested the effects of identified compounds in BALB/c nude mice bearing GA077 tumors; mice were given intraperitoneal injections of the compound or vehicle (control) twice daily for 24 days and tumor growth was monitored. Proteins associated with the toxicity of the compounds were overexpressed in MKN1 and SNU484 cells or knocked down in MKN45 and SNU719 using small interfering RNAs. We performed immunohistochemical analyses of 942 gastric cancer tissues and investigated associations between EMT markers and protein expression patterns. RESULTS The nicotinamide phosphoribosyltransferase inhibitor FK866 killed 6 of 7 gastric cancer cell lines with EMT-associated gene expression signatures but not gastric cancer cells without this signature. The 6 EMT-subtype gastric cell lines expressed significantly low levels of nicotinic acid phosphoribosyltransferase (NAPRT), which makes the cells hypersensitive to nicotinamide phosphoribosyltransferase inhibition. Gastric cell lines that expressed higher levels of NAPRT, regardless of EMT markers, were sensitized to FK866 after knockdown of NAPRT, whereas overexpression of NAPRT in deficient EMT cell lines protected them from FK866-mediated toxicity. Administration of FK866 to nude mice with tumors grown from GA077 cells (human gastric cancer tumors of the EMT subtype) led to tumor regression in 2 weeks; FK866 did not affect tumors grown from MKN45 cells without the EMT expression signature. Loss of NAPRT might promote the EMT, because it stabilizes β-catenin. We correlated the EMT gene expression signature with lower levels of NAPRT in 942 gastric tumors from patients; we also found lower levels of NAPRT mRNA in colorectal, pancreatic, and lung adenocarcinoma tissues with the EMT gene expression signature. CONCLUSIONS FK866 selectively kills gastric cancer cells with an EMT gene expression signature by inhibiting nicotinamide phosphoribosyltransferase in cells with NAPRT deficiency. Loss of NAPRT expression, frequently through promoter hypermethylation, is observed in many gastric tumors of the EMT subtype. FK866 might be used to treat patients with tumors of this subtype.
Collapse
Affiliation(s)
- Jooyoung Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hyosil Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Eun Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Hanyang University College of Medicine, Seoul, Korea
| | - Sejin Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Gino Kwon
- Graduate Program for Nanomedical Science, Yonsei University, Seoul, Korea
| | - Hakhyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Young Choi
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Michael A White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Soonmyung Paik
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Ho Cheong
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Department of Surgery, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| | - Hyun Seok Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
Zhou Q, Jin P, Liu J, Wang F, Xi S. HER2 and Src co-regulate proliferation, migration and transformation by downstream signaling pathways in arsenite-treated human uroepithelial cells. Metallomics 2018; 10:1141-1159. [PMID: 30066004 DOI: 10.1039/c8mt00131f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Epidemiological studies have established a strong association between arsenic exposure in drinking water and an increased incidence of bladder cancer in arseniasis-endemic areas. Increased expression of HER2 has been observed in various types of human malignancies including bladder cancer. This study investigated the role of HER2 in arsenite-induced transformation of uroepithelial cells SV-HUC-1 and the role of Src family kinases in HER2 signaling. We found that the expression HER2 and Src were increased following chronic arsenite exposure in a time-dependent fashion. Chronic arsenite exposure also led to an upregulation of proliferation factors such as cyclin D1, COX2, PCNA, VEGF, and HIF-1α. Furthermore, Ras/Raf/MAPK, PI3K/AKT, and JAK2/STAT3 signaling pathways were activated by arsenite treatment. Importantly, these changes were inhibited by HER2 inhibitors and in HER2 knocked down cells. In addition, downregulation of HER2 inhibited cell growth and migration properties of arsenite-treated cells. Inhibition of Src also inhibits activation of signaling pathways and malignant transformation of cells. And we obtained evidence of an interaction between HER2 and Src in SV-HUC-1 cell lines. These results suggest that HER2 and Src may play an important role in arsenite-induced transformation by multiple downstream signals pathways.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Environmental and Occupational Health, Liaoning Provincial Key Laboratory of Arsenic Biological Effect and Poisoning, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, P. R. China.
| | | | | | | | | |
Collapse
|
33
|
FBXL14 abolishes breast cancer progression by targeting CDCP1 for proteasomal degradation. Oncogene 2018; 37:5794-5809. [PMID: 29973690 DOI: 10.1038/s41388-018-0372-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/16/2018] [Accepted: 05/29/2018] [Indexed: 01/10/2023]
Abstract
Understanding the molecular mechanisms that underlie the aggressive behavior and relapse of breast cancer may help in the development of novel therapeutic interventions. CUB-domain-containing protein 1 (CDCP1), a transmembrane adaptor protein, is highly maintained and required in the context of cellular metastatic potential in triple-negative breast cancer (TNBC). For this reason, gene expression levels of CDCP1 have been considered as a prognostic marker in TNBC. However, not rarely, transcript levels of genes do not reflect always the levels of proteins, due to the post-transcriptional regulation. Here we show that miR-17/20a control the FBXL14 E3 ligase, establishing FBXL14 as an upstream regulator of the CDCP1 pathway. FBXL14 acts as an novel interaction partner of CDCP1, and facilitates its ubiquitination and proteasomal degradation with an enhanced capacity to suppress CDCP1 protein stability that eventually prevents CDCP1 target genes involved in breast cancer metastasis. Our findings first time uncovers the regulatory mechanism of CDCP-1 protein stabilization, more predictable criteria than gene expression levels for prognosis of breast cancer patients.
Collapse
|
34
|
Li W, Xu L, Che X, Li H, Zhang Y, Song N, Wen T, Hou K, Yang Y, Zhou L, Xin X, Xu L, Zeng X, Shi S, Liu Y, Qu X, Teng Y. C-Cbl reverses HER2-mediated tamoxifen resistance in human breast cancer cells. BMC Cancer 2018; 18:507. [PMID: 29720121 PMCID: PMC5930956 DOI: 10.1186/s12885-018-4387-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 04/17/2018] [Indexed: 01/12/2023] Open
Abstract
Background Tamoxifen is a frontline therapy for estrogen receptor (ER)-positive breast cancer in premenopausal women. However, many patients develop resistance to tamoxifen, and the mechanism underlying tamoxifen resistance is not well understood. Here we examined whether ER-c-Src-HER2 complex formation is involved in tamoxifen resistance. Methods MTT and colony formation assays were used to measure cell viability and proliferation. Western blot was used to detect protein expression and protein complex formations were detected by immunoprecipitation and immunofluorescence. SiRNA was used to examine the function of HER2 in of BT474 cells. An in vivo xenograft animal model was established to examine the role of c-Cbl in tumor growth. Results MTT and colony formation assay showed that BT474 cells are resistant to tamoxifen and T47D cells are sensitive to tamoxifen. Immunoprecipitation experiments revealed ER-c-Src-HER2 complex formation in BT474 cells but not in T47D cells. However, ER-c-Src-HER2 complex formation was detected after overexpressing HER2 in T47D cells and these cells were more resistant to tamoxifen. HER2 knockdown by siRNA in BT474 cells reduced ER-c-Src-HER2 complex formation and reversed tamoxifen resistance. ER-c-Src-HER2 complex formation was also disrupted and tamoxifen resistance was reversed in BT474 cells by the c-Src inhibitor PP2 and HER2 antibody trastuzumab. Nystatin, a lipid raft inhibitor, reduced ER-c-Src-HER2 complex formation and partially reversed tamoxifen resistance. ER-c-Src-HER2 complex formation was disrupted by overexpression of c-Cbl but not by the c-Cbl ubiquitin ligase mutant. In addition, c-Cbl could reverse tamoxifen resistance in BT474 cells, but the ubiquitin ligase mutant had no effect. The effect of c-Cbl was validated in BT474 tumor-bearing nude mice in vivo. Immunofluorescence also revealed ER-c-Src-HER2 complex formation was reduced in tumor tissues of nude mice with c-Cbl overexpression. Conclusions Our results suggested that c-Cbl can reverse tamoxifen resistance in HER2-overexpressing breast cancer cells by inhibiting the formation of the ER-c-Src-HER2 complex. Electronic supplementary material The online version of this article (10.1186/s12885-018-4387-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Ling Xu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Haizhou Li
- Jinzhou Center Hospital, Jinzhou, 121000, Liaoning, China
| | - Ye Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Na Song
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Ti Wen
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Kezuo Hou
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, Shenyang, 110001, Liaoning, China
| | - Lu Zhou
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xing Xin
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Lu Xu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xue Zeng
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Sha Shi
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Yunpeng Liu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China. .,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China.
| | - Yuee Teng
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, China. .,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, NO. 155, North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
35
|
Turdo F, Bianchi F, Gasparini P, Sandri M, Sasso M, De Cecco L, Forte L, Casalini P, Aiello P, Sfondrini L, Agresti R, Carcangiu ML, Plantamura I, Sozzi G, Tagliabue E, Campiglio M. CDCP1 is a novel marker of the most aggressive human triple-negative breast cancers. Oncotarget 2018; 7:69649-69665. [PMID: 27626701 PMCID: PMC5342505 DOI: 10.18632/oncotarget.11935] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 08/27/2016] [Indexed: 12/31/2022] Open
Abstract
CDCP1, a transmembrane noncatalytic receptor, the expression of which has been associated with a poor prognosis in certain epithelial cancers, was found to be expressed in highly aggressive triple-negative breast cancer (TNBC) cell models, in which it promoted aggressive activities—ie, migration, invasion, anchorage-independent tumor growth, and the formation of vascular-like structures in vitro. By immunohistochemical (IHC) analysis of 100 human TNBC specimens, CDCP1 was overexpressed in 57% of samples, 38% of which exhibited a gain in CDCP1 copy number by fluorescence in situ hybridization (FISH). CDCP1 positivity was significantly associated between FISH and IHC. CDCP1 expression and gains in CDCP1 copy number synergized with nodal (N) status in determining disease-free and distant disease-free survival. The hazard ratios (HRs) of the synergies between CDCP1 positivity by IHC and FISH and lymph node positivity in predicting relapse did not differ significantly, indicating that CDCP1 overexpression in human primary TNBCs, regardless of being driven by gains in CDCP1, is for a critical factor in the progression of N-positive TNBCs. Thus, CDCP1 is a novel marker of the most aggressive N-positive TNBCs and a potential therapeutic target.
Collapse
Affiliation(s)
- Federica Turdo
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Francesca Bianchi
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy.,Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Patrizia Gasparini
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marco Sandri
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marianna Sasso
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Loris De Cecco
- Functional Genomic Core Facility, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Luca Forte
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Patrizia Casalini
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Piera Aiello
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Roberto Agresti
- Division of Surgical Oncology, Breast Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Maria Luisa Carcangiu
- Division of Breast Anatomy Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Ilaria Plantamura
- Start-Up Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Manuela Campiglio
- Molecular Targeting Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| |
Collapse
|
36
|
Zhang X, Yao J, Guo K, Huang H, Huai S, Ye R, Niu B, Ji T, Han W, Li J. The functional mechanism of miR-125b in gastric cancer and its effect on the chemosensitivity of cisplatin. Oncotarget 2017; 9:2105-2119. [PMID: 29416757 PMCID: PMC5788625 DOI: 10.18632/oncotarget.23249] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have shown drug resistance of gastric cancer cells could be modulated by abnormal expression of microRNAs. Cisplatin (DDP) is one of the most commonly used drugs for chemotherapy of gastric cancer. In this study, the potential function of miR-125b on DDP resistance in gastric cancer cells was investigated. Sixteen miRNAs significantly differential expressed in gastric tumor tissues and adjacent tissues were characterized and their corresponding putative target genes were also screened. MiR-125b was selected as our focus for its evident down-regulated expression among candidate genes. Real-time polymerase chain reaction assay indicated that miR-125b was significantly down-regulated in gastric cancer tissues and various cell lines. HER2 was identified as a target gene of miR-125b by dual luciferase reporter assay and Western blot. Moreover, miR-125b overexpression inhibited not only the proliferation, migration, and invasion abilities of HGC-27 and MGC-803 cells, but also in vivo tumor growth of MGC-803 cells by an intratumoral delivery approach. Notably, we observed up-regulated miR-125b contributed to the chemosensitivity of DDP in HGC-27 and MGC-803 cells at different concentrations and also possessed sensibilization for DDP at different times. MiR-125b expression was found to be related to lymph node metastasis, HER2 expression and overall survival of patients through correlation analysis. Collectively, these results indicate miR-125b may regulate DDP resistance as a promising therapeutic target for gastric cancer treatment in future.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jie Yao
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Kai Guo
- Department of Gastroenterology, The 161th Hospital of PLA, Wuhan 430010, P.R. China
| | - Hu Huang
- Department of Oncology, The 161th Hospital of PLA, Wuhan 430010, P.R. China
| | - Siyuan Huai
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Rui Ye
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853, P.R. China.,Department of Oncology, Beidaihe Sanatorium of Beijing Military Command, Qinhuangdao 066100, P.R. China
| | - Baolong Niu
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Tiannan Ji
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Sciences, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jianxiong Li
- Department of Radiotherapy, Hainan Branch of Chinese PLA General Hospital, Sanya 572000, P.R. China
| |
Collapse
|
37
|
CDCP1 drives triple-negative breast cancer metastasis through reduction of lipid-droplet abundance and stimulation of fatty acid oxidation. Proc Natl Acad Sci U S A 2017; 114:E6556-E6565. [PMID: 28739932 DOI: 10.1073/pnas.1703791114] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is notoriously aggressive with high metastatic potential, which has recently been linked to high rates of fatty acid oxidation (FAO). Here we report the mechanism of lipid metabolism dysregulation in TNBC through the prometastatic protein, CUB-domain containing protein 1 (CDCP1). We show that a "low-lipid" phenotype is characteristic of breast cancer cells compared with normal breast epithelial cells and negatively correlates with invasiveness in 3D culture. Using coherent anti-Stokes Raman scattering and two-photon excited fluorescence microscopy, we show that CDCP1 depletes lipids from cytoplasmic lipid droplets (LDs) through reduced acyl-CoA production and increased lipid utilization in the mitochondria through FAO, fueling oxidative phosphorylation. These findings are supported by CDCP1's interaction with and inhibition of acyl CoA-synthetase ligase (ACSL) activity. Importantly, CDCP1 knockdown increases LD abundance and reduces TNBC 2D migration in vitro, which can be partially rescued by the ACSL inhibitor, Triacsin C. Furthermore, CDCP1 knockdown reduced 3D invasion, which can be rescued by ACSL3 co-knockdown. In vivo, inhibiting CDCP1 activity with an engineered blocking fragment (extracellular portion of cleaved CDCP1) lead to increased LD abundance in primary tumors, decreased metastasis, and increased ACSL activity in two animal models of TNBC. Finally, TNBC lung metastases have lower LD abundance than their corresponding primary tumors, indicating that LD abundance in primary tumor might serve as a prognostic marker for metastatic potential. Our studies have important implications for the development of TNBC therapeutics to specifically block CDCP1-driven FAO and oxidative phosphorylation, which contribute to TNBC migration and metastasis.
Collapse
|
38
|
Liu W, Chang J, Liu M, Yuan J, Zhang J, Qin J, Xia X, Wang Y. Quantitative proteomics profiling reveals activation of mTOR pathway in trastuzumab resistance. Oncotarget 2017; 8:45793-45806. [PMID: 28507275 PMCID: PMC5542228 DOI: 10.18632/oncotarget.17415] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/12/2017] [Indexed: 12/18/2022] Open
Abstract
Trastuzumab is an antibody-based therapy drug targeting HER2-overexpressing tumors. While it has been proven to be very successful initially, most patients eventually develop resistance to trastuzumab. The mechanism of drug resistance is not well understood. Identifying pathways that mediate trastuzumab resistance will improve our understanding of the underlying mechanism and is crucial for the development of therapeutic strategies to overcome resistance.Here we report a quantitative proteomics profiling of a trastuzumab-sensitive (T-S) gastric cancer cell line NCI N87 and a trastuzumab-resistant NCI N87 (T-R) subline generated by low-dose, continuous trastuzumab treatment. By identifying proteins differentially expressed in these two cell lines, we show that multiple pathways including mTOR, Wnt, DNA damage response and metabolic pathways are significantly altered. We further confirm by western blotting that protein levels of multiple components of the mTOR pathway, including mTOR, AKT and RPS6KB1, are increased, whereas AKT1S1 is decreased, suggesting the activation of mTOR pathway. Importantly, treatment of AZD8055, an mTOR inhibitor, leads to the decreased phosphorylation levels of mTOR downstream molecules RPS6KB1 at Thr421/Ser424 and AKT at Ser473. Furthermore, AZD8055 also preferentially reduces viability, and inhibits migration and invasion abilities of the T-R cells. Together, our findings indicate that mTOR pathway is among multiple signaling pathways that mediate trastuzumab resistance in NCI N87 T-R cells, and that mTOR inhibitors may be used to treat trastuzumab resistant, HER2-positive gastric cancer tumors.
Collapse
Affiliation(s)
- Wenhu Liu
- School of Pharmaceutical Sciences and Innovative Drug Research Center, Chongqing University, Chongqing 401331, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, Beijing 102206, China
- School of Pharmacy, North Sichuan Medical College, Nanchong 637007, China
| | - Jinxia Chang
- School of Basic Medical Sciences, North Sichuan Medical College, Nanchong 637007, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, Beijing 102206, China
| | - Jiangbei Yuan
- School of Pharmaceutical Sciences and Innovative Drug Research Center, Chongqing University, Chongqing 401331, China
| | - Jinqiang Zhang
- School of Pharmaceutical Sciences and Innovative Drug Research Center, Chongqing University, Chongqing 401331, China
| | - Jun Qin
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, Beijing 102206, China
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Xuefeng Xia
- School of Pharmaceutical Sciences and Innovative Drug Research Center, Chongqing University, Chongqing 401331, China
| | - Yi Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences, Beijing Proteome Research Center, Beijing 102206, China
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
39
|
Jin MH, Nam AR, Park JE, Bang JH, Bang YJ, Oh DY. Resistance Mechanism against Trastuzumab in HER2-Positive Cancer Cells and Its Negation by Src Inhibition. Mol Cancer Ther 2017; 16:1145-1154. [PMID: 28223426 DOI: 10.1158/1535-7163.mct-16-0669] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/21/2016] [Accepted: 02/04/2017] [Indexed: 11/16/2022]
Abstract
Trastuzumab in combination with chemotherapy is the standard of care for patients with human epidermal growth factor receptor 2 (HER2)-positive breast and gastric cancers. Several resistance mechanisms against anti-HER2 therapy have been proposed. Src activation has been suggested to be responsible for the resistance of HER2-positive breast cancer. In our study, we generated four trastuzumab-resistant (HR) cancer cell lines from HER2-amplified gastric and biliary tract cancer cell lines (SNU-216, NCI-N87, SNU-2670, and SNU-2773). Elevated Src phosphorylation was detected in SNU2670HR and NCI-N87HR cell lines, but not in SNU216HR or SNU2773HR cell lines. In SNU216HR and SNU2773HR cell lines, phospho-FAK (focal adhesion kinase) was elevated. Bosutinib as a Src inhibitor suppressed growth, cell-cycle progression, and migration in both parental and HR cell lines. Specifically, Src interacted with FAK to affect downstream molecules such as AKT, ERK, and STAT3. Bosutinib showed more potent antitumor effects in Src-activated HR cell lines than parental cell lines. Taken together, this study suggests that Src inhibition may be an effective measure to overcome trastuzumab resistance in HER2-positive cancer. Mol Cancer Ther; 16(6); 1145-54. ©2017 AACR.
Collapse
Affiliation(s)
- Mei Hua Jin
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ah-Rong Nam
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Eun Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ju-Hee Bang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yung-Jue Bang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Do-Youn Oh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Nakashima K, Uekita T, Yano S, Kikuchi JI, Nakanishi R, Sakamoto N, Fukumoto K, Nomoto A, Kawamoto K, Shibahara T, Yamaguchi H, Sakai R. Novel small molecule inhibiting CDCP1-PKCδ pathway reduces tumor metastasis and proliferation. Cancer Sci 2017; 108:1049-1057. [PMID: 28256037 PMCID: PMC5448658 DOI: 10.1111/cas.13218] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/23/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022] Open
Abstract
CUB domain‐containing protein‐1 (CDCP1) is a trans‐membrane protein predominantly expressed in various cancer cells and involved in tumor progression. CDCP1 is phosphorylated at tyrosine residues in the intracellular domain by Src family kinases and recruits PKCδ to the plasma membrane through tyrosine phosphorylation‐dependent association with the C2 domain of PKCδ, which in turn induces a survival signal in an anchorage‐independent condition. In this study, we used our cell‐free screening system to identify a small compound, glycoconjugated palladium complex (Pd‐Oqn), which significantly inhibited the interaction between the C2 domain of PKCδ and phosphorylated CDCP1. Immunoprecipitation assays demonstrated that Pd‐Oqn hindered the intercellular interaction of phosphorylated CDCP1 with PKCδ and also suppressed the phosphorylation of PKCδ but not that of ERK or AKT. In addition, Pd‐Oqn inhibited the colony formation of gastric adenocarcinoma 44As3 cells in soft agar as well as their invasion. In mouse models, Pd‐Oqn markedly reduced the peritoneal dissemination of gastric adenocarcinoma cells and the tumor growth of pancreatic cancer orthotopic xenografts. These results suggest that the novel compound Pd‐Oqn reduces tumor metastasis and growth by inhibiting the association between CDCP1 and PKCδ, thus potentially representing a promising candidate among therapeutic reagents targeting protein–protein interaction.
Collapse
Affiliation(s)
- Katsuhiko Nakashima
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Takamasa Uekita
- Department of Applied Chemistry, National Defense Academy, Yokosuka, Japan
| | - Shigenobu Yano
- Graduate School of Materials Science, Nara Institute of Science and Technology, Nara, Japan
| | - Jun-Ichi Kikuchi
- Graduate School of Materials Science, Nara Institute of Science and Technology, Nara, Japan
| | - Ruri Nakanishi
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Nozomi Sakamoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka, Japan
| | - Keisuke Fukumoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka, Japan
| | - Akihiro Nomoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, Osaka, Japan
| | - Keisuke Kawamoto
- Department of Chemistry, Graduate School of Natural Science, Kanazawa University, Kanazawa, Japan
| | - Takashi Shibahara
- Department of Chemistry, Okayama University of Science, Okayama, Japan
| | - Hideki Yamaguchi
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Tokyo, Japan
| | - Ryuichi Sakai
- Division of Refractory and Advanced Cancer, National Cancer Center Research Institute, Tokyo, Japan.,Division of Biochemistry, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
41
|
Extracellular vesicles for liquid biopsy in prostate cancer: where are we and where are we headed? Prostate Cancer Prostatic Dis 2017; 20:251-258. [PMID: 28374743 PMCID: PMC5569339 DOI: 10.1038/pcan.2017.7] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 12/23/2022]
Abstract
Background: Extracellular vesicles (EVs) are a heterogeneous class of lipid bound particles shed by any cell in the body in physiological and pathological conditions. EVs play critical functions in intercellular communication. EVs can actively travel in intercellular matrices and eventually reach the circulation. They can also be released directly in biological fluids where they appear to be stable. Because the molecular content of EVs reflects the composition of the cell of origin, they have recently emerged as a promising source of biomarkers in a number of diseases. EV analysis is particularly attractive in cancer patients that frequently present with increased numbers of circulating EVs. Methods: We sought to review the current literature on the molecular profile of prostate cancer-derived EVs in model systems and patient biological fluids in an attempt to draw some practical and universal conclusions on the use of EVs as a tool for liquid biopsy in clinical specimens. Results: We discuss advantages and limitations of EV-based liquid biopsy approaches summarizing salient studies on protein, DNA and RNA. Several candidate biomarkers have been identified so far but these results are difficult to apply to the clinic. However, the field is rapidly moving toward the implementation of novel tools to isolate cancer-specific EVs that are free of benign EVs and extra-vesicular contaminants. This can be achieved by identifying markers that are exquisitely present in tumor cell-derived EVs. An important contribution might also derive from a better understanding of EV types that may play specific functions in tumor progression and that may be a source of cancer-specific markers. Conclusions: EV analysis holds strong promises for the development of non-invasive biomarkers in patients with prostate cancer. Implementation of modern methods for EV isolation and characterization will enable to interrogate circulating EVs in vivo.
Collapse
|
42
|
Chen Y, Harrington BS, Lau KCN, Burke LJ, He Y, Iconomou M, Palmer JS, Meade B, Lumley JW, Hooper JD. Development of an enzyme-linked immunosorbent assay for detection of CDCP1 shed from the cell surface and present in colorectal cancer serum specimens. J Pharm Biomed Anal 2017; 139:65-72. [PMID: 28279929 DOI: 10.1016/j.jpba.2017.02.047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 11/17/2022]
Abstract
CUB domain containing protein 1 (CDCP1) is a transmembrane protein involved in progression of several cancers. When located on the plasma membrane, full-length 135kDa CDCP1 can undergo proteolysis mediated by serine proteases that cleave after two adjacent amino acids (arginine 368 and lysine 369). This releases from the cell surface two 65kDa fragments, collectively termed ShE-CDCP1, that differ by one carboxyl terminal residue. To evaluate the function of CDCP1 and its potential utility as a cancer biomarker, in this study we developed an enzyme-linked immunosorbent assay (ELISA) to reliably and easily measure the concentration of ShE-CDCP1 in biological samples. Using a reference standard we demonstrate that the developed ELISA has a working range of 0.68-26.5ng/ml, and the limit of detection is 0.25ng/ml. It displays high intra-assay (repeatability) and high inter-assay (reproducibility) precision with all coefficients of variation ≤7%. The ELISA also displays high accuracy detecting ShE-CDCP1 levels at ≥94.8% of actual concentration using quality control samples. We employed the ELISA to measure the concentration of ShE-CDCP1 in human serum samples with our results suggesting that levels are significantly higher in serum of colorectal cancer patients compared with serum from individuals with benign conditions (p<0.05). Our data also suggest that colorectal cancer patients with stage II-IV disease have at least 50% higher serum levels of ShE-CDCP1 compared with stage I cases (p<0.05). We conclude that the developed ELISA is a suitable method to quantify ShE-CDCP1 concentration in human serum.
Collapse
Affiliation(s)
- Yang Chen
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia
| | - Brittney S Harrington
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia
| | - Kevin C N Lau
- School of Medicine, The University of Queensland, Brisbane, Qld 4072, Australia
| | - Lez J Burke
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia
| | - Mary Iconomou
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Qld 4059, Australia
| | - James S Palmer
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia
| | - Brian Meade
- Colorectal Unit, Princess Alexandra Hospital, Woolloongabba Qld 4102, Australia
| | | | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Qld 4102, Australia.
| |
Collapse
|
43
|
O'Brien CM, Chy HS, Zhou Q, Blumenfeld S, Lambshead JW, Liu X, Kie J, Capaldo BD, Chung TL, Adams TE, Phan T, Bentley JD, McKinstry WJ, Oliva K, McMurrick PJ, Wang YC, Rossello FJ, Lindeman GJ, Chen D, Jarde T, Clark AT, Abud HE, Visvader JE, Nefzger CM, Polo JM, Loring JF, Laslett AL. New Monoclonal Antibodies to Defined Cell Surface Proteins on Human Pluripotent Stem Cells. Stem Cells 2017; 35:626-640. [PMID: 28009074 PMCID: PMC5412944 DOI: 10.1002/stem.2558] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/31/2016] [Accepted: 11/18/2016] [Indexed: 01/28/2023]
Abstract
The study and application of human pluripotent stem cells (hPSCs) will be enhanced by the availability of well‐characterized monoclonal antibodies (mAbs) detecting cell‐surface epitopes. Here, we report generation of seven new mAbs that detect cell surface proteins present on live and fixed human ES cells (hESCs) and human iPS cells (hiPSCs), confirming our previous prediction that these proteins were present on the cell surface of hPSCs. The mAbs all show a high correlation with POU5F1 (OCT4) expression and other hPSC surface markers (TRA‐160 and SSEA‐4) in hPSC cultures and detect rare OCT4 positive cells in differentiated cell cultures. These mAbs are immunoreactive to cell surface protein epitopes on both primed and naive state hPSCs, providing useful research tools to investigate the cellular mechanisms underlying human pluripotency and states of cellular reprogramming. In addition, we report that subsets of the seven new mAbs are also immunoreactive to human bone marrow‐derived mesenchymal stem cells (MSCs), normal human breast subsets and both normal and tumorigenic colorectal cell populations. The mAbs reported here should accelerate the investigation of the nature of pluripotency, and enable development of robust cell separation and tracing technologies to enrich or deplete for hPSCs and other human stem and somatic cell types. Stem Cells2017;35:626–640
Collapse
Affiliation(s)
- Carmel M O'Brien
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Hun S Chy
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Qi Zhou
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | | | - Jack W Lambshead
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Xiaodong Liu
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Joshua Kie
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Bianca D Capaldo
- The Walter and Eliza Hall Institute (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology
| | - Tung-Liang Chung
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Timothy E Adams
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia
| | - Tram Phan
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia
| | - John D Bentley
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia
| | | | - Karen Oliva
- Department of Surgery, Cabrini Monash University, Malvern, Victoria, Australia
| | - Paul J McMurrick
- Department of Surgery, Cabrini Monash University, Malvern, Victoria, Australia
| | - Yu-Chieh Wang
- Department of Chemical Physiology.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Fernando J Rossello
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Geoffrey J Lindeman
- The Walter and Eliza Hall Institute (WEHI), Parkville, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Di Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA
| | - Thierry Jarde
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Cancer Program, Monash Biomedicine Discovery Institute.,Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Amander T Clark
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA
| | - Helen E Abud
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.,Cancer Program, Monash Biomedicine Discovery Institute
| | - Jane E Visvader
- The Walter and Eliza Hall Institute (WEHI), Parkville, Victoria, Australia.,Department of Medical Biology
| | - Christian M Nefzger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Jose M Polo
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Jeanne F Loring
- Department of Chemical Physiology.,Center for Regenerative Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Andrew L Laslett
- Clayton and Parkville, CSIRO Manufacturing, Victoria, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
44
|
miR-217 and CAGE form feedback loop and regulates the response to anti-cancer drugs through EGFR and HER2. Oncotarget 2016; 7:10297-321. [PMID: 26863629 PMCID: PMC4891121 DOI: 10.18632/oncotarget.7185] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/23/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNA array analysis revealed that miR-217 expression was decreased in anti-cancer drug-resistant Malme3MR cancer cells. CAGE, a cancer/testis antigen, was predicted as a target of miR-217. Luciferase activity and ChIP assays revealed a negative feedback relationship between CAGE and miR-217. miR-217 and CAGE oppositely regulated the response to anti-cancer drugs such as taxol, gefitinib and trastuzumab, an inhibitor of HER2. miR-217 negatively regulated the tumorigenic, metastatic, angiogenic, migration and invasion potential of cancer cells. The xenograft of Malme3MR cells showed an increased expression of pEGFRY845. CAGE and miR-217 inhibitor regulated the expression of pEGFRY845. CAGE showed interactions with EGFR and HER2 and regulated the in vivo sensitivity to trastuzumab. The down-regulation of EGFR or HER2 enhanced the sensitivity to anti-cancer drugs. CAGE showed direct regulation of HER2 and was necessary for the interaction between EGFR and HER2 in Malme3MR cells. miR-217 inhibitor induced interactions of CAGE with EGFR and HER2 in Malme3M cells. The inhibition of EGFR by CAGE-binding GTGKT peptide enhanced the sensitivity to gefitinib and trastuzumab and prevented interactions of EGFR with CAGE and HER2. Our results show that miR-217-CAGE feedback loop serves as a target for overcoming resistance to various anti-cancer drugs, including EGFR and HER2 inhibitors.
Collapse
|
45
|
Wicki A, Mandalà M, Massi D, Taverna D, Tang H, Hemmings BA, Xue G. Acquired Resistance to Clinical Cancer Therapy: A Twist in Physiological Signaling. Physiol Rev 2016; 96:805-29. [DOI: 10.1152/physrev.00024.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although modern therapeutic strategies have brought significant progress to cancer care in the last 30 years, drug resistance to targeted monotherapies has emerged as a major challenge. Aberrant regulation of multiple physiological signaling pathways indispensable for developmental and metabolic homeostasis, such as hyperactivation of pro-survival signaling axes, loss of suppressive regulations, and impaired functionalities of the immune system, have been extensively investigated aiming to understand the diversity of molecular mechanisms that underlie cancer development and progression. In this review, we intend to discuss the molecular mechanisms of how conventional physiological signal transduction confers to acquired drug resistance in cancer patients. We will particularly focus on protooncogenic receptor kinase inhibition-elicited tumor cell adaptation through two major core downstream signaling cascades, the PI3K/Akt and MAPK pathways. These pathways are crucial for cell growth and differentiation and are frequently hyperactivated during tumorigenesis. In addition, we also emphasize the emerging roles of the deregulated host immune system that may actively promote cancer progression and attenuate immunosurveillance in cancer therapies. Understanding these mechanisms may help to develop more effective therapeutic strategies that are able to keep the tumor in check and even possibly turn cancer into a chronic disease.
Collapse
Affiliation(s)
- Andreas Wicki
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Mario Mandalà
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Massi
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Taverna
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Huifang Tang
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Brian A. Hemmings
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Gongda Xue
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| |
Collapse
|
46
|
Tamaskovic R, Schwill M, Nagy-Davidescu G, Jost C, Schaefer DC, Verdurmen WPR, Schaefer JV, Honegger A, Plückthun A. Intermolecular biparatopic trapping of ErbB2 prevents compensatory activation of PI3K/AKT via RAS-p110 crosstalk. Nat Commun 2016; 7:11672. [PMID: 27255951 PMCID: PMC4895728 DOI: 10.1038/ncomms11672] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 04/18/2016] [Indexed: 02/07/2023] Open
Abstract
Compensatory mechanisms, such as relief of AKT-ErbB3-negative feedback, are known to desensitize ErbB2-dependent tumours to targeted therapy. Here we describe an adaptation mechanism leading to reactivation of the PI3K/AKT pathway during trastuzumab treatment, which occurs independently of ErbB3 re-phosphorylation. This signalling bypass of phospho-ErbB3 operates in ErbB2-overexpressing cells via RAS-PI3K crosstalk and is attributable to active ErbB2 homodimers. As demonstrated by dual blockade of ErbB2/RAS and ErbB3 by means of pharmacological inhibition, RNA interference or by specific protein binders obstructing the RAS–p110α interaction, both routes must be blocked to prevent reactivation of the PI3K/AKT pathway. Applying these general principles, we developed biparatopic designed ankyrin repeat proteins (DARPins) trapping ErbB2 in a dimerization-incompetent state, which entail pan-ErbB inhibition and a permanent OFF state in the oncogenic signalling, thereby triggering extensive apoptosis in ErbB2-addicted tumours. Thus, these novel insights into mechanisms underlying network robustness provide a guide for overcoming adaptation response to ErbB2/ErbB3-targeted therapy. Targeted therapy of ErbB2-dependent tumours often provokes an adaptive response leading to reactivation of the PI3K/AKT pathway. Here the authors identify an ErbB3-independent compensatory mechanism comprising Ras/PI3K activation directly by ErbB2, and develop biparatopic panErbB inhibitors to block this mode of resistance.
Collapse
Affiliation(s)
- Rastislav Tamaskovic
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Martin Schwill
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Gabriela Nagy-Davidescu
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Christian Jost
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Dagmar C Schaefer
- Institute of Laboratory Animal Science, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Wouter P R Verdurmen
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Annemarie Honegger
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstr. 190, Zurich 8057, Switzerland
| |
Collapse
|
47
|
Ha JR, Siegel PM, Ursini-Siegel J. The Tyrosine Kinome Dictates Breast Cancer Heterogeneity and Therapeutic Responsiveness. J Cell Biochem 2016; 117:1971-90. [PMID: 27392311 DOI: 10.1002/jcb.25561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 03/24/2016] [Indexed: 12/13/2022]
Abstract
Phospho-tyrosine signaling networks control numerous biological processes including cellular differentiation, cell growth and survival, motility, and invasion. Aberrant regulation of the tyrosine kinome is a hallmark of malignancy and influences all stages of breast cancer progression, from initiation to the development of metastatic disease. The success of specific tyrosine kinase inhibitors strongly validates the clinical relevance of tyrosine phosphorylation networks in breast cancer pathology. However, a significant degree of redundancy exists within the tyrosine kinome. Numerous receptor and cytoplasmic tyrosine kinases converge on a core set of signaling regulators, including adaptor proteins and tyrosine phosphatases, to amplify pro-tumorigenic signal transduction pathways. Mutational activation, amplification, or overexpression of one or more components of the tyrosine kinome represents key contributing events responsible for the tumor heterogeneity that is observed in breast cancers. It is this molecular heterogeneity that has become the most significant barrier to durable clinical responses due to the development of therapeutic resistance. This review focuses on recent literature that supports a prominent role for specific components of the tyrosine kinome in the emergence of unique breast cancer subtypes and in shaping breast cancer plasticity, sensitivity to targeted therapies, and the eventual emergence of acquired resistance. J. Cell. Biochem. 117: 1971-1990, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jacqueline R Ha
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Peter M Siegel
- Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
48
|
Wright HJ, Arulmoli J, Motazedi M, Nelson LJ, Heinemann FS, Flanagan LA, Razorenova OV. CDCP1 cleavage is necessary for homodimerization-induced migration of triple-negative breast cancer. Oncogene 2016; 35:4762-72. [PMID: 26876198 DOI: 10.1038/onc.2016.7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 01/17/2023]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive and metastatic form of breast cancer that lacks the estrogen, progesterone and HER2 receptors and is resistant to targeted and hormone therapies. TNBCs express high levels of the transmembrane glycoprotein, complement C1r/C1s, Uegf, Bmp1 (CUB)-domain containing protein 1 (CDCP1), which has been correlated with the aggressiveness and poor prognosis of multiple carcinomas. Full-length CDCP1 (flCDCP1) can be proteolytically cleaved, resulting in a cleaved membrane-bound isoform (cCDCP1). CDCP1 is phosphorylated by Src family kinases in its full-length and cleaved states, which is important for its pro-metastatic signaling. We observed that cCDCP1, compared with flCDCP1, induced a dramatic increase in phosphorylation of the migration-associated proteins: PKCδ, ERK1/2 and p38 mitogen-activated protein kinase in HEK 293T. In addition, only cCDCP1 induced migration of HEK 293T cells and rescued migration of the TNBC cell lines expressing short hairpin RNA against CDCP1. Importantly, we found that only cCDCP1 is capable of dimerization, which can be blocked by expression of the extracellular portion of cCDCP1 (ECC), indicating that dimerization occurs through CDCP1's ectodomain. We found that ECC inhibited phosphorylation of PKCδ and migration of TNBC cells in two-dimensional culture. Furthermore, ECC decreased cell invasiveness, inhibited proliferation and stimulated apoptosis of TNBC cells in three-dimensional culture, indicating that the cCDCP1 dimer is an important contributor to TNBC aggressiveness. These studies have important implications for the development of a therapeutic to block CDCP1 activity and TNBC metastasis.
Collapse
Affiliation(s)
- H J Wright
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - J Arulmoli
- Department of Biomedical Engineering, University of California, Irvine, CA, USA
| | - M Motazedi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - L J Nelson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - F S Heinemann
- Department of Pathology, Hoag Memorial Hospital Presbyterian, Newport Beach, CA, USA
| | - L A Flanagan
- Department of Biomedical Engineering, University of California, Irvine, CA, USA.,Department of Neurology, University of California, Irvine, CA, USA
| | - O V Razorenova
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| |
Collapse
|
49
|
New crossroads for potential therapeutic intervention in cancer - intersections between CDCP1, EGFR family members and downstream signaling pathways. Oncoscience 2016; 3:5-8. [PMID: 26973855 PMCID: PMC4751911 DOI: 10.18632/oncoscience.286] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/22/2016] [Indexed: 02/03/2023] Open
Abstract
Signaling pathways regulated by the receptor CDCP1 play central roles in promoting cancer and in mediating resistance to chemo- and targeted-therapies. In this perspective we briefly summarize these findings as well as data demonstrating poorer outcomes for several malignancies that exhibit elevated CDCP1 expression. Promising data from preclinical studies suggest that CDCP1 targeted agents, including therapeutic antibodies, could be useful in the treatment of cancer patients selected on the basis of activation of CDCP1 and its signaling partners including EGFR, HER2, Met and Src.
Collapse
|