1
|
Pei G, Min J, Rajapakshe KI, Branchi V, Liu Y, Selvanesan BC, Thege F, Sadeghian D, Zhang D, Cho KS, Chu Y, Dai E, Han G, Li M, Yee C, Takahashi K, Garg B, Tiriac H, Bernard V, Semaan A, Grem JL, Caffrey TC, Burks JK, Lowy AM, Aguirre AJ, Grandgenett PM, Hollingsworth MA, Guerrero PA, Wang L, Maitra A. Spatial mapping of transcriptomic plasticity in metastatic pancreatic cancer. Nature 2025:10.1038/s41586-025-08927-x. [PMID: 40269162 DOI: 10.1038/s41586-025-08927-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/20/2025] [Indexed: 04/25/2025]
Abstract
Patients with treatment-refractory pancreatic cancer often succumb to systemic metastases1-3; however, the transcriptomic heterogeneity that underlies therapeutic recalcitrance remains understudied, particularly in a spatial context. Here we construct high-resolution maps of lineage states, clonal architecture and the tumour microenvironment (TME) using spatially resolved transcriptomics from 55 samples of primary tumour and metastases (liver, lung and peritoneum) collected from rapid autopsies of 13 people. We observe discernible transcriptomic shifts in cancer-cell lineage states as tumours transition from primary sites to organ-specific metastases, with the most pronounced intra-patient distinctions between liver and lung. Phylogenetic trees constructed from inferred copy number variations in primary and metastatic loci in each patient highlight diverse patient-specific evolutionary trajectories and clonal dissemination. We show that multiple tumour lineage states co-exist in each tissue, including concurrent metastatic foci in the same organ. Agnostic to tissue site, lineage states correlate with distinct TME features, such as the spatial proximity of TGFB1-expressing myofibroblastic cancer-associated fibroblasts (myCAFs) to aggressive 'basal-like' cancer cells, but not to cells in the 'classical' or 'intermediate' states. These findings were validated through orthogonal and cross-species analyses using mouse tissues and patient-derived organoids. Notably, basal-like cancer cells aligned with myCAFs correlate with plasma-cell exclusion from the tumour milieu, and neighbouring cell analyses suggest that CXCR4-CXCL12 signalling is the underlying basis for observed immune exclusion. Collectively, our findings underscore the profound transcriptomic heterogeneity and microenvironmental dynamics that characterize treatment-refractory pancreatic cancer.
Collapse
Affiliation(s)
- Guangsheng Pei
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jimin Min
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kimal I Rajapakshe
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Branchi
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yunhe Liu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Benson Chellakkan Selvanesan
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fredrik Thege
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dorsay Sadeghian
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daiwei Zhang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Kyung Serk Cho
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enyu Dai
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guangchun Han
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cassian Yee
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kazuki Takahashi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Bharti Garg
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Herve Tiriac
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Vincent Bernard
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Semaan
- Department of Surgery, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jean L Grem
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Thomas C Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jared K Burks
- Department of Leukemia and Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Andrew J Aguirre
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Paul M Grandgenett
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Paola A Guerrero
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
- James P. Allison Institute, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute for Data Science in Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Pirson S, Gautier-Isola M, Baudin L, Rouaud L, Vanwynsberghe A, Deroye J, Bekisz S, Gucciardo F, Lebeau A, Buntinx F, Ivanova E, Staumont B, Blacher S, Gilles C, Noël A. AXL promotes lymphangiogenesis by amplifying VEGF-C-mediated AKT pathway. Cell Mol Life Sci 2025; 82:95. [PMID: 40011241 PMCID: PMC11865408 DOI: 10.1007/s00018-024-05542-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/23/2024] [Accepted: 12/06/2024] [Indexed: 02/28/2025]
Abstract
Lymphangiogenesis has gained considerable interest due to its established role in cancer progression and dissemination of metastatic cells through lymph nodes. Deciphering the molecular mechanisms that govern lymphangiogenesis within lymph nodes holds promise for revealing novel targetable molecules and pathways to inhibit metastasis. In this study, we revealed a previously unrecognized role of AXL, a tyrosine kinase receptor, in the lymphatic vessel formation. We first validated the expression of AXL in lymphatic endothelial cells (LECs), followed by functional studies using RNA interference and pharmacological inhibition with R428/Bemcentinib. These approaches provided compelling evidence that AXL promotes LEC migration in both 2D and 3D culture systems. Our findings demonstrated that AXL activation was induced by VEGF-C (Vascular Endothelial Growth Factor C) and further amplified downstream signaling via the AKT pathway. In vivo, the role of AXL in lymphatic vessel sprouting was demonstrated using R428 in a model of VEGF-C-induced lymphangiogenesis in lymph nodes. Interestingly, we discovered that AXL was predominantly expressed in MARCO+ LECs. Strikingly, under metastatic conditions, there was a notable increase in the density and penetration extent of these AXL-expressing LECs into the lymph node parenchyma. Collectively, our findings pinpoint AXL as a potent enhancer of lymphangiogenesis operating through the VEGF-C/AKT pathway. Furthermore, the identification of AXL expression within a distinct LEC subpopulation, particularly in the context of metastasis, underscores the intricate interplay between AXL signaling and lymphatic dynamics within the lymph node microenvironment.
Collapse
Affiliation(s)
- Sébastien Pirson
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Marine Gautier-Isola
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Louis Baudin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Loïc Rouaud
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Aline Vanwynsberghe
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Jonathan Deroye
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Sophie Bekisz
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
- Biomechanics Research Unit, GIGA In Silico Medicine, ULiège, Liège, Belgium
| | - Fabrice Gucciardo
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Alizée Lebeau
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Florence Buntinx
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Elitsa Ivanova
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Bernard Staumont
- Biomechanics Research Unit, GIGA In Silico Medicine, ULiège, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium.
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Tour de Pathologie, B23, +4, Avenue Hippocrate, 13, Liège, 4000, Belgium.
| |
Collapse
|
3
|
Rao R, Gulfishan M, Kim MS, Kashyap MK. Deciphering Cancer Complexity: Integrative Proteogenomics and Proteomics Approaches for Biomarker Discovery. Methods Mol Biol 2025; 2859:211-237. [PMID: 39436604 DOI: 10.1007/978-1-0716-4152-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Proteomics has revolutionized the field of cancer biology because the use of a large number of in vivo (SILAC), in vitro (iTRAQ, ICAT, TMT, stable-isotope Dimethyl, and 18O) labeling techniques or label-free methods (spectral counting or peak intensities) coupled with mass spectrometry enables us to profile and identify dysregulated proteins in diseases such as cancer. These proteome and genome studies have led to many challenges, such as the lack of consistency or correlation between copy numbers, RNA, and protein-level data. This review covers solely mass spectrometry-based approaches used for cancer biomarker discovery. It also touches on the emerging role of oncoproteogenomics or proteogenomics in cancer biomarker discovery and how this new area is attracting the integration of genomics and proteomics areas to address some of the important questions to help impinge on the biology and pathophysiology of different malignancies to make these mass spectrometry-based studies more realistic and relevant to clinical settings.
Collapse
Affiliation(s)
- Rashmi Rao
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Mohd Gulfishan
- School of Life and Allied Health Sciences, Glocal University, Saharanpur, UP, India
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu-42988, Republic of Korea
| | - Manoj Kumar Kashyap
- Amity Stem Cell Institute (ASCI), Amity Medical School (AMS), Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
4
|
Wang X, Yang J, Ren B, Yang G, Liu X, Xiao R, Ren J, Zhou F, You L, Zhao Y. Comprehensive multi-omics profiling identifies novel molecular subtypes of pancreatic ductal adenocarcinoma. Genes Dis 2024; 11:101143. [PMID: 39253579 PMCID: PMC11382047 DOI: 10.1016/j.gendis.2023.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/04/2023] [Accepted: 09/10/2023] [Indexed: 09/11/2024] Open
Abstract
Pancreatic cancer, a highly fatal malignancy, is predicted to rank as the second leading cause of cancer-related death in the next decade. This highlights the urgent need for new insights into personalized diagnosis and treatment. Although molecular subtypes of pancreatic cancer were well established in genomics and transcriptomics, few known molecular classifications are translated to guide clinical strategies and require a paradigm shift. Notably, chronically developing and continuously improving high-throughput technologies and systems serve as an important driving force to further portray the molecular landscape of pancreatic cancer in terms of epigenomics, proteomics, metabonomics, and metagenomics. Therefore, a more comprehensive understanding of molecular classifications at multiple levels using an integrated multi-omics approach holds great promise to exploit more potential therapeutic options. In this review, we recapitulated the molecular spectrum from different omics levels, discussed various subtypes on multi-omics means to move one step forward towards bench-to-beside translation of pancreatic cancer with clinical impact, and proposed some methodological and scientific challenges in store.
Collapse
Affiliation(s)
- Xing Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Jinshou Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Bo Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Xiaohong Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Ruiling Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Jie Ren
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Feihan Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, China
- Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, China
- National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, China
| |
Collapse
|
5
|
DeRyckere D, Huelse JM, Earp HS, Graham DK. TAM family kinases as therapeutic targets at the interface of cancer and immunity. Nat Rev Clin Oncol 2023; 20:755-779. [PMID: 37667010 DOI: 10.1038/s41571-023-00813-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 09/06/2023]
Abstract
Novel treatment approaches are needed to overcome innate and acquired mechanisms of resistance to current anticancer therapies in cancer cells and the tumour immune microenvironment. The TAM (TYRO3, AXL and MERTK) family receptor tyrosine kinases (RTKs) are potential therapeutic targets in a wide range of cancers. In cancer cells, TAM RTKs activate signalling pathways that promote cell survival, metastasis and resistance to a variety of chemotherapeutic agents and targeted therapies. TAM RTKs also function in innate immune cells, contributing to various mechanisms that suppress antitumour immunity and promote resistance to immune-checkpoint inhibitors. Therefore, TAM antagonists provide an unprecedented opportunity for both direct and immune-mediated therapeutic activity provided by inhibition of a single target, and are likely to be particularly effective when used in combination with other cancer therapies. To exploit this potential, a variety of agents have been designed to selectively target TAM RTKs, many of which have now entered clinical testing. This Review provides an essential guide to the TAM RTKs for clinicians, including an overview of the rationale for therapeutic targeting of TAM RTKs in cancer cells and the tumour immune microenvironment, a description of the current preclinical and clinical experience with TAM inhibitors, and a perspective on strategies for continued development of TAM-targeted agents for oncology applications.
Collapse
Affiliation(s)
- Deborah DeRyckere
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Justus M Huelse
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - H Shelton Earp
- Department of Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Douglas K Graham
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA.
- Department of Paediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
6
|
MALDI-MSI: A Powerful Approach to Understand Primary Pancreatic Ductal Adenocarcinoma and Metastases. Molecules 2022; 27:molecules27154811. [PMID: 35956764 PMCID: PMC9369872 DOI: 10.3390/molecules27154811] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer-related deaths are very commonly attributed to complications from metastases to neighboring as well as distant organs. Dissociate response in the treatment of pancreatic adenocarcinoma is one of the main causes of low treatment success and low survival rates. This behavior could not be explained by transcriptomics or genomics; however, differences in the composition at the protein level could be observed. We have characterized the proteomic composition of primary pancreatic adenocarcinoma and distant metastasis directly in human tissue samples, utilizing mass spectrometry imaging. The mass spectrometry data was used to train and validate machine learning models that could distinguish both tissue entities with an accuracy above 90%. Model validation on samples from another collection yielded a correct classification of both entities. Tentative identification of the discriminative molecular features showed that collagen fragments (COL1A1, COL1A2, and COL3A1) play a fundamental role in tumor development. From the analysis of the receiver operating characteristic, we could further advance some potential targets, such as histone and histone variations, that could provide a better understanding of tumor development, and consequently, more effective treatments.
Collapse
|
7
|
Lautenbacher L, Samaras P, Muller J, Grafberger A, Shraideh M, Rank J, Fuchs ST, Schmidt TK, The M, Dallago C, Wittges H, Rost B, Krcmar H, Kuster B, Wilhelm M. ProteomicsDB: toward a FAIR open-source resource for life-science research. Nucleic Acids Res 2022; 50:D1541-D1552. [PMID: 34791421 PMCID: PMC8728203 DOI: 10.1093/nar/gkab1026] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 12/28/2022] Open
Abstract
ProteomicsDB (https://www.ProteomicsDB.org) is a multi-omics and multi-organism resource for life science research. In this update, we present our efforts to continuously develop and expand ProteomicsDB. The major focus over the last two years was improving the findability, accessibility, interoperability and reusability (FAIR) of the data as well as its implementation. For this purpose, we release a new application programming interface (API) that provides systematic access to essentially all data in ProteomicsDB. Second, we release a new open-source user interface (UI) and show the advantages the scientific community gains from such software. With the new interface, two new visualizations of protein primary, secondary and tertiary structure as well an updated spectrum viewer were added. Furthermore, we integrated ProteomicsDB with our deep-neural-network Prosit that can predict the fragmentation characteristics and retention time of peptides. The result is an automatic processing pipeline that can be used to reevaluate database search engine results stored in ProteomicsDB. In addition, we extended the data content with experiments investigating different human biology as well as a newly supported organism.
Collapse
Affiliation(s)
- Ludwig Lautenbacher
- Technical University of Munich, Computational Mass Spectrometry, 85354 Freising, Bavaria, Germany
| | - Patroklos Samaras
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
| | - Julian Muller
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
| | - Andreas Grafberger
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
| | - Marwin Shraideh
- Technical University of Munich, Chair for Information Systems, 85748 Garching, Bavaria, Germany
- Technical University of Munich, SAP University Competence Center, 85748 Garching, Bavaria, Germany
| | - Johannes Rank
- Technical University of Munich, Chair for Information Systems, 85748 Garching, Bavaria, Germany
- Technical University of Munich, SAP University Competence Center, 85748 Garching, Bavaria, Germany
| | - Simon T Fuchs
- Technical University of Munich, Chair for Information Systems, 85748 Garching, Bavaria, Germany
- Technical University of Munich, SAP University Competence Center, 85748 Garching, Bavaria, Germany
| | - Tobias K Schmidt
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
| | - Matthew The
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
| | - Christian Dallago
- Technical University of Munich, Department for Bioinformatics and Computational Biology, 85748 Garching, Bavaria, Germany
- Technical University of Munich, Center of Doctoral Studies in Informatics and its Applications (CeDoSIA), 85748 Garching, Bavaria, Germany
| | - Holger Wittges
- Technical University of Munich, Chair for Information Systems, 85748 Garching, Bavaria, Germany
- Technical University of Munich, SAP University Competence Center, 85748 Garching, Bavaria, Germany
| | - Burkhard Rost
- Technical University of Munich, Department for Bioinformatics and Computational Biology, 85748 Garching, Bavaria, Germany
- Technical University of Munich, Institute for Advanced Study (TUM-IAS), 85748 Freising, Bavaria, Germany
| | - Helmut Krcmar
- Technical University of Munich, Chair for Information Systems, 85748 Garching, Bavaria, Germany
- Technical University of Munich, SAP University Competence Center, 85748 Garching, Bavaria, Germany
| | - Bernhard Kuster
- Technical University of Munich, Chair of Proteomics and Bioanalytics, 85354 Freising, Bavaria, Germany
- Technical University of Munich, Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), 85354 Freising, Bavaria, Germany
| | - Mathias Wilhelm
- Technical University of Munich, Computational Mass Spectrometry, 85354 Freising, Bavaria, Germany
| |
Collapse
|
8
|
Dujardin P, Baginska AK, Urban S, Grüner BM. Unraveling Tumor Heterogeneity by Using DNA Barcoding Technologies to Develop Personalized Treatment Strategies in Advanced-Stage PDAC. Cancers (Basel) 2021; 13:4187. [PMID: 34439341 PMCID: PMC8394487 DOI: 10.3390/cancers13164187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/14/2022] Open
Abstract
Tumor heterogeneity is a hallmark of many solid tumors, including pancreatic ductal adenocarcinoma (PDAC), and an inherent consequence of the clonal evolution of cancers. As such, it is considered the underlying concept of many characteristics of the disease, including the ability to metastasize, adapt to different microenvironments, and to develop therapy resistance. Undoubtedly, the high mortality of PDAC can be attributed to a high extent to these properties. Despite its apparent importance, studying tumor heterogeneity has been a challenging task, mainly due to its complexity and lack of appropriate methods. However, in recent years molecular DNA barcoding has emerged as a sophisticated tool that allows mapping of individual cells or subpopulations in a cell pool to study heterogeneity and thus devise new personalized treatment strategies. In this review, we provide an overview of genetic and non-genetic inter- and intra-tumor heterogeneity and its impact on (personalized) treatment strategies in PDAC and address how DNA barcoding technologies work and can be applied to study this clinically highly relevant question.
Collapse
Affiliation(s)
- Philip Dujardin
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen at the University Duisburg-Essen, 45147 Essen, Germany
| | - Anna K Baginska
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen at the University Duisburg-Essen, 45147 Essen, Germany
| | - Sebastian Urban
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen at the University Duisburg-Essen, 45147 Essen, Germany
| | - Barbara M Grüner
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen at the University Duisburg-Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK) Partner Site Essen/Düsseldorf, 45147 Essen, Germany
| |
Collapse
|
9
|
Tyrosine Phosphoproteomics of Patient-Derived Xenografts Reveals Ephrin Type-B Receptor 4 Tyrosine Kinase as a Therapeutic Target in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13143404. [PMID: 34298619 PMCID: PMC8303779 DOI: 10.3390/cancers13143404] [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: 03/22/2021] [Revised: 06/04/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic cancer is one of the deadliest solid malignancies. Pancreatic ductal adenocarcinoma accounts for 90% of pancreatic cancer cases with minimal response to traditional chemotherapies. Protein tyrosine kinases have been shown to be hyperactivated in cancers and thus can serve as therapeutic targets. Patient-derived tumor xenografts (PDXs) in animal models such as mice are an appropriate resource to identify such activated kinases. PDXs models are excellent for the identification of therapeutic targets as compared to cell line models as they better reflect an in vivo environment. We identified ephrin type-B receptor 4 (EphB4) as hyperactivated in PDXs derived from pancreatic ductal adenocarcinoma. Abstract Pancreatic ductal adenocarcinoma is a recalcitrant tumor with minimal response to conventional chemotherapeutic approaches. Oncogenic signaling by activated tyrosine kinases has been implicated in cancers resulting in activation of diverse effector signaling pathways. Thus, the discovery of aberrantly activated tyrosine kinases is of great interest in developing novel therapeutic strategies in the treatment and management of pancreatic cancer. Patient-derived tumor xenografts (PDXs) in mice serve as potentially valuable preclinical models as they maintain the histological and molecular heterogeneity of the original human tumor. Here, we employed high-resolution mass spectrometry combined with immunoaffinity purification using anti-phosphotyrosine antibodies to profile tyrosine phosphoproteome across 13 pancreatic ductal adenocarcinoma PDX models. This analysis resulted in the identification of 1199 tyrosine-phosphorylated sites mapping to 704 proteins. The mass spectrometric analysis revealed widespread and heterogeneous activation of both receptor and non-receptor tyrosine kinases. Preclinical studies confirmed ephrin type-B receptor 4 (EphB4) as a potential therapeutic target based on the efficacy of human serum albumin-conjugated soluble EphB4 in mice bearing orthotopic xenografts. Immunohistochemistry-based validation using tissue microarrays from 346 patients with PDAC showed significant expression of EphB4 in >70% of patients. In summary, we present a comprehensive landscape of tyrosine phosphoproteome with EphB4 as a promising therapeutic target in pancreatic ductal adenocarcinoma.
Collapse
|
10
|
Le Large TYS, Bijlsma MF, El Hassouni B, Mantini G, Lagerweij T, Henneman AA, Funel N, Kok B, Pham TV, de Haas R, Morelli L, Knol JC, Piersma SR, Kazemier G, van Laarhoven HWM, Giovannetti E, Jimenez CR. Focal adhesion kinase inhibition synergizes with nab-paclitaxel to target pancreatic ductal adenocarcinoma. J Exp Clin Cancer Res 2021; 40:91. [PMID: 33750427 PMCID: PMC7941981 DOI: 10.1186/s13046-021-01892-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a very lethal disease, with minimal therapeutic options. Aberrant tyrosine kinase activity influences tumor growth and is regulated by phosphorylation. We investigated phosphorylated kinases as target in PDAC. METHODS Mass spectrometry-based phosphotyrosine proteomic analysis on PDAC cell lines was used to evaluate active kinases. Pathway analysis and inferred kinase activity analysis was performed to identify novel targets. Subsequently, we investigated targeting of focal adhesion kinase (FAK) in vitro with drug perturbations in combination with chemotherapeutics used against PDAC. Tyrosine phosphoproteomics upon treatment was performed to evaluate signaling. An orthotopic model of PDAC was used to evaluate the combination of defactinib with nab-paclitaxel. RESULTS PDAC cell lines portrayed high activity of multiple receptor tyrosine kinases to various degree. The non-receptor kinase, FAK, was identified in all cell lines by our phosphotyrosine proteomic screen and pathway analysis. Targeting of this kinase with defactinib validated reduced phosphorylation profiles. Additionally, FAK inhibition had anti-proliferative and anti-migratory effects. Combination with (nab-)paclitaxel had a synergistic effect on cell proliferation in vitro and reduced tumor growth in vivo. CONCLUSIONS Our study shows high phosphorylation of several oncogenic receptor tyrosine kinases in PDAC cells and validated FAK inhibition as potential synergistic target with Nab-paclitaxel against this devastating disease.
Collapse
Affiliation(s)
- T Y S Le Large
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - M F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - B El Hassouni
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - G Mantini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC-Start-Up, Fondazione Pisana per la Scienza, Pisa, Italy
| | - T Lagerweij
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, The Netherlands
| | - A A Henneman
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - N Funel
- Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - B Kok
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - T V Pham
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - R de Haas
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - L Morelli
- Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - J C Knol
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - S R Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - G Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, The Netherlands
| | - H W M van Laarhoven
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - E Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, AIRC-Start-Up, Fondazione Pisana per la Scienza, Pisa, Italy.
| | - C R Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, Cancer, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Singh K, Pruski M, Bland R, Younes M, Guha S, Thosani N, Maitra A, Cash BD, McAllister F, Logsdon CD, Chang JT, Bailey-Lundberg JM. Kras mutation rate precisely orchestrates ductal derived pancreatic intraepithelial neoplasia and pancreatic cancer. J Transl Med 2021; 101:177-192. [PMID: 33009500 PMCID: PMC8172380 DOI: 10.1038/s41374-020-00490-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related death in the United States. Despite the high prevalence of Kras mutations in pancreatic cancer patients, murine models expressing the oncogenic mutant Kras (Krasmut) in mature pancreatic cells develop PDAC at a low frequency. Independent of cell of origin, a second genetic hit (loss of tumor suppressor TP53 or PTEN) is important for development of PDAC in mice. We hypothesized ectopic expression and elevated levels of oncogenic mutant Kras would promote PanIN arising in pancreatic ducts. To test our hypothesis, the significance of elevating levels of K-Ras and Ras activity has been explored by expression of a CAG driven LGSL-KrasG12V allele (cKras) in pancreatic ducts, which promotes ectopic Kras expression. We predicted expression of cKras in pancreatic ducts would generate neoplasia and PDAC. To test our hypothesis, we employed tamoxifen dependent CreERT2 mediated recombination. Hnf1b:CreERT2;KrasG12V (cKrasHnf1b/+) mice received 1 (Low), 5 (Mod) or 10 (High) mg per 20 g body weight to recombine cKras in low (cKrasLow), moderate (cKrasMod), and high (cKrasHigh) percentages of pancreatic ducts. Our histologic analysis revealed poorly differentiated aggressive tumors in cKrasHigh mice. cKrasMod mice had grades of Pancreatic Intraepithelial Neoplasia (PanIN), recapitulating early and advanced PanIN observed in human PDAC. Proteomics analysis revealed significant differences in PTEN/AKT and MAPK pathways between wild type, cKrasLow, cKrasMod, and cKrasHigh mice. In conclusion, in this study, we provide evidence that ectopic expression of oncogenic mutant K-Ras in pancreatic ducts generates early and late PanIN as well as PDAC. This Ras rheostat model provides evidence that AKT signaling is an important early driver of invasive ductal derived PDAC.
Collapse
Affiliation(s)
- Kanchan Singh
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Melissa Pruski
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Rachael Bland
- Kings College London, Department of Pharmacology, London, UK
| | - Mamoun Younes
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Sushovan Guha
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Nirav Thosani
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Anirban Maitra
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brooks D Cash
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Florencia McAllister
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Craig D Logsdon
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Jennifer M Bailey-Lundberg
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA.
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Abyadeh M, Meyfour A, Gupta V, Zabet Moghaddam M, Fitzhenry MJ, Shahbazian S, Hosseini Salekdeh G, Mirzaei M. Recent Advances of Functional Proteomics in Gastrointestinal Cancers- a Path towards the Identification of Candidate Diagnostic, Prognostic, and Therapeutic Molecular Biomarkers. Int J Mol Sci 2020; 21:ijms21228532. [PMID: 33198323 PMCID: PMC7697099 DOI: 10.3390/ijms21228532] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/02/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) cancer remains one of the common causes of morbidity and mortality. A high number of cases are diagnosed at an advanced stage, leading to a poor survival rate. This is primarily attributed to the lack of reliable diagnostic biomarkers and limited treatment options. Therefore, more sensitive, specific biomarkers and curative treatments are desirable. Functional proteomics as a research area in the proteomic field aims to elucidate the biological function of unknown proteins and unravel the cellular mechanisms at the molecular level. Phosphoproteomic and glycoproteomic studies have emerged as two efficient functional proteomics approaches used to identify diagnostic biomarkers, therapeutic targets, the molecular basis of disease and mechanisms underlying drug resistance in GI cancers. In this review, we present an overview on how functional proteomics may contribute to the understanding of GI cancers, namely colorectal, gastric, hepatocellular carcinoma and pancreatic cancers. Moreover, we have summarized recent methodological developments in phosphoproteomics and glycoproteomics for GI cancer studies.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (M.A.); (G.H.S.)
| | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1985717413, Iran
- Cell Science Research Center, Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
- Correspondence: (A.M.); (M.M.)
| | - Vivek Gupta
- Department of Clinical Medicine, Macquarie University, Macquarie Park, NSW 2113, Australia;
| | | | - Matthew J. Fitzhenry
- Australian Proteome Analysis Facility, Macquarie University, Macquarie Park, NSW 2113, Australia;
| | - Shila Shahbazian
- Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2113, Australia;
| | - Ghasem Hosseini Salekdeh
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (M.A.); (G.H.S.)
- Department of Molecular Sciences, Macquarie University, Macquarie Park, NSW 2113, Australia;
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Macquarie University, Macquarie Park, NSW 2113, Australia;
- Correspondence: (A.M.); (M.M.)
| |
Collapse
|
13
|
von Itzstein MS, Burke MC, Brekken RA, Aguilera TA, Zeh HJ, Beg MS. Targeting TAM to Tame Pancreatic Cancer. Target Oncol 2020; 15:579-588. [PMID: 32996059 DOI: 10.1007/s11523-020-00751-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer is expected to become the second leading cause of cancer-related death within the next few years. Current therapeutic strategies have limited effectiveness and therefore there is an urgency to develop novel effective therapies. The receptor tyrosine kinase subfamily TAM (Tyro3, Axl, MerTK) is directly implicated in the pathogenesis of the metastatic, chemoresistant, and immunosuppressive phenotype in pancreatic cancer. TAM inhibitors are promising investigational therapies for pancreatic cancer due to their potential to target multiple aspects of pancreatic cancer biology. Specifically, recent mechanistic investigations and therapeutic combinations in the preclinical setting suggest that TAM inhibition with chemotherapy, targeted therapy, and immunotherapy should be evaluated clinically.
Collapse
Affiliation(s)
- Mitchell S von Itzstein
- Division of Hematology/Oncology, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8852, USA
- Division of Hematology and Medical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael C Burke
- Division of Hematology/Oncology, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8852, USA
- Division of Hematology and Medical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rolf A Brekken
- Division of Surgical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Todd A Aguilera
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Herbert J Zeh
- Division of Surgical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Muhammad Shaalan Beg
- Division of Hematology/Oncology, Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8852, USA.
- Division of Hematology and Medical Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
14
|
Armacki M, Polaschek S, Waldenmaier M, Morawe M, Ruhland C, Schmid R, Lechel A, Tharehalli U, Steup C, Bektas Y, Li H, Kraus JM, Kestler HA, Kruger S, Ormanns S, Walther P, Eiseler T, Seufferlein T. Protein Kinase D1, Reduced in Human Pancreatic Tumors, Increases Secretion of Small Extracellular Vesicles From Cancer Cells That Promote Metastasis to Lung in Mice. Gastroenterology 2020; 159:1019-1035.e22. [PMID: 32446697 DOI: 10.1053/j.gastro.2020.05.052] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Pancreatic tumor cells release small extracellular vesicles (sEVs, exosomes) that contain lipids and proteins, RNA, and DNA molecules that might promote formation of metastases. It is not clear what cargo these vesicles contain and how they are released. Protein kinase D1 (PRKD1) inhibits cell motility and is believed to be dysregulated in pancreatic ductal adenocarcinomas. We investigated whether it regulates production of sEVs in pancreatic cancer cells and their ability to form premetastatic niches for pancreatic cancer cells in mice. METHODS We analyzed data from UALCAN and human pancreatic tissue microarrays to compare levels of PRKD1 between tumor and nontumor tissues. We studied mice with pancreas-specific disruption of Prkd1 (PRKD1KO mice), mice that express oncogenic KRAS (KC mice), and KC mice with disruption of Prkd1 (PRKD1KO-KC mice). Subcutaneous xenograft tumors were grown in NSG mice from Panc1 cells; some mice were then given injections of sEVs. Pancreata and lung tissues from mice were analyzed by histology, immunohistochemistry, and/or quantitative polymerase chain reaction; we performed nanoparticle tracking analysis of plasma sEVs. The Prkd1 gene was disrupted in Panc1 cells using CRISPR-Cas9 or knocked down with small hairpin RNAs, or PRKD1 activity was inhibited with the selective inhibitor CRT0066101. Pancreatic cancer cell lines were analyzed by gene-expression microarray, quantitative polymerase chain reaction, immunoblot, and immunofluorescence analyses. sEVs secreted by Panc1 cell lines were analyzed by flow cytometry, transmission electron microscopy, and mass spectrometry. RESULTS Levels of PRKD1 were reduced in human pancreatic ductal adenocarcinoma tissues compared with nontumor tissues. PRKD1KO-KC mice developed more pancreatic intraepithelial neoplasia, at a faster rate, than KC mice, and had more lung metastases and significantly shorter average survival time. Serum from PRKD1KO-KC mice had increased levels of sEVs compared with KC mice. Pancreatic cancer cells with loss or inhibition of PRKD1 increased secretion of sEVs; loss of PRKD1 reduced phosphorylation of its substrate, cortactin, resulting in increased F-actin levels at the plasma membrane. sEVs from cells with loss or reduced expression of PRKD1 had altered content, and injection of these sEVs into mice increased metastasis of xenograft tumors to lung, compared with sEVs from pancreatic cells that expressed PRKD1. PRKD1-deficient pancreatic cancer cells showed increased loading of integrin α6β4 into sEVs-a process that required CD82. CONCLUSIONS Human pancreatic ductal adenocarcinoma has reduced levels of PRKD1 compared with nontumor pancreatic tissues. Loss of PRKD1 results in reduced phosphorylation of cortactin in pancreatic cancer cell lines, resulting in increased in F-actin at the plasma membrane and increased release of sEVs, with altered content. These sEVs promote metastasis of xenograft and pancreatic tumors to lung in mice.
Collapse
Affiliation(s)
- Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Sandra Polaschek
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | - Mareen Morawe
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Claudia Ruhland
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Rebecca Schmid
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - André Lechel
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Umesh Tharehalli
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Christoph Steup
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Yasin Bektas
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Hongxia Li
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Johann M Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Stephan Kruger
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Faculty of Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, University of Ulm, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany.
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
15
|
Peng J, Liu L, Huang D, Chen H, Dai M, Guo J, Zhang T, Liao Q, Jiang J, Wang W, Guo D, Cao D, Xuan Z, Li D, Zhao Y, Wu W. Impact of ischemia on sample quality of human pancreatic tissues. Pancreatology 2020; 20:265-277. [PMID: 31956070 DOI: 10.1016/j.pan.2019.11.005] [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] [Received: 06/21/2019] [Revised: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Successful clinical evaluation of human tumors relies on proper handling of tissue samples to maximally preserve the cellular and metabolic states in vivo. Pancreatic samples are particularly sensitive to sample mishandling due to the abundance of digestive enzymes. We study how the duration of ischemia, in vivo and ex vivo, both of which are unavoidable lagging periods following surgical dissection, significantly impact the utility of pancreatic samples. METHODS We systematically characterize a wide range of tissue integrity features, including histological patterns, cellular structures, DNA/RNA quality and activity of major signaling pathways in normal pancreases and pancreatic ductal adenocarcinoma (PDAC) tumor tissues from 41 patients with different ischemia. RESULTS We reveal that tissues experiencing longer periods of ischemia exhibit significant deterioration and could potentially mislead disease diagnosis and preclinical research. Based on these analyses, we propose an optimal procedure that balances better clinical practice and high tissue sample quality. CONCLUSIONS Our work provides a guideline for pancreatic sample handling and could have wide implications in clinical diagnosis and translational research.
Collapse
Affiliation(s)
- Junya Peng
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Lulu Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Dan Huang
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Hao Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Jialin Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Wenze Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Dan Guo
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China; Department of Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Dingyan Cao
- Department of Clinical Biobank, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Zhixuan Xuan
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Dongjing Li
- Department of Health Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China; Tsinghua University-Peking University Joint Center for Life Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China.
| | - Wenming Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China.
| |
Collapse
|
16
|
Zhao Y, Li J, Li D, Wang Z, Zhao J, Wu X, Sun Q, Lin PP, Plum P, Damanakis A, Gebauer F, Zhou M, Zhang Z, Schlösser H, Jauch KW, Nelson PJ, Bruns CJ. Tumor biology and multidisciplinary strategies of oligometastasis in gastrointestinal cancers. Semin Cancer Biol 2020; 60:334-343. [PMID: 31445220 DOI: 10.1016/j.semcancer.2019.08.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/20/2019] [Indexed: 12/11/2022]
Abstract
More than 70% of gastrointestinal (GI) cancers are diagnosed with metastases, leading to poor prognosis. For some cancer patients with limited sites of metastatic tumors, the term oligometastatic disease (OMD) has been coined as opposed to systemic polymetastasis (PMD) disease. Stephan Paget first described an organ-specific pattern of metastasis in 1889, now known as the "seed and soil" theory where distinct cancer types are found to metastasize to different tumor-specific sites. Our understanding of the biology of tumor metastasis and specifically the molecular mechanisms driving their formation are still limited, in particular, as it relates to the genesis of oligometastasis. In the following review, we discuss recent advances in general understanding of this metastatic behavior including the role of specific signaling pathways, various molecular features and biomarkers, as well as the interaction of carcinoma cells with their tissue microenvironments (both primary and metastatic niches). The unique features that underlie OMD provide potential targets for localized therapy. As it relates to clinical practice, OMD is emerging as treatable with surgical resection and/or other local therapy options. Strategies currently being applied in the clinical management of OMD will be discussed including surgical, radiation-based therapy, ablation procedures, and the results of emerging clinical trials involving immunotherapy.
Collapse
Affiliation(s)
- Yue Zhao
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Department of General, Visceral und Vascular Surgery, Otto von Guericke University, Magdeburg, Germany.
| | - Jiahui Li
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Dai Li
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Department of Anethesiology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Zhefang Wang
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Jiangang Zhao
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Department of General, Visceral und Vascular Surgery, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Xiaolin Wu
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Qiye Sun
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | | | - Patrick Plum
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Institute for Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexander Damanakis
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Menglong Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hans Schlösser
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Center for Integrated Oncology (CIO) Achen, Bonn, Cologne and Düsseldorf, Cologne, Germany
| | - Karl-Walter Jauch
- Department of General, Visceral und Vascular Surgery, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Peter J Nelson
- Department of Internal Medicine IV, University Hospital of Munich, Ludwig-Maximilians-University Munich, Germany
| | - Christiane J Bruns
- Department of General, Visceral und Tumor Surgery, University Hospital Cologne, Kerpener Straße 62, 50937, Cologne, Germany; Center for Integrated Oncology (CIO) Achen, Bonn, Cologne and Düsseldorf, Cologne, Germany.
| |
Collapse
|
17
|
Tao L, Su L, Yuan C, Ma Z, Zhang L, Bo S, Niu Y, Lu S, Xiu D. Postoperative metastasis prediction based on portal vein circulating tumor cells detected by flow cytometry in periampullary or pancreatic cancer. Cancer Manag Res 2019; 11:7405-7425. [PMID: 31496801 PMCID: PMC6689556 DOI: 10.2147/cmar.s210332] [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: 03/28/2019] [Accepted: 06/20/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose The aim of this study was to evaluate the value of flow cytometry (FCM) detection of portal vein circulating tumor cells (CTCs) in predicting postoperative metastasis. Methods Samples of portal venous blood and peripheral blood were collected from 39 patients during surgery, and CTCs were detected by FCM, with confirmation by laser confocal microscopy and single-cell sequencing. Results Among all patients, a portal EpCAM+CD45- percentage ≥24.5×10−4 (P=0.06), peripheral EpCAM+CD45- count ≥97/5 mL (P=0.034), peripheral EpCAM+CD45- percentage ≥4.4×10−4 (P=0.042), and CA242≥3.5 U/mL (P=0.027) were significant predictors of metastasis. Further analysis showed that the portal EpCAM+CD45- ratio ≥24.5×10−4 is a predictor of metastasis (P=0.025) in pancreatic cancer after curative resection. Conclusion CTCs detected by FCM in portal venous blood are of significant value for the prediction of postoperative metastasis in pancreatic or periampullary tumors.
Collapse
Affiliation(s)
- Lianyuan Tao
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, People's Republic of China.,Department of Hepatobiliary Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, People's Republic of China
| | - Li Su
- Medical and Health Analytical Center, Peking University Health Science Center, Beijing 100191, People's Republic of China
| | - Chunhui Yuan
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Zhaolai Ma
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Lingfu Zhang
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Shiping Bo
- Department of Clinical Research, Yikon Genomics Co. Ltd., Shanghai, People's Republic of China
| | - Yunyun Niu
- Department of Clinical Research, Yikon Genomics Co. Ltd., Shanghai, People's Republic of China
| | - Sijia Lu
- Department of Clinical Research, Yikon Genomics Co. Ltd., Shanghai, People's Republic of China
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing 100191, People's Republic of China
| |
Collapse
|
18
|
Zhang X, Nguyen KD, Rudnick PA, Roper N, Kawaler E, Maity TK, Awasthi S, Gao S, Biswas R, Venugopalan A, Cultraro CM, Fenyö D, Guha U. Quantitative Mass Spectrometry to Interrogate Proteomic Heterogeneity in Metastatic Lung Adenocarcinoma and Validate a Novel Somatic Mutation CDK12-G879V. Mol Cell Proteomics 2019; 18:622-641. [PMID: 30617155 PMCID: PMC6442362 DOI: 10.1074/mcp.ra118.001266] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in both men and women. Tumor heterogeneity is an impediment to targeted treatment of all cancers, including lung cancer. Here, we sought to characterize tumor proteome and phosphoproteome changes by longitudinal, prospective collection of tumor tissue from an exceptional responder lung adenocarcinoma patient who survived with metastatic lung adenocarcinoma for over seven years while undergoing HER2-directed therapy in combination with chemotherapy. We employed "Super-SILAC" and TMT labeling strategies to quantify the proteome and phosphoproteome of a lung metastatic site and eight distinct metastatic progressive lymph nodes collected during these seven years, including five lymph nodes procured at autopsy. We identified specific signaling networks enriched in lung compared with the lymph node metastatic sites. We correlated the changes in protein abundance with changes in copy number alteration (CNA) and transcript expression. ERBB2/HER2 protein expression was higher in lung, consistent with a higher degree of ERBB2 amplification in lung compared with the lymph node metastatic sites. To further interrogate the mass spectrometry data, a patient-specific database was built by incorporating all the somatic and germline variants identified by whole genome sequencing (WGS) of genomic DNA from the lung, one lymph node metastatic site and blood. An extensive validation pipeline was built to confirm variant peptides. We validated 360 spectra corresponding to 55 germline and 6 somatic variant peptides. Targeted MRM assays revealed two novel variant somatic peptides, CDK12-G879V and FASN-R1439Q, expressed in lung and lymph node metastatic sites, respectively. The CDK12-G879V mutation likely results in a nonfunctional CDK12 kinase and chemotherapy susceptibility in lung metastatic sites. Knockdown of CDK12 in lung adenocarcinoma cells increased chemotherapy sensitivity which was rescued by wild type, but not CDK12-G879V expression, consistent with the complete resolution of the lung metastatic sites in this patient.
Collapse
Affiliation(s)
- Xu Zhang
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Khoa Dang Nguyen
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Paul A Rudnick
- §Spectragen Informatics LLC, Bainbridge Island, Washington
| | - Nitin Roper
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Emily Kawaler
- ¶Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - Tapan K Maity
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Shivangi Awasthi
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Shaojian Gao
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Romi Biswas
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Abhilash Venugopalan
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Constance M Cultraro
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - David Fenyö
- ¶Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - Udayan Guha
- From the ‡Thoracic and GI Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland;.
| |
Collapse
|
19
|
Chandana S, Babiker HM, Mahadevan D. Therapeutic trends in pancreatic ductal adenocarcinoma (PDAC). Expert Opin Investig Drugs 2018; 28:161-177. [DOI: 10.1080/13543784.2019.1557145] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Sreenivasa Chandana
- Phase I program, START Midwest, Grand Rapids, MI, USA
- Department of Gastrointestinal Medical Oncology, Cancer and Hematology Centers of Western Michigan, Grand Rapids, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Hani M. Babiker
- Early Phase Therapeutics Program, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Daruka Mahadevan
- Early Phase Therapeutics Program, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
20
|
Wang X, Jia Y, Deng H, Liu Y, Liu Y. Intratumoral heterogeneity of esophageal squamous cell carcinoma and its clinical significance. Pathol Res Pract 2018; 215:308-314. [PMID: 30528923 DOI: 10.1016/j.prp.2018.11.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 11/13/2018] [Accepted: 11/23/2018] [Indexed: 12/29/2022]
Abstract
Recent studies have shown that intratumoral heterogeneity is prevalent in esophageal squamous cell cancer (ESCC) based on DNA sequencing and chromosome analysis in multiple regions from the same tumor. This study aimed to investigate the expression of ZNF750, EP300, MTOR and KMT2D and their intratumoral heterogeneity (ITH) in patients with ESCC. A total of 106 cases, who underwent esophagectomy from 2008 to 2010, with two foci from each case, were tested by immunohistochemistry(IHC) as well as 12 cases were tested by RNAscope in this study.We found that 58/106 (54.72%), 66/106 (62.26%), 75/106 (70.75%%) of ESCC showed high expression of ZNF750, EP300, MTOR, respectively by IHC, and 8/12 (66.67%), 10/12 (83.33%), 4/12 (33.33%) and 6/12 (50%) showed high expression of ZNF750, EP300, MTOR and KMT2D, respectively by RNAscope. Multivariate analysis showed that MTOR expression was an independent infavorable prognostic factor of overall survival (OS) (HR = 1.921; P = 0.000). This study also found that 44/106(4151%), 37/106 (34.91%), 39/106(36.79%) of ESCC showed heterogeneous expression of ZNF750, EP300 and MTOR respectively by IHC, 8/12(66.67%), 8/12(66.67%), 4/12(33.33%), 4/12(33.33%) of ZNF750, EP300, MTOR and KMT2D respectively by RNAscope, IHC and RNAscope could successfully detect a high prevalence of ITH. In conclusion, findings of this study showed that ZNF750, EP300, MTOR and KMT2D heterogeneously expressed in ESCC. High expression of ZNF750 related to a better outcome, while EP300 and MTOR related to a poor prognosis.
Collapse
Affiliation(s)
- Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Ying Jia
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Huiyan Deng
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yao Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China.
| |
Collapse
|
21
|
Du W, Brekken RA. Does Axl have potential as a therapeutic target in pancreatic cancer? Expert Opin Ther Targets 2018; 22:955-966. [PMID: 30244621 PMCID: PMC6292430 DOI: 10.1080/14728222.2018.1527315] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Pancreatic cancer is a leading cause of cancer-related death. Metastasis, therapy resistance, and immunosuppression are dominant characteristics of pancreatic tumors. Strategies that enhance the efficacy of standard of care and/or immune therapy are likely the most efficient route to improve overall survival in this disease. Areas covered: Axl, a member of the TAM (Tyro3, Axl, MerTK) family of receptor tyrosine kinases, is involved in cell plasticity, chemoresistance, immune suppression, and metastasis in various cancers, including pancreatic cancer. This review provides an overview of Axl and its function in normal conditions, summarizes the regulation and function of Axl in cancer, and highlights the contribution of Axl to pancreatic cancer as well as its potential as a therapeutic target. Expert opinion: Axl is an attractive therapeutic target in pancreatic cancer because it contributes to many of the roadblocks that hamper therapeutic efficacy. Clinical evidence supporting Axl inhibition in pancreatic cancer is currently limited; however, multiple clinical trials have been initiated or are in the planning phase to test the effect of inhibiting Axl in conjunction with standard therapy in pancreatic cancer patients. We anticipate that these studies will provide robust validation of Axl as a therapeutic target in pancreatic cancer.
Collapse
|
22
|
Signal-Targeted Therapies and Resistance Mechanisms in Pancreatic Cancer: Future Developments Reside in Proteomics. Cancers (Basel) 2018; 10:cancers10060174. [PMID: 29865155 PMCID: PMC6025626 DOI: 10.3390/cancers10060174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/18/2022] Open
Abstract
For patients with metastatic pancreatic cancer that are not eligible for surgery, signal-targeted therapies have so far failed to significantly improve survival. These therapeutic options have been tested in phase II/III clinical trials mostly in combination with the reference treatment gemcitabine. Innovative therapies aim to annihilate oncogenic dependency, or to normalize the tumoural stroma to allow immune cells to function and/or re-vascularisation to occur. Large scale transcriptomic and genomic analysis revealed that pancreatic cancers display great heterogeneity but failed to clearly delineate specific oncogene dependency, besides oncogenic Kras. Beyond these approaches, proteomics appears to be an appropriate approach to classify signal dependency and to identify specific alterations at the targetable level. However, due to difficulties in sampling, proteomic data for this pathology are scarce. In this review, we will discuss the current state of clinical trials for targeted therapies against pancreatic cancer. We will then highlight the most recent proteomic data for pancreatic tumours and their metastasis, which could help to identify major oncogenic signalling dependencies, as well as provide future leads to explain why pancreatic tumours are intrinsically resistant to signal-targeted therapies. We will finally discuss how studies on phosphatidylinositol-3-kinase (PI3K) signalling, as the paradigmatic pro-tumoural signal downstream of oncogenic Kras in pancreatic cancer, would benefit from exploratory proteomics to increase the efficiency of targeted therapies.
Collapse
|
23
|
Yadav DK, Bai X, Yadav RK, Singh A, Li G, Ma T, Chen W, Liang T. Liquid biopsy in pancreatic cancer: the beginning of a new era. Oncotarget 2018; 9:26900-26933. [PMID: 29928492 PMCID: PMC6003564 DOI: 10.18632/oncotarget.24809] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/25/2018] [Indexed: 12/21/2022] Open
Abstract
With dismal survival rate pancreatic cancer remains one of the most aggressive and devastating malignancy. Predominantly, due to the absence of a dependable methodology for early identification and limited therapeutic options for advanced disease. However, it takes over 17 years to develop pancreatic cancer from initiation of mutation to metastatic cancer; therefore, if diagnosed early; it may increase overall survival dramatically, thus, providing a window of opportunity for early detection. Recently, genomic expression analysis defined 4 subtypes of pancreatic cancer based on mutated genes. Hence, we need simple and standard, minimally invasive test that can monitor those altered genes or their associated pathways in time for the success of precision medicine, and liquid biopsy seems to be one answer to all these questions. Again, liquid biopsy has an ability to pair with genomic tests. Additionally, liquid biopsy based development of circulating tumor cells derived xenografts, 3D organoids system, real-time monitoring of genetic mutations by circulating tumor DNA and exosome as the targeted drug delivery vehicle holds lots of potential for the treatment and cure of pancreatic cancer. At present, diagnosis of pancreatic cancer is frantically done on the premise of CA19-9 and radiological features only, which doesn't give a picture of genetic mutations and epigenetic alteration involved. In this manner, the current diagnostic paradigm for pancreatic cancer diagnosis experiences low diagnostic accuracy. This review article discusses the current state of liquid biopsy in pancreatic cancer as diagnostic and therapeutic tools and future perspectives of research in the light of circulating tumor cells, circulating tumor DNA and exosomes.
Collapse
Affiliation(s)
- Dipesh Kumar Yadav
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Rajesh Kumar Yadav
- Department of Pharmacology, Gandaki Medical College, Tribhuwan University, Institute of Medicine, Pokhara 33700, Nepal
| | - Alina Singh
- Department of Surgery, Bir Hospital, National Academy of Medical Science, Kanti Path, Kathmandu 44600, Nepal
| | - Guogang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tao Ma
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wei Chen
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
24
|
Hsieh CC, Shyr YM, Liao WY, Chen TH, Wang SE, Lu PC, Lin PY, Chen YB, Mao WY, Han HY, Hsiao M, Yang WB, Li WS, Sher YP, Shen CN. Elevation of β-galactoside α2,6-sialyltransferase 1 in a fructoseresponsive manner promotes pancreatic cancer metastasis. Oncotarget 2018; 8:7691-7709. [PMID: 28032597 PMCID: PMC5352353 DOI: 10.18632/oncotarget.13845] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive type of pancreatic cancer with clinical characteristics of local invasion and early metastasis. Recent cohort studies indicate high fructose intake is associated with an increase in pancreatic cancer risk. However, the mechanisms by which fructose promotes pancreatic tumorigenesis remain unclear. Herein, Kras+/LSLG12D mice were crossed with Elas-CreER transgenic mice to determine whether fructose intake directly contributes to tumor formation. Orthotopic tumor-xenograft experiments were performed to determine whether fructose substitution enhances the metastatic potential of PDAC cells. The mechanisms underlying the effects of fructose were explored by RNAseq analysis in combination with high-performance anion exchange chromatography. Dietary fructose was initially found to promote the development of aggressive pancreatic cancer in mice conditionally expressing KrasG12D in the adult pancreas. We further revealed that fructose substitution enhanced the metastatic potential of human PDAC cell via selective outgrowth of aggressive ABCG2-positive subpopulations and elevating N-acetylmannosamine levels that upregulated β-galactoside α2,6-sialyltransferase 1 (ST6Gal1), thereby promoting distant metastasis. Finally, we observed that PDAC patients expressing higher levels of ST6Gal1 and GLUT5 presented poorer prognosis compared to other groups. In conclusion, our findings have elucidated a crucial role of ST6Gal1 in regulating the invasiveness of PDACs in a fructose-responsive manner.
Collapse
Affiliation(s)
- Chi-Che Hsieh
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taiwan.,Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Yi-Ming Shyr
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-Ying Liao
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Tien-Hua Chen
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Anatomy and Cell Biology and National Yang-Ming University, Taipei, Taiwan
| | - Shin-E Wang
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Peir-Chuen Lu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Yu Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Yan-Bo Chen
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Wan-Yu Mao
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Hsin-Ying Han
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Wen-Bin Yang
- Genomics Research Center and Academia Sinica, Taipei, Taiwan
| | - Wen-Shan Li
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yuh-Pyng Sher
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center and Academia Sinica, Taipei, Taiwan.,Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
25
|
The binding landscape of a partially-selective isopeptidase inhibitor with potent pro-death activity, based on the bis(arylidene)cyclohexanone scaffold. Cell Death Dis 2018; 9:184. [PMID: 29416018 PMCID: PMC5833369 DOI: 10.1038/s41419-017-0259-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/07/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023]
Abstract
Diaryldienone derivatives with accessible β-carbons show strong anti-neoplastic properties, related to their ability to make covalent adducts with free thiols by Michael addition, and low toxicity in vivo. Accumulation of poly-ubiquitylated proteins, activation of the unfolded protein response (UPR) and induction of cell death are universal hallmarks of their activities. These compounds have been characterized as inhibitors of isopeptidases, a family of cysteine-proteases, which de-conjugate ubiquitin and ubiquitin-like proteins from their targets. However, it is unclear whether they can also react with additional proteins. In this work, we utilized the biotin-conjugated diaryldienone-derivative named 2c, as a bait to purify novel cellular targets of these small molecules. Proteomic analyses have unveiled that, in addition to isopeptidases, these inhibitors can form stable covalent adducts with different intracellular proteins, thus potentially impacting on multiple functions of the cells, from cytoskeletal organization to metabolism. These widespread activities can explain the ability of diaryldienone derivatives to efficiently trigger different cell death pathways.
Collapse
|
26
|
Wang Z, Kim MS, Martinez-Ferrando I, Koleske AJ, Pandey A, Cole PA. Analysis of Cellular Tyrosine Phosphorylation via Chemical Rescue of Conditionally Active Abl Kinase. Biochemistry 2018; 57:1390-1398. [PMID: 29341593 DOI: 10.1021/acs.biochem.7b01158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Identifying direct substrates targeted by protein kinases is important in understanding cellular physiology and intracellular signal transduction. Mass spectrometry-based quantitative proteomics provides a powerful tool for comprehensively characterizing the downstream substrates of protein kinases. This approach is efficiently applied to receptor kinases that can be precisely, directly, and rapidly activated by some agent, such as a growth factor. However, nonreceptor tyrosine kinase Abl lacks the experimental advantage of extracellular growth factors as immediate and direct stimuli. To circumvent this limitation, we combine a chemical rescue approach with quantitative phosphoproteomics to identify targets of Abl and their phosphorylation sites with enhanced temporal resolution. Both known and novel putative substrates are identified, presenting opportunities for studying unanticipated functions of Abl under physiological and pathological conditions.
Collapse
Affiliation(s)
- Zhihong Wang
- Department of Chemistry & Biochemistry, University of the Sciences , Philadelphia, Pennsylvania 19104, United States
| | - Min-Sik Kim
- Global Center for Pharmaceutical Ingredient Materials, Department of Applied Chemistry, Kyung Hee University , Yongin, Gyeonggi, Republic of Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University , Seoul, Republic of Korea
| | - Isabel Martinez-Ferrando
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Anthony J Koleske
- Departments of Molecular Biophysics and Biochemistry and Neuroscience, Yale University , New Haven, Connecticut 06520, United States
| | - Akhilesh Pandey
- Departments of Oncology and Biological Chemistry, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Philip A Cole
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States.,Division of Genetics, Brigham and Women's Hospital, Departments of Medicine and Biological Chemistry and Molecular Pharmacology, Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
27
|
Lim LC, Lim YM. Proteome Heterogeneity in Colorectal Cancer. Proteomics 2018; 18. [PMID: 29316255 DOI: 10.1002/pmic.201700169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/17/2017] [Indexed: 01/26/2023]
Abstract
Tumor heterogeneity is an important feature of colorectal cancer (CRC) manifested by dynamic changes in gene expression, protein expression, and availability of different tumor subtypes. Recent publications in the past 10 years have revealed proteome heterogeneity between different colorectal tumors and within the same tumor site. This paper reviews recent research works on the proteome heterogeneity in CRC, which includes the heterogeneity within a single tumor (intratumor heterogeneity), between different anatomical sites at the same organ, and between primary and metastatic sites (intertumor heterogeneity). The potential use of proteome heterogeneity in precision medicine and its implications in biomarker discovery and therapeutic outcomes will be discussed. Identification of the unique proteome landscape between and within individual tumors is imperative for understanding cancer biology and the management of CRC patients.
Collapse
Affiliation(s)
- Lay Cheng Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| | - Yang Mooi Lim
- Centre for Cancer Research, Faculty of Medicine and Health Sciences, University of Tunku Abdul Rahman, Selangor, Malaysia
| |
Collapse
|
28
|
Torphy RJ, Wang Z, True-Yasaki A, Volmar KE, Rashid N, Yeh B, Johansen JS, Hollingsworth MA, Yeh JJ, Collisson EA. Stromal Content Is Correlated With Tissue Site, Contrast Retention, and Survival in Pancreatic Adenocarcinoma. JCO Precis Oncol 2018; 2018:PO.17.00121. [PMID: 30506016 PMCID: PMC6262879 DOI: 10.1200/po.17.00121] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Desmoplastic stroma is a cardinal feature of primary pancreatic ductal adenocarcinoma (PDAC), but its effects on the biology, prognosis and therapeutic outcomes are not known. We developed an automated method to assess tumor stroma density (TSD) and investigated computed tomography (CT)-correlates of stroma in PDAC. PATIENTS AND METHODS We collected PDAC samples from rapid autopsy and resection series and digitally annotated samples to quantify TSD. A series of resected patients also underwent preoperative multiphasic CT. RESULTS Automated and manual assessments of TSD were highly correlated (ρ= 0.65, P < 0.001). Solid organ metastases had a lower median TSD than primary tumors (P < 0.001). Patients with high TSD enjoyed prolonged recurrence free survival (RFS) (P = 0.003; HR = 0.51) and overall survival (P = 0.008, HR = 0.57). In another independent dataset, patients with high TSD had decreased risk for recurrence (P = 0.003, HR = 0.03) and death (P = 0.003, HR = 0.03) at time of resection, however the protective effect diminished over time. Patients with normalized portovenous phase CT tumor enhancement ratio ≥0.40 had a longer RFS following resection (P = 0.020). Normalized portovenous phase CT tumor enhancement ratio was significantly correlated with TSD (P = 0.003). CONCLUSIONS Objective quantitative assessment of stroma in PDAC revealed several clinically relevant observations. Firstly, stroma was less abundant in metastatic PDAC, the cause of most PDAC mortality. Secondly, high stromal content correlates with favorable outcome in resected cases, implying a protective effect of stroma and suggesting careful consideration of active stromal depletion therapies. Finally, standard multiphase CT imaging correlates with stroma content as well as clinical outcome, indicating that non-invasive assessment of stroma is a feasible sensitivity enrichment approach in PDAC.
Collapse
Affiliation(s)
- Robert J. Torphy
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Zhen Wang
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Aisha True-Yasaki
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Keith E. Volmar
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Naim Rashid
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Benjamin Yeh
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Julia S. Johansen
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Michael A. Hollingsworth
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Jen Jen Yeh
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| | - Eric A. Collisson
- Robert J. Torphy, University of Colorado, Aurora, CO; Zhen Wang Aisha True-Yasaki, Benjamin Yeh, and Eric A. Collisson, University of California, San Francisco, CA; Keith E. Volmar, Rex Healthcare; Keith E. Volmar, Naim Rashid, and Jen Jen Yeh, University of North Carolina, Chapel Hill, Chapel Hill, NC; Julia S. Johansen, University of Copenhagen, Copenhagen, Denmark; and Michael A. Hollingsworth, University of Nebraska, Lincoln, NE
| |
Collapse
|
29
|
Thillai K, Lam H, Sarker D, Wells CM. Deciphering the link between PI3K and PAK: An opportunity to target key pathways in pancreatic cancer? Oncotarget 2017; 8:14173-14191. [PMID: 27845911 PMCID: PMC5355171 DOI: 10.18632/oncotarget.13309] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/27/2016] [Indexed: 02/07/2023] Open
Abstract
The development of personalised therapies has ushered in a new and exciting era of cancer treatment for a variety of solid malignancies. Yet pancreatic ductal adenocarcinoma (PDAC) has failed to benefit from this paradigm shift, remaining notoriously refractory to targeted therapies. Chemotherapy is the cornerstone of management but can offer only modest survival benefits of a few months with 5-year survival rates rarely exceeding 3%. Despite these disappointing statistics, significant strides have been made towards understanding the complex biology of pancreatic cancer, with deep genomic sequencing identifying novel genetic aberrations and key signalling pathways. The PI3K-PDK1-AKT pathway has received great attention due to its prominence in carcinogenesis. However, efforts to target several components of this network have resulted in only a handful of drugs demonstrating any survival benefit in solid tumors; despite promising pre-clinical results. p-21 activated kinase 4 (PAK4) is a gene that is recurrently amplified or overexpressed in PDAC and both PAK4 and related family member PAK1, have been linked to aberrant RAS activity, a common feature in pancreatic cancer. As regulators of PI3K, PAKs have been highlighted as a potential prognostic marker and therapeutic target. In this review, we discuss the biology of pancreatic cancer and the close interaction between PAKs and the PI3K pathway. We also suggest proposals for future research that may see the development of effective targeted therapies that could finally improve outcomes for this disease.
Collapse
Affiliation(s)
- Kiruthikah Thillai
- Division of Cancer Studies, King's College London, London, United Kingdom.,Department of Medical Oncology, Guy's and St Thomas' NHS Trust, London, United Kingdom
| | - Hoyin Lam
- Division of Cancer Studies, King's College London, London, United Kingdom
| | - Debashis Sarker
- Division of Cancer Studies, King's College London, London, United Kingdom.,Department of Medical Oncology, Guy's and St Thomas' NHS Trust, London, United Kingdom
| | - Claire M Wells
- Division of Cancer Studies, King's College London, London, United Kingdom
| |
Collapse
|
30
|
Mechanisms and clinical implications of tumor heterogeneity and convergence on recurrent phenotypes. J Mol Med (Berl) 2017; 95:1167-1178. [PMID: 28871446 DOI: 10.1007/s00109-017-1587-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/11/2017] [Accepted: 08/20/2017] [Indexed: 10/18/2022]
Abstract
Tumor heterogeneity has been identified at various -omic levels. The tumor genome, transcriptome, proteome, and phenome can vary widely across cells in patient tumors and are influenced by tumor cell interactions with heterogeneous physical conditions and cellular components of the tumor microenvironment. Here, we explore the concept that while variation exists at multiple -omic levels, changes at each of these levels converge on the same pathways and lead to convergent phenotypes in tumors that can provide common drug targets. These phenotypes include cellular growth and proliferation, sustained oncogenic signaling, and immune avoidance, among others. Tumor heterogeneity complicates treatment of patient cancers as it leads to varied response to therapies. Identification of convergent cellular phenotypes arising in patient cancers and targeted therapies that reverse them has the potential to transform the way clinicians treat these cancers and to improve patient outcome.
Collapse
|
31
|
Le Large TYS, Bijlsma MF, Kazemier G, van Laarhoven HWM, Giovannetti E, Jimenez CR. Key biological processes driving metastatic spread of pancreatic cancer as identified by multi-omics studies. Semin Cancer Biol 2017; 44:153-169. [PMID: 28366542 DOI: 10.1016/j.semcancer.2017.03.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy, characterized by a high metastatic burden, already at the time of diagnosis. The metastatic potential of PDAC is one of the main reasons for the poor outcome next to lack of significant improvement in effective treatments in the last decade. Key mutated driver genes, such as activating KRAS mutations, are concordantly expressed in primary and metastatic tumors. However, the biology behind the metastatic potential of PDAC is not fully understood. Recently, large-scale omic approaches have revealed new mechanisms by which PDAC cells gain their metastatic potency. In particular, genomic studies have shown that multiple heterogeneous subclones reside in the primary tumor with different metastatic potential. The development of metastases may be correlated to a more mesenchymal transcriptomic subtype. However, for cancer cells to survive in a distant organ, metastatic sites need to be modulated into pre-metastatic niches. Proteomic studies identified the influence of exosomes on the Kuppfer cells in the liver, which could function to prepare this tissue for metastatic colonization. Phosphoproteomics adds an extra layer to the established omic techniques by unravelling key functional signaling. Future studies integrating results from these large-scale omic approaches will hopefully improve PDAC prognosis through identification of new therapeutic targets and patient selection tools. In this article, we will review the current knowledge on the biology of PDAC metastasis unravelled by large scale multi-omic approaches.
Collapse
Affiliation(s)
- T Y S Le Large
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Laboratory of Experimental Oncology and Radiobiology, Academic Medical Center, Amsterdam, The Netherlands; Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - M F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Academic Medical Center, Amsterdam, The Netherlands
| | - G Kazemier
- Department of Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - H W M van Laarhoven
- Department of Medical Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - E Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy; CNR-Nano, Institute of Nanoscience and Nanotechnology, Pisa, Italy
| | - C R Jimenez
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Zhang X, Maity T, Kashyap MK, Bansal M, Venugopalan A, Singh S, Awasthi S, Marimuthu A, Charles Jacob HK, Belkina N, Pitts S, Cultraro CM, Gao S, Kirkali G, Biswas R, Chaerkady R, Califano A, Pandey A, Guha U. Quantitative Tyrosine Phosphoproteomics of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor-treated Lung Adenocarcinoma Cells Reveals Potential Novel Biomarkers of Therapeutic Response. Mol Cell Proteomics 2017; 16:891-910. [PMID: 28331001 PMCID: PMC5417828 DOI: 10.1074/mcp.m117.067439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
Mutations in the Epidermal growth factor receptor (EGFR) kinase domain, such as the L858R missense mutation and deletions spanning the conserved sequence 747LREA750, are sensitive to tyrosine kinase inhibitors (TKIs). The gatekeeper site residue mutation, T790M accounts for around 60% of acquired resistance to EGFR TKIs. The first generation EGFR TKIs, erlotinib and gefitinib, and the second generation inhibitor, afatinib are FDA approved for initial treatment of EGFR mutated lung adenocarcinoma. The predominant biomarker of EGFR TKI responsiveness is the presence of EGFR TKI-sensitizing mutations. However, 30-40% of patients with EGFR mutations exhibit primary resistance to these TKIs, underscoring the unmet need of identifying additional biomarkers of treatment response. Here, we sought to characterize the dynamics of tyrosine phosphorylation upon EGFR TKI treatment of mutant EGFR-driven human lung adenocarcinoma cell lines with varying sensitivity to EGFR TKIs, erlotinib and afatinib. We employed stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative mass spectrometry to identify and quantify tyrosine phosphorylated peptides. The proportion of tyrosine phosphorylated sites that had reduced phosphorylation upon erlotinib or afatinib treatment correlated with the degree of TKI-sensitivity. Afatinib, an irreversible EGFR TKI, more effectively inhibited tyrosine phosphorylation of a majority of the substrates. The phosphosites with phosphorylation SILAC ratios that correlated with the TKI-sensitivity of the cell lines include sites on kinases, such as EGFR-Y1197 and MAPK7-Y221, and adaptor proteins, such as SHC1-Y349/350, ERRFI1-Y394, GAB1-Y689, STAT5A-Y694, DLG3-Y705, and DAPP1-Y139, suggesting these are potential biomarkers of TKI sensitivity. DAPP1, is a novel target of mutant EGFR signaling and Y-139 is the major site of DAPP1 tyrosine phosphorylation. We also uncovered several off-target effects of these TKIs, such as MST1R-Y1238/Y1239 and MET-Y1252/1253. This study provides unique insight into the TKI-mediated modulation of mutant EGFR signaling, which can be applied to the development of biomarkers of EGFR TKI response.
Collapse
Affiliation(s)
- Xu Zhang
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Tapan Maity
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Manoj K Kashyap
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Mukesh Bansal
- ¶Department of System Biology, Columbia University, New York, New York, 10032
- ‖PsychoGenics Inc., Tarrytown, New York, 10591
| | - Abhilash Venugopalan
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Sahib Singh
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shivangi Awasthi
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | | | | | - Natalya Belkina
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Stephanie Pitts
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Constance M Cultraro
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shaojian Gao
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Guldal Kirkali
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Romi Biswas
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Raghothama Chaerkady
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
- **Medimmune LLC, Gaithersburg, Maryland, 20878
| | - Andrea Califano
- ¶Department of System Biology, Columbia University, New York, New York, 10032
| | - Akhilesh Pandey
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Udayan Guha
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892;
| |
Collapse
|
33
|
Cutler JA, Tahir R, Sreenivasamurthy SK, Mitchell C, Renuse S, Nirujogi RS, Patil AH, Heydarian M, Wong X, Wu X, Huang TC, Kim MS, Reddy KL, Pandey A. Differential signaling through p190 and p210 BCR-ABL fusion proteins revealed by interactome and phosphoproteome analysis. Leukemia 2017; 31:1513-1524. [DOI: 10.1038/leu.2017.61] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 12/15/2022]
|
34
|
Abstract
OBJECTIVES Mass spectrometry-based proteomics enables near-comprehensive protein expression profiling. We aimed to compare quantitatively the relative expression levels of thousands of proteins across 5 pancreatic cell lines. METHODS Using tandem mass tags (TMT10-plex), we profiled the global proteomes of 5 cell lines in duplicate in a single multiplexed experiment. We selected cell lines commonly used in pancreatic research: CAPAN-1, HPAC, HPNE, PANC1, and PaSCs. In addition, we examined the effects of different proteases (Lys-C and Lys-C plus trypsin) on the dataset depth. RESULTS We quantified over 8000 proteins across the 5 cell lines. Analysis of variance testing of cell lines within each data set resulted in over 1400 statistically significant differences in protein expression levels. Comparing the data sets, 10% more proteins and 30% more peptides were identified in the Lys-C/trypsin data set than in the Lys-C-only data set. The correlation coefficient of quantified proteins common between the data sets was greater than 0.85. CONCLUSIONS We illustrate protein level differences across pancreatic cell lines. In addition, we highlight the advantages of Lys-C/trypsin over Lys-C-only digests for discovery proteomics. These data sets provide a valuable resource of cell line-dependent peptide and protein differences for future targeted analyses, including those investigating on- or off-target drug effects across cell lines.
Collapse
|
35
|
Coleman O, Henry M, McVey G, Clynes M, Moriarty M, Meleady P. Proteomic strategies in the search for novel pancreatic cancer biomarkers and drug targets: recent advances and clinical impact. Expert Rev Proteomics 2016; 13:383-94. [PMID: 26985644 DOI: 10.1586/14789450.2016.1167601] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers; despite a low incidence rate it is the fourth leading cause of cancer-related death in the world. Improvement of the diagnosis, prognosis and treatment remains the main focus of pancreatic cancer research. Rapid developments in proteomic technologies has improved our understanding of the pancreatic cancer proteome. Here, the authors summarise the recent proteomic strategies undertaken in the search for: novel biomarkers for early diagnosis, pancreatic cancer-specific proteins which may be used for novel targeted therapies and proteins which may be useful for monitoring disease progression post-therapy. Recent advances and findings discussed here provide great promise of having a significant clinical impact and improving the outcome of patients with this malignancy.
Collapse
Affiliation(s)
- Orla Coleman
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Michael Henry
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Gerard McVey
- b St. Lukes Hospital , Rathgar , Dublin 6 , Ireland
| | - Martin Clynes
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| | - Michael Moriarty
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland.,b St. Lukes Hospital , Rathgar , Dublin 6 , Ireland
| | - Paula Meleady
- a Department of Proteomics, National Institute for Cellular Biotechnology , Dublin City University , Glasnevin , Dublin 9 , Ireland
| |
Collapse
|
36
|
Kim MS, Zhong J, Pandey A. Common errors in mass spectrometry-based analysis of post-translational modifications. Proteomics 2016; 16:700-14. [PMID: 26667783 DOI: 10.1002/pmic.201500355] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/05/2015] [Accepted: 12/08/2015] [Indexed: 12/29/2022]
Abstract
Mass spectrometry (MS) is a powerful tool to analyze complex mixtures of proteins in a high-throughput fashion. Proteome analysis has already become a routine task in biomedical research with the emergence of proteomics core facilities in most research institutions. Post-translational modifications (PTMs) represent a mechanism by which complex biological processes are orchestrated dynamically at the systems level. MS is rapidly becoming popular to discover new modifications and novel sites of known PTMs, revolutionizing the current understanding of diverse signaling pathways and biological processes. However, MS-based analysis of PTMs has its own caveats and pitfalls that can lead to erroneous conclusions. Here, we review the most common errors in MS-based PTM analyses with the goal of adopting strategies that maximize correct interpretation in the context of biological questions that are being addressed. Finally, we provide suggestions that should help mass spectrometrists, bioinformaticians and biologists to perform and interpret MS-based PTM analyses more accurately.
Collapse
Affiliation(s)
- Min-Sik Kim
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Zhong
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Abstract
Metastases that are resistant to conventional therapy are the major cause of death from cancer. In most patients, metastasis has already occurred by the time of diagnosis. Thus, the prevention of metastasis is unlikely to be of therapeutic benefit. The biological heterogeneity of metastases presents a major obstacle to treatment. However, the growth and survival of metastases depend on interactions between tumor cells and host homeostatic mechanisms. Targeting these interactions, in addition to the tumor cells, can produce synergistic therapeutic effects against existing metastases.
Collapse
Affiliation(s)
- Isaiah J Fidler
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 173, Houston, TX, 77030, USA.
| | - Margaret L Kripke
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 173, Houston, TX, 77030, USA
| |
Collapse
|
38
|
Carvalho AS, Matthiesen R. Global MS-Based Proteomics Drug Profiling. Methods Mol Biol 2016; 1449:469-79. [PMID: 27613057 DOI: 10.1007/978-1-4939-3756-1_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
DNA-based technologies such as RNAi, chemical-genetic profiling, or gene expression profiling by DNA microarrays combined with other biochemical methods are established strategies for surveying drug mechanisms. Such approaches can provide mechanistic information on how drugs act and affect cellular pathways. By studying how cancer cells compensate for the drug treatment, novel targets used in a combined treatment can be designed. Furthermore, toxicity effects on cells not targeted can be obtained on a molecular level. For example, drug companies are particularly interested in studying the molecular side effects of drugs in the liver. In addition, experiments with the purpose of elucidating liver toxicity can be studied using samples obtained from animal models exposed to different concentrations of a drug over time. More recently considerable advances in mass spectrometry (MS) technologies and bioinformatics tools allows informative global drug profiling experiments to be performed at a cost comparable to other large-scale technologies such as DNA-based technologies. Moreover, MS-based proteomics provides an additional layer of information on the dynamic regulation of proteins translation and particularly protein degradation. MS-based proteomics approaches combined with other biochemical methods delivers information on regulatory networks, signaling cascades, and metabolic pathways upon drug treatment. Furthermore, MS-based proteomics can provide additional information on single amino acid polymorphisms, protein isoform distribution, posttranslational modifications, and subcellular localization. In this chapter, we will share our experience using MS based proteomics as a pharmacoproteomics strategy to characterize drug mechanisms of action in single drug therapy or in multidrug combination. Finally, the emergence of integrated proteogenomics analysis, such as "The Cancer Genome Atlas" program, opened interesting perspectives to extend this approach to drug target discovery and validation.
Collapse
Affiliation(s)
- Ana Sofia Carvalho
- Computational and Experimental Biology Group, Department of Health Promotion and Chronic Diseases, National Health Institute Dr. Ricardo Jorge, INSA, I.P., Av Padre Cruz, Lisboa, 1649-016, Portugal.
| | - Rune Matthiesen
- Computational and Experimental Biology Group, Department of Health Promotion and Chronic Diseases, National Health Institute Dr. Ricardo Jorge, INSA, I.P., Av Padre Cruz, Lisboa, 1649-016, Portugal
| |
Collapse
|
39
|
Humphrey ES, Su SP, Nagrial AM, Hochgräfe F, Pajic M, Lehrbach GM, Parton RG, Yap AS, Horvath LG, Chang DK, Biankin AV, Wu J, Daly RJ. Resolution of Novel Pancreatic Ductal Adenocarcinoma Subtypes by Global Phosphotyrosine Profiling. Mol Cell Proteomics 2016; 15:2671-85. [PMID: 27259358 PMCID: PMC4974343 DOI: 10.1074/mcp.m116.058313] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 05/11/2016] [Indexed: 12/20/2022] Open
Abstract
Comprehensive characterization of signaling in pancreatic ductal adenocarcinoma (PDAC) promises to enhance our understanding of the molecular aberrations driving this devastating disease, and may identify novel therapeutic targets as well as biomarkers that enable stratification of patients for optimal therapy. Here, we use immunoaffinity-coupled high-resolution mass spectrometry to characterize global tyrosine phosphorylation patterns across two large panels of human PDAC cell lines: the ATCC series (19 cell lines) and TKCC series (17 cell lines). This resulted in the identification and quantification of over 1800 class 1 tyrosine phosphorylation sites and the consistent segregation of both PDAC cell line series into three subtypes with distinct tyrosine phosphorylation profiles. Subtype-selective signaling networks were characterized by identification of subtype-enriched phosphosites together with pathway and network analyses. This revealed that the three subtypes characteristic of the ATCC series were associated with perturbations in signaling networks associated with cell-cell adhesion and epithelial-mesenchyme transition, mRNA metabolism, and receptor tyrosine kinase (RTK) signaling, respectively. Specifically, the third subtype exhibited enhanced tyrosine phosphorylation of multiple RTKs including the EGFR, ERBB3 and MET. Interestingly, a similar RTK-enriched subtype was identified in the TKCC series, and 'classifier' sites for each series identified using Random Forest models were able to predict the subtypes of the alternate series with high accuracy, highlighting the conservation of the three subtypes across the two series. Finally, RTK-enriched cell lines from both series exhibited enhanced sensitivity to the small molecule EGFR inhibitor erlotinib, indicating that their phosphosignature may provide a predictive biomarker for response to this targeted therapy. These studies highlight how resolution of subtype-selective signaling networks can provide a novel taxonomy for particular cancers, and provide insights into PDAC biology that can be exploited for improved patient management.
Collapse
Affiliation(s)
- Emily S Humphrey
- From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia; §St Vincent's Hospital Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia
| | - Shih-Ping Su
- ¶Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Level 1, Building 77, Monash University, VIC 3800, Australia
| | - Adnan M Nagrial
- From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia; §St Vincent's Hospital Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia
| | - Falko Hochgräfe
- ‖Competence Center Functional Genomics, University of Greifswald, F.-L-Jahnstr. 15, 17489 Greifswald, Germany
| | - Marina Pajic
- From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia; §St Vincent's Hospital Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia
| | - Gillian M Lehrbach
- From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia
| | - Robert G Parton
- **Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane QLD 4072, Australia
| | - Alpha S Yap
- **Division of Cell Biology and Molecular Medicine, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Brisbane QLD 4072, Australia
| | - Lisa G Horvath
- From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia; ‡‡Chris O'Brien Lifehouse, Missenden Road, Camperdown, NSW 2050, Australia
| | - David K Chang
- §§Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Andrew V Biankin
- §§Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK;
| | - Jianmin Wu
- ¶¶Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, 52 Fu-Cheng Road, Hai-Dian District, Beijing 100142, China From the ‡Cancer Division and Kinghorn Cancer Centre, Garvan Institute of Medical Research, 384 Victoria St, Sydney, NSW 2010, Australia; §St Vincent's Hospital Clinical School, Faculty of Medicine, University of New South Wales, NSW 2052, Australia;
| | - Roger J Daly
- ¶Cancer Program, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Level 1, Building 77, Monash University, VIC 3800, Australia;
| |
Collapse
|
40
|
Pinato DJ, Chowdhury S, Stebbing J. TAMing resistance to multi-targeted kinase inhibitors through Axl and Met inhibition. Oncogene 2016; 35:2684-6. [PMID: 26434595 DOI: 10.1038/onc.2015.374] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/28/2015] [Indexed: 02/08/2023]
Abstract
TAM (Tyro3-Axl-Mer) receptor tyrosine kinases and Met are implicated in several hallmarks of cancer progression including sustained angiogenesis, enhanced motility, tissue invasion and acquisition of metastatic potential through the upregulation of epithelial-to-mesenchymal transition. Increasing evidence has confirmed Axl and Met as emerging central drivers of adaptive resistance to targeted therapies across a wide variety of cancers. In this issue of Oncogene, Zhou et al. describe the mechanisms linking Axl and Met activation to acquired resistance to sunitinib in renal cell carcinoma (RCC), providing a pre-clinical rationale for the development of Axl and Met inhibitors including cabozantinib in anti-angiogenic resistant RCC.
Collapse
Affiliation(s)
- D J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - S Chowdhury
- Department of Medical Oncology, Guy's Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - J Stebbing
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
41
|
Abstract
The interaction between Axl receptor tyrosine kinase and its main ligand Gas6 has been implicated in the progression of a wide number of malignancies. More recently, overexpression of Axl has emerged as a key molecular determinant underlying the development of acquired resistance to targeted anticancer agents. The activation of Axl is overexpression-dependent and controls a number of hallmarks of cancer progression including proliferation, migration, resistance to apoptosis and survival through a complex network of intracellular second messengers. Axl has been noted to influence clinically meaningful end points including metastatic recurrence and survival in the vast majority of tumour types. With Axl inhibitors having gained momentum as novel anticancer therapies, we provide an overview of the biological and clinical relevance of this molecular pathway, outlining the main directions of research.
Collapse
Affiliation(s)
- Matthew Brown
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - James R M Black
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Rohini Sharma
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
42
|
Mitchell CJ, Kim MS, Zhong J, Nirujogi RS, Bose AK, Pandey A. Unbiased identification of substrates of protein tyrosine phosphatase ptp-3 in C. elegans. Mol Oncol 2016; 10:910-20. [PMID: 27067626 DOI: 10.1016/j.molonc.2016.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/26/2016] [Accepted: 03/15/2016] [Indexed: 01/11/2023] Open
Abstract
The leukocyte antigen related (LAR) family of receptor-like protein tyrosine phosphatases has three members in humans - PTPRF, PTPRD and PTPRS - that have been implicated in diverse processes including embryonic development, inhibition of cell growth and axonal guidance. Mutations in the LAR family are associated with developmental defects such as cleft palate as well as various cancers including breast, neck, lung, colon and brain. Although this family of tyrosine phosphatases is important for many developmental processes, little is known of their substrates. This is partially due to functional redundancy within the LAR family, as deletion of a single gene in the LAR family does not have an appreciable phenotype, but a dual knockout is embryonically lethal in mouse models. To circumvent the inability to knockout multiple members of the LAR family in mouse models, we used a knockout of ptp-3, which is the only known ortholog of the LAR family in Caenorhabditis elegans and allows for the study of the LAR family at the organismal level. Using SILAC-based quantitative phosphoproteomics, we identified 255 putative substrates of ptp-3, which included four of the nine known annotated substrates of the LAR family. A motif analysis of the identified phosphopeptides allowed for the determination of sequences that appear to be preferentially dephosphorylated. Finally, we discovered that kinases were overrepresented in the list of identified putative substrates and tyrosine residues whose phosphorylation is known to increase kinase activity were dephosphorylated by ptp-3. These data are suggestive of ptp-3 as a potential negative regulator of several kinase families, such as the mitogen activated kinases (MAPKs), and multiple tyrosine kinases including FER, MET, and NTRK2.
Collapse
Affiliation(s)
- Christopher J Mitchell
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Min-Sik Kim
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Zhong
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Raja Sekhar Nirujogi
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute of Bioinformatics, Bangalore, India
| | - Anjun K Bose
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
43
|
Josić D, Andjelković U. The Role of Proteomics in Personalized Medicine. Per Med 2016. [DOI: 10.1007/978-3-319-39349-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
44
|
Dosch JS, Ziemke EK, Shettigar A, Rehemtulla A, Sebolt-Leopold JS. Cancer stem cell marker phenotypes are reversible and functionally homogeneous in a preclinical model of pancreatic cancer. Cancer Res 2015; 75:4582-92. [PMID: 26359451 DOI: 10.1158/0008-5472.can-14-2793] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 07/25/2015] [Indexed: 01/16/2023]
Abstract
Survival rates associated with pancreatic cancer remain dismal despite advancements in detection and experimental treatment strategies. Genetically engineered mouse models of pancreatic tumorigenesis have gained considerable attention based on their ability to recapitulate key clinical features of human disease including chemotherapeutic resistance and fibrosis. However, it is unclear if transgenic systems exemplified by the Kras(G12D)/Trp53(R172H)/Pdx-1-Cre (KPC) mouse model recapitulate the functional heterogeneity of human pancreatic tumors harboring distinct cells with tumorigenic properties. To facilitate tracking of heterogeneous tumor cell populations, we incorporated a luciferase-based tag into the genetic background of the KPC mouse model. We isolated pancreatic cancer cells from multiple independent tumor lines and found that roughly 1 out of 87 cells exhibited tumorigenic capability. Notably, this frequency is significantly higher than reported for human pancreatic adenocarcinomas. Cancer stem cell (CSC) markers, including CD133, CD24, Sca-1, and functional Aldefluor activity, were unable to discriminate tumorigenic from nontumorigenic cells in syngeneic transplants. Furthermore, three-dimensional spheroid cultures originating from KPC tumors did not enrich for cells with stem-like characteristics and were not significantly more tumorigenic than cells cultured as monolayers. Additionally, we did not observe significant differences in response to gemcitabine or salinomycin in several isolated subpopulations. Taken together, these studies show that the hierarchical organization of CSCs in human disease is not recapitulated in a commonly used mouse model of pancreatic cancer and therefore provide a new view of the phenotypic and functional heterogeneity of tumor cells.
Collapse
Affiliation(s)
- Joseph S Dosch
- Translational Oncology Program, University of Michigan Medical School, Ann Arbor, Michigan. Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Elizabeth K Ziemke
- Translational Oncology Program, University of Michigan Medical School, Ann Arbor, Michigan. Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Amrith Shettigar
- Translational Oncology Program, University of Michigan Medical School, Ann Arbor, Michigan. Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Alnawaz Rehemtulla
- Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan. Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Judith S Sebolt-Leopold
- Translational Oncology Program, University of Michigan Medical School, Ann Arbor, Michigan. Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan. Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
45
|
Logsdon CD, Arumugam T, Ramachandran V. Animal Models of Gastrointestinal and Liver Diseases. The difficulty of animal modeling of pancreatic cancer for preclinical evaluation of therapeutics. Am J Physiol Gastrointest Liver Physiol 2015; 309:G283-91. [PMID: 26159697 PMCID: PMC4556944 DOI: 10.1152/ajpgi.00169.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 06/25/2015] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is relatively rare but extremely lethal. Standard cytotoxic therapeutics provide little benefit. To date, newer targeted therapeutics have also not been highly successful. Often novel therapeutics that have appeared to perform well in preclinical models have failed in the clinic. Many factors contribute to these failures, but the one most often attributed is the shortcomings of the preclinical models. A plethora of animal models now exist for PDAC, including cell line xenografts, patient-derived xenografts, a wide variety of genetic mouse models, and syngeneic xenografts. These models have generated a tremendous amount of information useful for the understanding of PDAC. Yet none seems to well predict clinical outcomes of new treatments. This review will discuss how genetic instability and cellular heterogeneity make this disease so difficult to model accurately. We will also discuss the strengths and weaknesses of many of the popular models. Ultimately we will argue that there is no perfect model and that the best approach to understanding clinical performance is the use of multiple preclinical models with an understanding of their salient features.
Collapse
Affiliation(s)
- Craig D. Logsdon
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and ,2Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thiruvengadam Arumugam
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| | - Vijaya Ramachandran
- 1Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas and
| |
Collapse
|
46
|
Reardon ES, Hong JA, Straughan DM, Azoury SC, Zhang M, Schrump DS. Pulmonary Metastases Exhibit Epigenetic Clonality: Implications for Precision Cancer Therapy. Ann Thorac Surg 2015; 100:1839-48; discussion 1848. [PMID: 26298164 DOI: 10.1016/j.athoracsur.2015.05.089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/12/2015] [Accepted: 05/15/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Development of effective cancer therapies may be limited by intratumoral heterogeneity, which facilitates outgrowth and organ-specific dissemination of treatment resistant clones. At present, limited information is available regarding epigenetic landscapes of pulmonary metastases. This study was undertaken to characterize epigenetic signatures of pulmonary metastases and to identify potential therapeutic targets. METHODS RNA and DNA were extracted from 65 pulmonary metastases resected from 12 patients (5 with sarcoma, 7 with adrenocortical carcinoma). Quantitative reverse transcription polymerase chain reaction techniques were used to evaluate expression levels of cancer-testis (CT) genes (NY-ESO-1, MAGE-A3, MAGE-A9, MAGE-A12, GAGE1, CT-45, SSX-1, and SSX-2), tumor suppressor (TS) genes (p16 and RASSF1A), and genes encoding epigenetic modifiers (DNMT1, DNMT3A, DNMT3B, EZH2, EED, and SUZ12), aberrantly expressed in human malignant diseases. Pyrosequencing techniques were used to quantitate DNA methylation levels in LINE1, NBL2, and D4Z4 repetitive sequences and promoter methylation status of differentially regulated genes. Results of these analyses were compared with a standardized panel of normal lung tissues. RESULTS Pulmonary metastases exhibited histologically related and patient-specific global DNA demethylation. Significant interpatient heterogeneity of gene expression was observed even among patients with similar tumor histologic features. Epigenetic signatures appeared consistent among metastases from the same patient, irrespective of the time of resection (synchronous/metachronous) or the anatomic location. EZH2, EED, and SUZ12 (core components of Polycomb repressive complex-2 [PRC-2]) were upregulated in the majority of metastases. CONCLUSIONS Pulmonary metastases exhibit patient-specific epigenetic clonality, which may be exploited for precision therapies targeting aberrant CT or TS gene expression. PRC-2 may be a shared target for epigenetic therapy of pulmonary metastases.
Collapse
Affiliation(s)
- Emily S Reardon
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Julie A Hong
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - David M Straughan
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Saïd C Azoury
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mary Zhang
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - David S Schrump
- Thoracic Epigenetics Laboratory, Thoracic and GI Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
47
|
Deeb A, Haque SU, Olowokure O. Pulmonary metastases in pancreatic cancer, is there a survival influence? J Gastrointest Oncol 2015; 6:E48-51. [PMID: 26029466 DOI: 10.3978/j.issn.2078-6891.2014.114] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 12/08/2014] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer is known to be one of the most lethal cancers. The majority of patients present with advanced stage disease, making curative approach unachievable. In untreated patients, the median survival does not exceed 6 months in metastatic disease and 10 months in locally advanced disease. Furthermore, the 5-year survival rate remains poor even in patients with early stage disease who are surgical candidates. The detrimental outcome is related to the high potency of developing metastasis which can be detected at diagnosis, when the disease progresses or relapses after surgery. Although the liver is the most common site of pancreatic cancer metastases, the cancer can escape the liver in some cases and metastasize to the lung or other distant organs. The involvement of some sites not others might reflect subgroups of this cancer with different molecular backgrounds. Identifying these groups may have utility in determining prognosis and stratifying treatment for patients.
Collapse
Affiliation(s)
- Ayham Deeb
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio-45229, USA
| | - Sulsal-Ul Haque
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio-45229, USA
| | - Olugbenga Olowokure
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, Ohio-45229, USA
| |
Collapse
|
48
|
Loroch S, Schommartz T, Brune W, Zahedi RP, Sickmann A. Multidimensional electrostatic repulsion–hydrophilic interaction chromatography (ERLIC) for quantitative analysis of the proteome and phosphoproteome in clinical and biomedical research. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:460-8. [DOI: 10.1016/j.bbapap.2015.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/10/2015] [Accepted: 01/15/2015] [Indexed: 11/29/2022]
|
49
|
Cantor DI, Nice EC, Baker MS. Recent findings from the human proteome project: opening the mass spectrometry toolbox to advance cancer diagnosis, surveillance and treatment. Expert Rev Proteomics 2015; 12:279-93. [DOI: 10.1586/14789450.2015.1040770] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
50
|
Blair BG, Wu X, Zahari MS, Mohseni M, Cidado J, Wong HY, Beaver JA, Cochran RL, Zabransky DJ, Croessmann S, Chu D, Toro PV, Cravero K, Pandey A, Park BH. A phosphoproteomic screen demonstrates differential dependence on HER3 for MAP kinase pathway activation by distinct PIK3CA mutations. Proteomics 2014; 15:318-26. [PMID: 25367220 DOI: 10.1002/pmic.201400342] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/02/2014] [Accepted: 10/29/2014] [Indexed: 11/07/2022]
Abstract
The PIK3CA gene encodes for the p110 alpha isoform of PI3 kinase and is one of the most frequently mutated oncogenes in human cancers. However, the mechanisms by which PIK3CA mutations activate cell signaling are not fully understood. Here we used a phosphoproteomic approach to compare differential phosphorylation patterns between human breast epithelial cells and two isogenic somatic cell knock in derivatives, each harboring a distinct PIK3CA mutation. We demonstrated differential phosphorylation patterns between isogenic cell lines containing a PIK3CA helical domain mutation (E545K) compared to cells with a PIK3CA kinase domain mutation (H1047R). In particular, the receptor tyrosine kinase, HER3, showed increased phosphorylation at tyrosine 1328 in H1047R cells versus E545K cells. Genetic studies using shRNA demonstrated that H1047R cells have a profound decrease in growth factor independent proliferation upon HER3 knock down, but this effect was attenuated in E545K cells. In addition, HER3 knock down led to reductions in both PI3 kinase and MAP kinase pathway activation in H1047R cells, but in E545K cells only PI3 kinase pathway diminution was observed. These studies demonstrate the power of using paired isogenic cell lines for proteomic analysis to gain new insights into oncogenic signal transduction pathways.
Collapse
Affiliation(s)
- Brian G Blair
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|