51
|
Ho WJ, Jaffee EM, Zheng L. The tumour microenvironment in pancreatic cancer - clinical challenges and opportunities. Nat Rev Clin Oncol 2020; 17:527-540. [PMID: 32398706 PMCID: PMC7442729 DOI: 10.1038/s41571-020-0363-5] [Citation(s) in RCA: 639] [Impact Index Per Article: 159.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
Metastatic pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumours despite the use of multi-agent conventional chemotherapy regimens. Such poor outcomes have fuelled ongoing efforts to exploit the tumour microenvironment (TME) for therapy, but strategies aimed at deconstructing the surrounding desmoplastic stroma and targeting the immunosuppressive pathways have largely failed. In fact, evidence has now shown that the stroma is multi-faceted, which illustrates the complexity of exploring features of the TME as isolated targets. In this Review, we describe ways in which the PDAC microenvironment has been targeted and note the current understanding of the clinical outcomes that have unexpectedly contradicted preclinical observations. We also consider the more sophisticated therapeutic strategies under active investigation - multi-modal treatment approaches and exploitation of biologically integrated targets - which aim to remodel the TME against PDAC.
Collapse
Affiliation(s)
- Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, The Skip Viragh Pancreatic Cancer Center for Clinical Research and Care, and The Bloomberg-Kimmel Institute for Immunotherapy at Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
52
|
Zeng F, Zhang Y, Han X, Weng J, Gao Y. Liver Buds and Liver Organoids: New Tools for Liver Development, Disease and Medical Application. Stem Cell Rev Rep 2020; 15:774-784. [PMID: 31863336 DOI: 10.1007/s12015-019-09909-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The current understanding and effective treatment of liver disease is far from satisfactory. Liver organoids and liver buds (LBs) transforming cell culture from two dimensions(2D) to three dimensions(3D) has provided infinite possibilities for stem cells to use in clinic. Recent technological advances in the 3D culture have shown the potentiality of liver organoids and LBs as the promising tool to model in vitro liver diseases. The induced LBs and liver organoids provide a platform for cell-based therapy, liver disease models, liver organogenesis and drugs screening. And its genetic heterogeneity supplies a way for the realization of precision medicine.
Collapse
Affiliation(s)
- Fanhong Zeng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Yue Zhang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Xu Han
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China
| | - Jun Weng
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China.
| | - Yi Gao
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, 253 Gongye Street, Haizhu, Guangzhou, 510280, China.
| |
Collapse
|
53
|
Kandil SB, Jones SR, Smith S, Hiscox SE, Westwell AD. Structure-Based Virtual Screening, Synthesis and Biological Evaluation of Potential FAK-FAT Domain Inhibitors for Treatment of Metastatic Cancer. Molecules 2020; 25:molecules25153488. [PMID: 32751931 PMCID: PMC7435868 DOI: 10.3390/molecules25153488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 11/16/2022] Open
Abstract
Focal adhesion kinase (FAK) is a tyrosine kinase that is overexpressed and activated in several advanced-stage solid cancers. In cancer cells, FAK promotes the progression and metastasis of tumours. In this study, we used structure-based virtual screening to filter a library of more than 210K compounds against the focal adhesion targeting FAK-focal adhesion targeting (FAT) domain to identify 25 virtual hit compounds which were screened in the invasive breast cancer line (MDA-MB-231). Most notably, compound I showed low micromolar antiproliferative activity, as well as antimigratory activity. Moreover, examination in a model of triple negative breast cancer (TNBC), revealed that, despite not effecting FAK phosphorylation, compound I significantly impairs proliferation whilst impairing focal adhesion growth and turnover leading to reduced migration. Further optimisation and synthesis of analogues of the lead compound I using a four-step synthetic procedure was performed, and analogues were assessed for their antiproliferative activity against three breast cancer (MDA-MB-231, T47D, BT474) cell lines and one pancreatic cancer (MIAPaCa2) cell line. Compound 5f was identified as a promising lead compound with IC50 values in the range of 4.59–5.28 μM in MDA-MB-231, T47D, BT474, and MIAPaCa2. Molecular modelling and pharmacokinetic studies provided more insight into the therapeutic features of this new series.
Collapse
|
54
|
Indovina P, Forte IM, Pentimalli F, Giordano A. Targeting SRC Family Kinases in Mesothelioma: Time to Upgrade. Cancers (Basel) 2020; 12:cancers12071866. [PMID: 32664483 PMCID: PMC7408838 DOI: 10.3390/cancers12071866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant mesothelioma (MM) is a deadly tumor mainly caused by exposure to asbestos. Unfortunately, no current treatment is able to change significantly the natural history of the disease, which has a poor prognosis in the majority of patients. The non-receptor tyrosine kinase SRC and other SRC family kinase (SFK) members are frequently hyperactivated in many cancer types, including MM. Several works have indeed suggested that SFKs underlie MM cell proliferation, survival, motility, and invasion, overall affecting multiple oncogenic pathways. Consistently, SFK inhibitors effectively counteracted MM cancerous features at the preclinical level. Dasatinib, a multi-kinase inhibitor targeting SFKs, was also assessed in clinical trials either as second-line treatment for patients with unresectable MM or, more recently, as a neoadjuvant agent in patients with resectable MM. Here, we provide an overview of the molecular mechanisms implicating SFKs in MM progression and discuss possible strategies for a more successful clinical application of SFK inhibitors. Our aim is to stimulate discussion and further consideration of these agents in better designed preclinical and clinical studies to make the most of another class of powerful antitumoral drugs, which too often are lost in translation when applied to MM.
Collapse
Affiliation(s)
- Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Institute for High Performance Computing and Networking, National Research Council of Italy (ICAR-CNR), I-80131 Naples, Italy
- Correspondence: (P.I.); (F.P.)
| | - Iris Maria Forte
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
| | - Francesca Pentimalli
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, I-80131 Naples, Italy;
- Correspondence: (P.I.); (F.P.)
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
- Department of Medical Biotechnologies, University of Siena, I-53100 Siena, Italy
| |
Collapse
|
55
|
Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers (Basel) 2020; 12:cancers12051347. [PMID: 32466266 PMCID: PMC7281461 DOI: 10.3390/cancers12051347] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is the 4th leading cause of cancer deaths in the United States. The pancreatic cancer phenotype is primarily a consequence of oncogenes disturbing the resident pancreas parenchymal cell repair program. Many solid tumor types including pancreatic cancer have severe tumor fibrosis called desmoplasia. Desmoplastic stroma is coopted by the tumor as a support structure and CAFs aid in tumor growth, invasion, and metastases. This stroma is caused by cancer associated fibroblasts (CAFs), which lay down extensive connective tissue in and around the tumor cells. CAFs represent a heterogeneous population of cells that produce various paracrine molecules such as transforming growth factor-beta (TGF-beta) and platelet derived growth factors (PDGFs) that aid tumor growth, local invasion, and development of metastases. The hard, fibrotic shell of desmoplasia serves as a barrier to the infiltration of both chemo- and immunotherapy drugs and host immune cells to the tumor. Although there have been recent improvements in chemotherapy and surgical techniques for management of pancreatic cancer, the majority of patients will die from this disease. Therefore, new treatment strategies are clearly needed. CAFs represent an under-explored potential therapeutic target. This paper discusses what we know about the role of CAFs in pancreatic cancer cell growth, invasion, and metastases. Additionally, we present different strategies that are being and could be explored as anti-CAF treatments for pancreatic cancer.
Collapse
|
56
|
Chen R, Sugiyama A, Kataoka N, Sugimoto M, Yokoyama S, Fukuda A, Takaishi S, Seno H. Promoter-Level Transcriptome Identifies Stemness Associated With Relatively High Proliferation in Pancreatic Cancer Cells. Front Oncol 2020; 10:316. [PMID: 32266133 PMCID: PMC7099289 DOI: 10.3389/fonc.2020.00316] [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: 11/02/2019] [Accepted: 02/21/2020] [Indexed: 11/13/2022] Open
Abstract
Both pancreatic intraepithelial neoplasia (PanIN), a frequent precursor of pancreatic cancer, and intraductal papillary mucinous neoplasm (IPMN), a less common precursor, undergo several phases of molecular conversions and finally develop into highly malignant solid tumors with negative effects on the quality of life. We approached this long-standing issue by examining the following PanIN/IPMN cell lines derived from mouse models of pancreatic cancer: Ptf1a-Cre; KrasG12D; p53f/+ and Ptf1a-Cre; KrasG12D; and Brg1f/f pancreatic ductal adenocarcinomas (PDAs). The mRNA from these cells was subjected to a cap analysis of gene expression (CAGE) to map the transcription starting sites and quantify the expression of promoters across the genome. Two RNA samples extracted from three individual subcutaneous tumors generated by the transplantation of PanIN or IPMN cancer cell lines were used to generate libraries and Illumina Seq, with four RNA samples in total, to depict discrete transcriptional network between IPMN and PanIN. Moreover, in IPMN cells, the transcriptome tended to be enriched for suppressive and inhibitory biological processes. In contrast, the transcriptome of PanIN cells exhibited properties of stemness. Notably, the proliferation capacity of the latter cells in culture was only minimally constrained by well-known chemotherapy drugs such as GSK690693 and gemcitabine. The various transcriptional factor network systems detected in PanIN and IPMN cells reflect the distinct molecular profiles of these cell types. Further, we hope that these findings will enhance our mechanistic understanding of the characteristic molecular alterations underlying pancreatic cancer precursors. These data may provide a promising direction for therapeutic research.
Collapse
Affiliation(s)
- Ru Chen
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aiko Sugiyama
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoyuki Kataoka
- Laboratory of Cell Regulation, Department of Applied Animal Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masahiro Sugimoto
- Research and Development Center for Minimally Invasive Therapies Health Promotion and Preemptive Medicine, Tokyo Medical University, Tokyo, Japan
| | - Shoko Yokoyama
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Takaishi
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
57
|
Advances in molecular mechanisms of drugs affecting abnormal glycosylation and metastasis of breast cancer. Pharmacol Res 2020; 155:104738. [PMID: 32151681 DOI: 10.1016/j.phrs.2020.104738] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Abstract
Breast cancer remains the leading cause of cancer-related death among women worldwide, and its incidence is also increasing. High recurrence rate and metastasis rate are the key causes of poor prognosis and death. It is suggested that abnormal glycosylation plays an important role in the growth, invasion, metastasis and resistance to therapy of breast cancer cells. Meanwhile, it can be used as the biomarkers for the early detection and prognosis of breast cancer and the potential attractive targets for drug treatment. However, only a few attentions have been paid to the molecular mechanism of abnormal glycosylation in the epithelial-mesenchymal transition (EMT) of breast cancer cells and the related intervention of drugs. This manuscript thus investigated the relationship between abnormal glycosylation, the EMT, and breast cancer metastasis. Then, the process of abnormal glycosylation, the classification and their molecular regulatory mechanisms of breast cancer were analyzed in detail. Last, potential drugs are introduced in different categories, which are expected to reverse or intervene the abnormal glycosylation of breast cancer. This review is conducive to an in-depth understanding of the metastasis and drug resistance of breast cancer cells, which will provide new ideas for the clinical regulation of glycosylation and related drug treatments in breast cancer.
Collapse
|
58
|
Nevala-Plagemann C, Hidalgo M, Garrido-Laguna I. From state-of-the-art treatments to novel therapies for advanced-stage pancreatic cancer. Nat Rev Clin Oncol 2020; 17:108-123. [PMID: 31705130 DOI: 10.1038/s41571-019-0281-6] [Citation(s) in RCA: 236] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2019] [Indexed: 12/13/2022]
Abstract
Improvements in the outcomes of patients with pancreatic ductal adenocarcinoma (PDAC) have lagged behind advances made in the treatment of many other malignancies over the past few decades. For most patients with PDAC, cytotoxic chemotherapy remains the mainstay of treatment. For patients with resectable disease, modified 5-fluorouracil, leucovorin, irinotecan and oxaliplatin (mFOLFIRINOX) is the standard-of-care adjuvant therapy, although data from several randomized trials have shown improved outcomes with neoadjuvant treatment strategies. For patients with advanced-stage or metastatic disease, comprehensive genomic profiling has revealed several potentially actionable alterations in small subsets of patients and the feasibility of implementing such strategies is beginning to be confirmed. Novel therapies targeting certain aberrations, most notably BRCA1/2 mutations, mismatch repair (MMR) deficiencies or NTRK1-3 fusions, have shown considerable activity in clinical trials, and larotrectinib, entrectinib and pembrolizumab have received FDA approval for the treatment of patients with tumours harbouring NTRK fusions and MMR deficiencies, respectively, regardless of primary tumour histology. In this Review, we describe the available data on the activity of these and other agents in patients with PDAC. Our discussion is structured according to the acronym 'PRIME' to organize the various treatment strategies currently undergoing evaluation in clinical trials: Pathway inhibition, alteration of DNA Repair pathways, Immunotherapy, cancer Metabolism and targeting the Extracellular tumour microenvironment.
Collapse
Affiliation(s)
| | - Manuel Hidalgo
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA.
- Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
59
|
|
60
|
Ligeiro D, Rao M, Maia A, Castillo M, Beltran A, Maeurer M. B Cells in the Gastrointestinal Tumor Microenvironment with a Focus on Pancreatic Cancer: Opportunities for Precision Medicine? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1273:175-195. [PMID: 33119882 DOI: 10.1007/978-3-030-49270-0_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We review state-of-the-art in translational and clinical studies focusing on the tumor microenvironment (TME) with a focus on tumor-infiltrating B cells (TIBs). The TME is a dynamic matrix of mutations, immune-regulatory networks, and distinct cell-to-cell interactions which collectively impact on disease progress. We discuss relevant findings concerning B cells in pancreatic cancer, the concepts of "bystander" B cells, the role of antigen-specific B cells contributing to augmenting anticancer-directed immune responses, the role of B cells as prognostic markers for response to checkpoint inhibitors (ICBs), and the potential use in adoptive cell tumor-infiltrating lymphocyte (TIL) products.
Collapse
Affiliation(s)
- Dário Ligeiro
- Immunogenetics Unit, Lisbon Centre for Blood and Transplantation (Instituto Português do Sangue e Transplantação, IPST), Lisbon, Portugal
| | - Martin Rao
- Immunosurgery Unit, Champalimaud Center for the Unknown, Lisbon, Portugal
| | - Andreia Maia
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Mireia Castillo
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Antonio Beltran
- Department of Pathology, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Markus Maeurer
- Immunosurgery Unit, Champalimaud Center for the Unknown, Lisbon, Portugal.
- I Med Clinical University of Mainz, Mainz, Germany.
| |
Collapse
|
61
|
Yan D, Dong W, He Q, Yang M, Huang L, Kong J, Qin H, Lin T, Huang J. Circular RNA circPICALM sponges miR-1265 to inhibit bladder cancer metastasis and influence FAK phosphorylation. EBioMedicine 2019; 48:316-331. [PMID: 31648990 PMCID: PMC6838432 DOI: 10.1016/j.ebiom.2019.08.074] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/30/2019] [Accepted: 08/30/2019] [Indexed: 12/12/2022] Open
Abstract
Background Metastasis is a major obstacle in the treatment of bladder cancer (BC). Circular RNAs exert various functions in the aggressive biological behaviour of cancers. In this study, we aimed to elucidate how circPICALM influences BC metastasis. Methods The expression of circPICALM was analysed by real-time PCR. The tumourigenic properties of BC cells were evaluated using in vitro migration, invasion, and wound healing assays and an in vivo footpad model. The interaction between circPICALM and miR-1265 was confirmed by pull-down and dual-luciferase reporter assays and biotin-labelled miRNA capture. The interaction of STEAP4 and focal adhesion kinase (FAK) was confirmed by co-immunoprecipitation. Findings CircPICALM was downregulated in BC tissues, and low circPICALM expression was related to advanced T stage, high grade, lymph node positivity and poor overall survival. Overexpression of circPICALM inhibited the metastasis of BC cells, and DHX9 negatively regulated circPICALM levels. CircPICALM colocalized with miR-1265 and acted as a sponge for this miRNA, and the pro-invasion effect of miR-1265 was abolished by circPICALM overexpression. STEAP4, a target of miR-1265, suppressed metastasis; it bound to FAK to prevent autophosphorylation at Y397 and influenced EMT in BC cells. Interpretation CircPICALM can inhibit BC metastasis and bind to miR-1265 to block its pro-invasion activity. STEAP4 is a target of miR-1265 and is related to FAK phosphorylation and EMT. Fund This research was supported by National Natural Science Foundation of China, No.81772728, National Natural Science Foundation of China, No.81772719, National Natural Science Foundation of China No.81572514.
Collapse
Affiliation(s)
- Dong Yan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingqing He
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meihua Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lifang Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianqiu Kong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haide Qin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
62
|
Nguyen BT, Pyun JC, Lee SG, Kang MJ. Identification of new binding proteins of focal adhesion kinase using immunoprecipitation and mass spectrometry. Sci Rep 2019; 9:12908. [PMID: 31501460 PMCID: PMC6733923 DOI: 10.1038/s41598-019-49145-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion kinase (FAK) is a 125 kDa protein recruited as a participant in focal adhesion dynamics and serves as a signaling scaffold for the assembly and subsequent maturation of focal contact. Identification of new FAK binding proteins could reveal potential signaling targets and contribute to further development of therapeutic drugs in the treatment of colon cancer. Here, we applied a functional proteomic strategy to identify proteins that interact with FAK in human colon cancer cell line HCT-116. Proteins were targeted by coimmunoprecipitation with an anti-FAK antibody and resolved on 1D-SDS-PAGE. The gel was excised, reduced, alkylated, and trypsin digested. Tryptic peptides were separated by nano-LC-MS/MS by an LTQ-Orbitrap-Velos spectrometer. We identified 101 proteins in the immunocomplex under epithelial growth factor (EGF) stimulation. Three proteins, zyxin, nesprin-1, and desmoplakin, were discovered and validated using reciprocal immunoprecipitation and Western blot analysis. Then, we sought to study the biological relevance of these proteins by siRNA transfection of HCT-116 cells. According to the results, zyxin might play a central role as an upstream regulator to mediate critical cancer-related signaling pathways. Zyxin and nesprin-1 depletion significantly impaired cell migration and invasion capabilities. Additionally, we performed ELISA assays on serum samples from patients with colon cancer instead of cell models to quantify the protein levels of zyxin and nesprin-1. Our results suggested that zyxin and nesprin-1 are not only promising therapeutic targets but also potential diagnostic biomarkers for colon cancer.
Collapse
Affiliation(s)
- Binh Thanh Nguyen
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea.,Division of Bio-Medical Science and Technology (Biological Chemistry), Korea University of Science and Technology (UST), Daejeon, 34113, South Korea
| | - Jae-Chul Pyun
- Department of Materials and Sciences, Yonsei University, Seoul, 120-749, South Korea
| | - Sang-Guk Lee
- Department of Laboratory Medicine, Severance Hospital, Seoul, 120-752, South Korea. .,Yonsei University College of Medicine, Seoul, 120-752, South Korea.
| | - Min-Jung Kang
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea. .,Division of Bio-Medical Science and Technology (Biological Chemistry), Korea University of Science and Technology (UST), Daejeon, 34113, South Korea.
| |
Collapse
|
63
|
Parkin A, Man J, Timpson P, Pajic M. Targeting the complexity of Src signalling in the tumour microenvironment of pancreatic cancer: from mechanism to therapy. FEBS J 2019; 286:3510-3539. [PMID: 31330086 PMCID: PMC6771888 DOI: 10.1111/febs.15011] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 05/26/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer, a disease with extremely poor prognosis, has been notoriously resistant to virtually all forms of treatment. The dynamic crosstalk that occurs between tumour cells and the surrounding stroma, frequently mediated by intricate Src/FAK signalling, is increasingly recognised as a key player in pancreatic tumourigenesis, disease progression and therapeutic resistance. These important cues are fundamental for defining the invasive potential of pancreatic tumours, and several components of the Src and downstream effector signalling have been proposed as potent anticancer therapeutic targets. Consequently, numerous agents that block this complex network are being extensively investigated as potential antiinvasive and antimetastatic therapeutic agents for this disease. In this review, we will discuss the latest evidence of Src signalling in PDAC progression, fibrotic response and resistance to therapy. We will examine future opportunities for the development and implementation of more effective combination regimens, targeting key components of the oncogenic Src signalling axis, and in the context of a precision medicine-guided approach.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Jennifer Man
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
| | - Paul Timpson
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| | - Marina Pajic
- The Kinghorn Cancer CentreThe Garvan Institute of Medical ResearchSydneyAustralia
- Faculty of MedicineSt Vincent's Clinical SchoolUniversity of NSWSydneyAustralia
| |
Collapse
|
64
|
Kircher DA, Trombetti KA, Silvis MR, Parkman GL, Fischer GM, Angel SN, Stehn CM, Strain SC, Grossmann AH, Duffy KL, Boucher KM, McMahon M, Davies MA, Mendoza MC, VanBrocklin MW, Holmen SL. AKT1 E17K Activates Focal Adhesion Kinase and Promotes Melanoma Brain Metastasis. Mol Cancer Res 2019; 17:1787-1800. [PMID: 31138602 PMCID: PMC6726552 DOI: 10.1158/1541-7786.mcr-18-1372] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/18/2019] [Accepted: 05/22/2019] [Indexed: 02/03/2023]
Abstract
Alterations in the PI3K/AKT pathway occur in up to 70% of melanomas and are associated with disease progression. The three AKT paralogs are highly conserved but data suggest they have distinct functions. Activating mutations of AKT1 and AKT3 occur in human melanoma but their role in melanoma formation and metastasis remains unclear. Using an established melanoma mouse model, we evaluated E17K, E40K, and Q79K mutations in AKT1, AKT2, and AKT3 and show that mice harboring tumors expressing AKT1E17K had the highest incidence of brain metastasis and lowest mean survival. Tumors expressing AKT1E17K displayed elevated levels of focal adhesion factors and enhanced phosphorylation of focal adhesion kinase (FAK). AKT1E17K expression in melanoma cells increased invasion and this was reduced by pharmacologic inhibition of either AKT or FAK. These data suggest that the different AKT paralogs have distinct roles in melanoma brain metastasis and that AKT and FAK may be promising therapeutic targets. IMPLICATIONS: This study suggests that AKT1E17K promotes melanoma brain metastasis through activation of FAK and provides a rationale for the therapeutic targeting of AKT and/or FAK to reduce melanoma metastasis.
Collapse
Affiliation(s)
- David A Kircher
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Kirby A Trombetti
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Mark R Silvis
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Gennie L Parkman
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Grant M Fischer
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stephanie N Angel
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Christopher M Stehn
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Sean C Strain
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Allie H Grossmann
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- ARUP Institute for Clinical and Experimental Pathology, Salt Lake City, Utah
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Keith L Duffy
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Kenneth M Boucher
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Martin McMahon
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Dermatology, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Michael A Davies
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michelle C Mendoza
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Matthew W VanBrocklin
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah.
- Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah
- Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah
| |
Collapse
|
65
|
Pancreatic ductal adenocarcinoma: biological hallmarks, current status, and future perspectives of combined modality treatment approaches. Radiat Oncol 2019; 14:141. [PMID: 31395068 PMCID: PMC6688256 DOI: 10.1186/s13014-019-1345-6] [Citation(s) in RCA: 251] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/24/2019] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly devastating disease with poor prognosis and rising incidence. Late detection and a particularly aggressive biology are the major challenges which determine therapeutic failure. In this review, we present the current status and the recent advances in PDAC treatment together with the biological and immunological hallmarks of this cancer entity. On this basis, we discuss new concepts combining distinct treatment modalities in order to improve therapeutic efficacy and clinical outcome - with a specific focus on protocols involving radio(chemo)therapeutic approaches.
Collapse
|
66
|
Wang L, Ai M, Yu J, Jin L, Wang C, Liu Z, Shu X, Tang Z, Liu K, Luo H, Guan W, Sun X, Ma X. Structure-based modification of carbonyl-diphenylpyrimidines (Car-DPPYs) as a novel focal adhesion kinase (FAK) inhibitor against various stubborn cancer cells. Eur J Med Chem 2019; 172:154-162. [PMID: 30978560 DOI: 10.1016/j.ejmech.2019.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 12/15/2022]
Abstract
A family of carbonyl-substituted diphenylpyrimidine derivatives (Car-DPPYs) with strong activity against focal adhesion kinase (FAK), were described in this manuscript. Among them, compounds 7a (IC50 = 5.17 nM) and 7f (IC50 = 2.58 nM) displayed equal anti-FAK enzymatic activity to the lead compound TAE226 (6.79 nM). In particular, compound 7a also exhibited strong antiproliferative activity against several stubborn cancer cells, including AsPC-1 cells (IC50 = 0.105 μM), BxPC-3 cells (IC50 = 0.090 μM), and MCF-7/ADR cells (IC50 = 0.59 μM). Additionally, compound 7a also showed great antitumor efficacy in vivo via aAsPC-1 cancer Xenograft mouse model. The preliminary mechanism study by Western blot analysis revealed that 7a repressed FAK phosphorylation in AsPC cancer cells. Taken together, the results indicate that compound 7a may serve as a promising preclinical candidate for treatment of stubborn cancers.
Collapse
Affiliation(s)
- Luhong Wang
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China; College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Min Ai
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Jiawen Yu
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China
| | - Lingling Jin
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Zhihao Liu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Xiaohong Shu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Zeyao Tang
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Kexin Liu
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China
| | - Hui Luo
- Liaoning Ben-healthy Natural Technology and Dalian Buyun Biotechnology Co., Ltds., 116085, PR China
| | - Wenshun Guan
- Liaoning Ben-healthy Natural Technology and Dalian Buyun Biotechnology Co., Ltds., 116085, PR China
| | - Xiuli Sun
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, PR China.
| | - Xiaodong Ma
- College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, PR China.
| |
Collapse
|
67
|
Xie ZB, Zhang YF, Jin C, Mao YS, Fu DL. LRG-1 promotes pancreatic cancer growth and metastasis via modulation of the EGFR/p38 signaling. J Exp Clin Cancer Res 2019; 38:75. [PMID: 30760292 PMCID: PMC6374912 DOI: 10.1186/s13046-019-1088-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/06/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The abnormal expression of leucine-rich-alpha-2-glycoprotein 1 (LRG-1) is reported to be associated with multiple malignancies, but its role in the progression of pancreatic ductal adenocarcinoma (PDAC) remains to be determined. METHODS The expression of LRG-1 was assessed in PDAC tissues by RT-PCR, Western blot and immunohistochemistry. LRG-1-silenced or overexpressed cell lines were constructed using shRNA or LRG-1-overexpressing plasmids. EdU incorporation assay, Transwell invasion and wound-healing assays were performed to evaluate the proliferation, invasion and migration of PDAC cells. In addition, protein expression of the mitogen-activated protein kinase (MAPK) pathway was detected using Western blot. Finally, Co-immunoprecipitation assay was conducted in search of the potential interaction between LRG-1 and epidermal growth factor receptor (EGFR). RESULTS The expression of LRG-1 in PDAC tissue was significantly higher than that in adjacent normal tissue, and high LRG-1 expression predicted poor survival and a late tumor stage. In addition, LRG-1 markedly promoted the viability, proliferation, migration and invasion of PDAC cells in vitro and facilitated tumor growth in vivo. More importantly, we revealed that these bioactivities of LRG-1 might result from its selective interaction with EGFR, which might further activate the p38/MAPK signaling pathways. CONCLUSION LRG-1 may prove to be a promising biomarker for predicting prognosis of PDAC patients. Inhibition of LRG-1 or its downstream pathway could be a potential therapeutic target for the treatment of PDAC.
Collapse
Affiliation(s)
- Zhi-Bo Xie
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Central Urumqi Road, Shanghai, 200040 China
| | - Yi-Fan Zhang
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, 639 ZhizaojuRoad, Shanghai, 200011 China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Central Urumqi Road, Shanghai, 200040 China
| | - Yi-Shen Mao
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Central Urumqi Road, Shanghai, 200040 China
| | - De-Liang Fu
- Department of Pancreatic Surgery, Huashan Hospital, Fudan University, 12 Central Urumqi Road, Shanghai, 200040 China
| |
Collapse
|
68
|
Laszlo V, Valko Z, Ozsvar J, Kovacs I, Garay T, Hoda MA, Klikovits T, Stockhammer P, Aigner C, Gröger M, Klepetko W, Berger W, Grusch M, Tovari J, Waizenegger IC, Dome B, Hegedus B. The FAK inhibitor BI 853520 inhibits spheroid formation and orthotopic tumor growth in malignant pleural mesothelioma. J Mol Med (Berl) 2019; 97:231-242. [PMID: 30539198 PMCID: PMC6348072 DOI: 10.1007/s00109-018-1725-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023]
Abstract
No tyrosine kinase inhibitors are approved for malignant pleural mesothelioma (MPM). Preclinical studies identified focal adhesion kinase (FAK) as a target in MPM. Accordingly, we assessed the novel, highly selective FAK inhibitor (BI 853520) in 2D and 3D cultures and in vivo. IC50 values were measured by adherent cell viability assay. Cell migration and 3D growth were quantified by video microscopy and spheroid formation, respectively. Phosphorylation of FAK, Akt, S6, and Erk was measured by immunoblot. The mRNA expression of the putative tumor stem cell markers SOX2, Nanog, CD44, ALDH1, c-myc, and Oct4 was analyzed by qPCR. Cell proliferation, apoptosis, and tumor tissue microvessel density (MVD) were investigated in orthotopic MPM xenografts. In all 12 MPM cell lines, IC50 exceeded 5 μM and loss of NF2 did not correlate with sensitivity. No synergism was found with cisplatin in adherent cells. BI 853520 decreased migration in 3 out of 4 cell lines. FAK phosphorylation was reduced upon treatment but activation of Erk, Akt, or S6 remained unaffected. Nevertheless, BI 853520 inhibited spheroid growth and significantly reduced tumor weight, cell proliferation, and MVD in vivo. BI 853520 has limited effect in adherent cultures but demonstrates potent activity in spheroids and in orthotopic tumors in vivo. Based on our findings, further studies are warranted to explore the clinical utility of BI 853520 in human MPM. KEY MESSAGES: Response to FAK inhibition in MPM is independent of NF2 expression or histotype. FAK inhibition strongly interfered with MPM spheroid formation. BI 853520 has been shown to exert anti-tumor effect in MPM.
Collapse
Affiliation(s)
- Viktoria Laszlo
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Valko
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Judit Ozsvar
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Ildiko Kovacs
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Tamas Garay
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Mir Alireza Hoda
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Thomas Klikovits
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Paul Stockhammer
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, University Duisburg-Essen, Tüschener Weg 40, 45239, Essen, Germany
| | - Clemens Aigner
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, University Duisburg-Essen, Tüschener Weg 40, 45239, Essen, Germany
| | - Marion Gröger
- Core Facility Imaging, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Jozsef Tovari
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
- KINETO Lab Ltd, Budapest, Hungary
| | | | - Balazs Dome
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary.
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, Budapest, Hungary.
| | - Balazs Hegedus
- Division of Thoracic Surgery, Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria.
- 2nd Department of Pathology, Semmelweis University, Budapest, Hungary.
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, University Duisburg-Essen, Tüschener Weg 40, 45239, Essen, Germany.
| |
Collapse
|
69
|
Parkin A, Man J, Chou A, Nagrial AM, Samra J, Gill AJ, Timpson P, Pajic M. The Evolving Understanding of the Molecular and Therapeutic Landscape of Pancreatic Ductal Adenocarcinoma. Diseases 2018; 6:diseases6040103. [PMID: 30428574 PMCID: PMC6313363 DOI: 10.3390/diseases6040103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is the third leading cause of cancer-related deaths, characterised by poor survival, marked molecular heterogeneity and high intrinsic and acquired chemoresistance. Only 10⁻20% of pancreatic cancer patients present with surgically resectable disease and even then, 80% die within 5 years. Our increasing understanding of the genomic heterogeneity of cancer suggests that the failure of definitive clinical trials to demonstrate efficacy in the majority of cases is likely due to the low proportion of responsive molecular subtypes. As a consequence, novel treatment strategies to approach this disease are urgently needed. Significant developments in the field of precision oncology have led to increasing molecular stratification of cancers into subtypes, where individual cancers are selected for optimal therapy depending on their molecular or genomic fingerprint. This review provides an overview of the current status of clinically used and emerging treatment strategies, and discusses the advances in and the potential for the implementation of precision medicine in this highly lethal malignancy, for which there are currently no curative systemic therapies.
Collapse
Affiliation(s)
- Ashleigh Parkin
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Jennifer Man
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Angela Chou
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- University of Sydney, Sydney, NSW 2006, Australia.
| | - Adnan M Nagrial
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW 2145, Australia.
| | - Jaswinder Samra
- Department of Surgery, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia.
| | - Anthony J Gill
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- University of Sydney, Sydney, NSW 2006, Australia.
- Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia.
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW 2065, Australia.
| | - Paul Timpson
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia.
| | - Marina Pajic
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of NSW, Sydney, NSW 2010, Australia.
| |
Collapse
|
70
|
Fan S, Liang Z, Gao Z, Pan Z, Han S, Liu X, Zhao C, Yang W, Pan Z, Feng W. Identification of the key genes and pathways in prostate cancer. Oncol Lett 2018; 16:6663-6669. [PMID: 30405806 PMCID: PMC6202544 DOI: 10.3892/ol.2018.9491] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in men globally. The aim of the present study was to identify the key genes and pathways involved in the occurrence of PCa. Gene expression profile (GSE55945) was downloaded from Gene Expression Omnibus, and the differentially expressed genes (DEGs) were identified. Subsequently, Gene ontology analysis, KEGG pathway analysis and protein-protein interaction (PPI) analysis of DEGs were performed. Finally, the identified key genes were confirmed by immunohistochemistry. The GO analysis results showed that the DEGs were mainly participated in cell cycle, cell division, cell development and cell junction. The KEGG pathway analysis showed that the DEGs were mainly enriched in proteoglycans in cancer, endocytosis, focal adhesion and hippo signaling pathway. The PPI analysis results showed that RPS21, FOXO1, BIRC5, POLR2H, RPL22L1 and NPM1 were the key genes involved in the occurrence of PCa, and the Module analysis indicated that the occurrence of PCa was associated with cell cycle, oocyte meiosis and ribosome biogenesis. IHC result showed that the expression of RPS21, BIRC5, POLR2H, RPL22L1 and NPM1 were significantly upregulated in PCa, while the expression of FOXO1 was significantly downregulated in PCa, matching with the bioinformatics analysis. Taken together, several key genes and pathways were identified involved in PCa, which might provide the potential biomarker for prognosis, diagnosis and drug targets.
Collapse
Affiliation(s)
- Shutong Fan
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zumu Liang
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiqin Gao
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiwei Pan
- Department of Internal Medicine, Laizhou Development Zone Hospital, Yantai, Shandong 261400, P.R. China
| | - Shaojie Han
- Animal Epidemic Prevention and Epidemic Control Center, Changle County Bureau of Animal Health and Production, Weifang, Shandong 262400, P.R. China
| | - Xiaoying Liu
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Chunling Zhao
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Weiwei Yang
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhifang Pan
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Weiguo Feng
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
71
|
Lu P, Chen J, Yan L, Yang L, Zhang L, Dai J, Hao Z, Bai T, Xi Y, Li Y, Kang Z, Xv J, Sun G, Yang T. RasGRF2 promotes migration and invasion of colorectal cancer cells by modulating expression of MMP9 through Src/Akt/NF-κB pathway. Cancer Biol Ther 2018; 20:435-443. [PMID: 30359168 DOI: 10.1080/15384047.2018.1529117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Ras-specific guanine nucleotide-releasing factor 2 (RasGRF2) is a member of the guanine nucleotide exchange factors family which is expressed in a variety of tissues and cancer. However, the role of RasGRF2 in cancer is less reported, especially in colorectal cancer(CRC). Hence, the present study aimed to investigated the function of RasGRF2 and ways in which it affects tumor progression in CRC samples and cell lines. We first measured RasGRF2 mRNA level in 26 paired tumor and nontumor colon tissues after colon cancer surgical resection, and determined RasGRF2 protein level in 97 paired paraffin-embedded colon cancer tissues, and found that levels of RasGRF2 mRNA and protein were increased in colorectal tumor tissues, compared with adjacent non-tumor tissues. We then examined the effects of RasGRF2 knockdown on proliferation, migration and invasion were analyzed in CRC cells (SW480, HCT116 and LS174T). HCT116 cells with RasGRF2 knockdown were injected into the tail vein in nude mice to yield metastatic model, and tumor metastasis was measured as well. We found that knockdown of RasGRF2 in CRC cells reduced their migration and invasion in vitro and metastasis in mice. Furthermore, we explored the underlying molecular mechanism for RasGRF2-mediated CRC migration and invasion. The results showed that knockdown of RasGRF2 in CRC cells impairing the expression of MMP9 and inhibiting the activation of Src/Akt and NF-κB signaling. We conclude that RasGRF2 plays a role in controlling migration and invasion of CRC and modulates the expression of MMP9 through Src/PI 3-kinase and the NF-κB pathways.
Collapse
Affiliation(s)
- Peifen Lu
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Junjun Chen
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Lihong Yan
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Lijun Yang
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Litao Zhang
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Jie Dai
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Zixuan Hao
- b Department of Optometry , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Tao Bai
- c Department of Pathology , First Affiliated Hospital of Shanxi Medical University , Taiyuan , Shanxi , China
| | - Yanfeng Xi
- d Department of Pathology , Shanxi Provincial Cancer Hospital , Taiyuan , Shanxi , China
| | - Yahui Li
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Zhiming Kang
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| | - Jun Xv
- e Department of General Surgery , Second Hospital of Shanxi Medical University , Taiyuan , Shanxi , China
| | - Gongqin Sun
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China.,f Department of Cell and Molecular Biology , University of Rhode Island , Kingston , RI , USA
| | - Tao Yang
- a Department of Biochemistry & Molecular Biology , Shanxi Medical University , Taiyuan , Shanxi , China
| |
Collapse
|
72
|
Li ZY, Sun XY. Molecular targets regulating invasion and metastasis of pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2018; 26:1651-1659. [DOI: 10.11569/wcjd.v26.i28.1651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most malignant tumors of the digestive system. Invasion and metastasis are important biological characteristics of pancreatic cancer and contribute greatly to the poor prognosis of the patients. Many lines of evidence have recently revealed that many molecules, genes and proteins regulate the invasion and metastasis of pancreatic cancer cells. Therefore, exploration and a deep understanding of the molecular mechanism accounting for the invasion and metastasis of pancreatic cancer can help find novel pancreatic cancer biomarkers, improve early diagnosis, develop novel and effective treatment strategies, and predict the prognosis. This review summarizes the latest progress in the research of molecular targets for pancreatic cancer and the mechanisms by which they participate in the invasion and metastasis of this aggressive malignancy.
Collapse
Affiliation(s)
- Zi-Yi Li
- The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Xue-Ying Sun
- The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
73
|
Kaushik G, Ponnusamy MP, Batra SK. Concise Review: Current Status of Three-Dimensional Organoids as Preclinical Models. Stem Cells 2018; 36:1329-1340. [PMID: 29770526 DOI: 10.1002/stem.2852] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/10/2018] [Accepted: 05/01/2018] [Indexed: 12/15/2022]
Abstract
Three-dimensional (3D) cultures use the property of some cells to self-organize in matrices and generate structures that can be programmed to represent an organ or a pathology. Organoid cultures are the 3D cultivation of source tissue (ranging from cells to tissue fragments) in a support matrix and specialized media that nearly resembles the physiological environment. Depending on the source tissue, growth factors, and inhibitors provided, organoids can be programmed to recapitulate the biology of a system and progression of pathology. Organoids are genetically stable, and genetically amenable, making them very suitable tools to study tissue homeostasis and cancer. In this Review, we focus on providing recent technical advances from published literature to efficiently use organoids as a tool for disease modeling and therapeutics. Also, we discuss stem cell biology principles used to generate multiple organoids and their characteristics, with a brief description of methodology. A major theme of this review is to expand organoid applications to the study disease progression and drug response in different cancers. We also discuss shortcomings, limitations, and advantages of developed 3D cultures, with the rationale behind the methodology. Stem Cells 2018;36:1329-1340.
Collapse
Affiliation(s)
- Garima Kaushik
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Omaha, Nebraska, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, Omaha, Nebraska, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
74
|
Proline-Rich Protein Tyrosine Kinase 2 in Inflammation and Cancer. Cancers (Basel) 2018; 10:cancers10050139. [PMID: 29738483 PMCID: PMC5977112 DOI: 10.3390/cancers10050139] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 01/13/2023] Open
Abstract
Focal adhesion kinase (FAK) and its homologous FAK-related proline-rich tyrosine kinase 2 (Pyk2) contain the same domain, exhibit high sequence homology and are defined as a distinct family of non-receptor tyrosine kinases. This group of kinases plays critical roles in cytoskeletal dynamics and cell adhesion by regulating survival and growth signaling. This review summarizes the physiological and pathological functions of Pyk2 in inflammation and cancers. In particular, overexpression of Pyk2 in cancerous tissues is correlated with poor outcomes. Pyk2 stimulates multiple oncogenic signaling pathways, such as Wnt/β-catenin, PI3K/Akt, MAPK/ERK, and TGF-β/EGFR/VEGF, and facilitates carcinogenesis, migration, invasion, epithelial⁻mesenchymal transition and metastasis. Therefore, Pyk2 is a high-value therapeutic target and has clinical significance.
Collapse
|