LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 369

Search options

  1. Article ; Online: Proteome-based molecular subtyping and therapeutic target prediction in gastric cancer.

    Hu, Changyuan / Song, Jiangning / Kwok, Terry / Nguyen, Elizabeth V / Shen, Xian / Daly, Roger J

    Molecular oncology

    2024  

    Abstract: Different molecular classifications for gastric cancer (GC) have been proposed based on multi-omics platforms with the long-term goal of improved precision treatment. However, the GC (phospho)proteome remains incompletely characterized, particularly at ... ...

    Abstract Different molecular classifications for gastric cancer (GC) have been proposed based on multi-omics platforms with the long-term goal of improved precision treatment. However, the GC (phospho)proteome remains incompletely characterized, particularly at the level of tyrosine phosphorylation. In addition, previous multiomics-based stratification of patient cohorts has lacked identification of corresponding cell line models and comprehensive validation of broad or subgroup-selective therapeutic targets. To address these knowledge gaps, we applied a reverse approach, undertaking the most comprehensive (phospho)proteomic analysis of GC cell lines to date and cross-validating this using publicly available data. Mass spectrometry (MS)-based (phospho)proteomic and tyrosine phosphorylation datasets were subjected to individual or integrated clustering to identify subgroups that were subsequently characterized in terms of enriched molecular processes and pathways. Significant congruence was detected between cell line proteomic and specific patient-derived transcriptomic subclassifications. Many protein kinases exhibiting 'outlier' expression or phosphorylation in the cell line dataset exhibited genomic aberrations in patient samples and association with poor prognosis, with casein kinase I isoform delta/epsilon (CSNK1D/E) being experimentally validated as potential therapeutic targets. Src family kinases were predicted to be commonly hyperactivated in GC cell lines, consistent with broad sensitivity to the next-generation Src inhibitor eCF506. In addition, phosphoproteomic and integrative clustering segregated the cell lines into two subtypes, with epithelial-mesenchyme transition (EMT) and proliferation-associated processes enriched in one, designated the EMT subtype, and metabolic pathways, cell-cell junctions, and the immune response dominating the features of the other, designated the metabolism subtype. Application of kinase activity prediction algorithms and interrogation of gene dependency and drug sensitivity databases predicted that the mechanistic target of rapamycin kinase (mTOR) and dual specificity mitogen-activated protein kinase kinase 2 (MAP2K2) represented potential therapeutic targets for the EMT and metabolism subtypes, respectively, and this was confirmed using selective inhibitors. Overall, our study provides novel, in-depth insights into GC proteomics, kinomics, and molecular taxonomy and reveals potential therapeutic targets that could provide the basis for precision treatments.
    Language English
    Publishing date 2024-04-16
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2415106-3
    ISSN 1878-0261 ; 1574-7891
    ISSN (online) 1878-0261
    ISSN 1574-7891
    DOI 10.1002/1878-0261.13654
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Preface: Novel Therapeutic Targets and Biomarkers in Prostate Cancer.

    Horvath, Lisa G / Taylor, Renea A / Daly, Roger J

    Critical reviews in oncogenesis

    2022  Volume 27, Issue 1, Page(s) vii–ix

    Abstract: Preface: CRO special issue: Novel Therapeutic Targets and Biomarkers in Prostate Cancer. ...

    Abstract Preface: CRO special issue: Novel Therapeutic Targets and Biomarkers in Prostate Cancer.
    MeSH term(s) Biomarkers ; Biomarkers, Tumor/genetics ; Humans ; Male ; Prostatic Neoplasms/diagnosis ; Prostatic Neoplasms/genetics ; Prostatic Neoplasms/therapy
    Chemical Substances Biomarkers ; Biomarkers, Tumor
    Language English
    Publishing date 2022-08-22
    Publishing country United States
    Document type Journal Article
    ZDB-ID 1036388-9
    ISSN 0893-9675
    ISSN 0893-9675
    DOI 10.1615/CritRevOncog.2022043661
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: Novel Therapeutic Targets and Biomarkers Associated with Prostate Cancer-Associated Fibroblasts (CAFs).

    Clark, Kimberley C / Wu, Yunjian / Taylor, Renea A / Daly, Roger J

    Critical reviews in oncogenesis

    2022  Volume 27, Issue 1, Page(s) 1–24

    Abstract: Despite advances in treatment, prostate cancer remains a significant cause of morbidity and mortality worldwide. While the vast majority of prostate cancer research has centered on malignant epithelial cells, the tumor mi croenvironment (TME) has ... ...

    Abstract Despite advances in treatment, prostate cancer remains a significant cause of morbidity and mortality worldwide. While the vast majority of prostate cancer research has centered on malignant epithelial cells, the tumor mi croenvironment (TME) has recently become increasingly recognized as an important regulator of tumor progression and response to treatment. Among the diverse cell types within the TME, stromal fibroblasts, in particular cancer-associated fibroblasts (CAFs), play an important role in prostate cancer progression. This is highlighted by the prognostic value of CAF markers in prostate cancer, which can predict disease recurrence, metastasis, and patient survival. There are also an increasing number of studies that demonstrate the critical role of CAFs in mediating response to specific therapies and CAF signaling pathways as potential therapeutic targets. However, further investigation into the mechanisms that underpin the interactions between cancer cells and CAFs are required to develop novel therapeutic approaches and identify predictive and prognostic biomarkers in CAFs. In this review, we discuss the current knowledge of CAF-dependent regulatory pathways in prostate tumorigenesis and their prognostic and therapeutic potential. Furthermore, we explore the emerging models and technologies that are likely to progress this field of research in terms of discovery and translation to the clinic.
    MeSH term(s) Biomarkers/metabolism ; Cancer-Associated Fibroblasts ; Fibroblasts/metabolism ; Fibroblasts/pathology ; Humans ; Male ; Prostate/metabolism ; Prostate/pathology ; Prostatic Neoplasms/diagnosis ; Prostatic Neoplasms/genetics ; Prostatic Neoplasms/therapy ; Tumor Microenvironment/genetics
    Chemical Substances Biomarkers
    Language English
    Publishing date 2022-08-22
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 1036388-9
    ISSN 0893-9675
    ISSN 0893-9675
    DOI 10.1615/CritRevOncog.2022043478
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Enhancing therapeutic anti-cancer responses by combining immune checkpoint and tyrosine kinase inhibition.

    Daly, Roger J / Scott, Andrew M / Klein, Oliver / Ernst, Matthias

    Molecular cancer

    2022  Volume 21, Issue 1, Page(s) 189

    Abstract: Over the past decade, immune checkpoint inhibitor (ICI) therapy has been established as the standard of care for many types of cancer, but the strategies employed have continued to evolve. Recently, much clinical focus has been on combining targeted ... ...

    Abstract Over the past decade, immune checkpoint inhibitor (ICI) therapy has been established as the standard of care for many types of cancer, but the strategies employed have continued to evolve. Recently, much clinical focus has been on combining targeted therapies with ICI for the purpose of manipulating the immune setpoint. The latter concept describes the equilibrium between factors that promote and those that suppress anti-cancer immunity. Besides tumor mutational load and other cancer cell-intrinsic determinants, the immune setpoint is also governed by the cells of the tumor microenvironment and how they are coerced by cancer cells to support the survival and growth of the tumor. These regulatory mechanisms provide therapeutic opportunities to intervene and reduce immune suppression via application of small molecule inhibitors and antibody-based therapies against (receptor) tyrosine kinases and thereby improve the response to ICIs. This article reviews how tyrosine kinase signaling in the tumor microenvironment can promote immune suppression and highlights how therapeutic strategies directed against specific tyrosine kinases can be used to lower the immune setpoint and elicit more effective anti-tumor immunity.
    MeSH term(s) Humans ; Immune Checkpoint Inhibitors/pharmacology ; Immune Checkpoint Inhibitors/therapeutic use ; Neoplasms/therapy ; Protein-Tyrosine Kinases ; Receptor Protein-Tyrosine Kinases ; Tumor Microenvironment ; Tyrosine/therapeutic use
    Chemical Substances Immune Checkpoint Inhibitors ; Tyrosine (42HK56048U) ; Protein-Tyrosine Kinases (EC 2.7.10.1) ; Receptor Protein-Tyrosine Kinases (EC 2.7.10.1)
    Language English
    Publishing date 2022-09-29
    Publishing country England
    Document type Journal Article ; Review ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 2091373-4
    ISSN 1476-4598 ; 1476-4598
    ISSN (online) 1476-4598
    ISSN 1476-4598
    DOI 10.1186/s12943-022-01656-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Proteomics-based interrogation of the kinome and its implications for precision oncology.

    Lim Kam Sian, Terry C C / Chüeh, Anderly C / Daly, Roger J

    Proteomics

    2021  Volume 21, Issue 17-18, Page(s) e2000161

    Abstract: The identification of specific protein kinases as oncogenic drivers in a variety of cancer types, coupled with the clinical success of particular kinase-directed targeted therapies, has cemented the human kinome as an attractive source of "actionable" ... ...

    Abstract The identification of specific protein kinases as oncogenic drivers in a variety of cancer types, coupled with the clinical success of particular kinase-directed targeted therapies, has cemented the human kinome as an attractive source of "actionable" targets for cancer therapy. However, "mining" of the human kinome for precision oncology applications has yet to yield its full potential. This reflects a variety of issues, including oncogenic kinase dysregulation at levels not detectable by genomic sequencing and the uncharacterized nature of a considerable fraction of the kinome. In addition, selective therapeutic targeting of specific kinases requires efficient mapping of total kinome space impacted by candidate small molecule drugs. Fortunately, recent developments in proteomics techniques, particularly in mass spectrometry-based phosphoproteomics and kinomics, provide the necessary technology platforms to address these impediments. Moreover, initiatives such as the Clinical Proteomic Tumour Analysis Consortium have enabled the generation, deposition and integration of genomic, transcriptomic and (phospho)proteomic data for many cancer types, providing unprecedented insights into oncogenic kinases and cancer cell signalling generally. These multi-omic data are identifying novel therapeutic targets, highlighting opportunities for drug re-purposing, and helping assign optimal therapies to specific tumour subtypes, heralding a new era of "enhanced" precision oncology.
    MeSH term(s) Humans ; Mass Spectrometry ; Neoplasms/drug therapy ; Neoplasms/genetics ; Precision Medicine ; Protein Kinase Inhibitors/pharmacology ; Proteomics
    Chemical Substances Protein Kinase Inhibitors
    Language English
    Publishing date 2021-03-08
    Publishing country Germany
    Document type Journal Article ; Review
    ZDB-ID 2032093-0
    ISSN 1615-9861 ; 1615-9853
    ISSN (online) 1615-9861
    ISSN 1615-9853
    DOI 10.1002/pmic.202000161
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article: A Boolean-based machine learning framework identifies predictive biomarkers of HSP90-targeted therapy response in prostate cancer.

    Shin, Sung-Young / Centenera, Margaret M / Hodgson, Joshua T / Nguyen, Elizabeth V / Butler, Lisa M / Daly, Roger J / Nguyen, Lan K

    Frontiers in molecular biosciences

    2023  Volume 10, Page(s) 1094321

    Abstract: Precision medicine has emerged as an important paradigm in oncology, driven by the significant heterogeneity of individual patients' tumour. A key prerequisite for effective implementation of precision oncology is the development of companion biomarkers ... ...

    Abstract Precision medicine has emerged as an important paradigm in oncology, driven by the significant heterogeneity of individual patients' tumour. A key prerequisite for effective implementation of precision oncology is the development of companion biomarkers that can predict response to anti-cancer therapies and guide patient selection for clinical trials and/or treatment. However, reliable predictive biomarkers are currently lacking for many anti-cancer therapies, hampering their clinical application. Here, we developed a novel machine learning-based framework to derive predictive multi-gene biomarker panels and associated expression signatures that accurately predict cancer drug sensitivity. We demonstrated the power of the approach by applying it to identify response biomarker panels for an Hsp90-based therapy in prostate cancer, using proteomic data profiled from prostate cancer patient-derived explants. Our approach employs a rational feature section strategy to maximise model performance, and innovatively utilizes Boolean algebra methods to derive specific expression signatures of the marker proteins. Given suitable data for model training, the approach is also applicable to other cancer drug agents in different tumour settings.
    Language English
    Publishing date 2023-01-19
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2814330-9
    ISSN 2296-889X
    ISSN 2296-889X
    DOI 10.3389/fmolb.2023.1094321
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Integrative characterisation of secreted factors involved in intercellular communication between prostate epithelial or cancer cells and fibroblasts.

    Wu, Yunjian / Clark, Kimberley C / Niranjan, Birunthi / Chüeh, Anderly C / Horvath, Lisa G / Taylor, Renea A / Daly, Roger J

    Molecular oncology

    2023  Volume 17, Issue 3, Page(s) 469–486

    Abstract: Reciprocal interactions between prostate cancer cells and carcinoma-associated fibroblasts (CAFs) mediate cancer development and progression; however, our understanding of the signalling pathways mediating these cellular interactions remains incomplete. ... ...

    Abstract Reciprocal interactions between prostate cancer cells and carcinoma-associated fibroblasts (CAFs) mediate cancer development and progression; however, our understanding of the signalling pathways mediating these cellular interactions remains incomplete. To address this, we defined secretome changes upon co-culture of prostate epithelial or cancer cells with fibroblasts that mimic bi-directional communication in tumours. Using antibody arrays, we profiled conditioned media from mono- and co-cultures of prostate fibroblasts, epithelial and cancer cells, identifying secreted proteins that are upregulated in co-culture compared to mono-culture. Six of these (CXCL10, CXCL16, CXCL6, FST, PDGFAA, IL-17B) were functionally screened by siRNA knockdown in prostate cancer cell/fibroblast co-cultures, revealing a key role for follistatin (FST), a secreted glycoprotein that binds and bioneutralises specific members of the TGF-β superfamily, including activin A. Expression of FST by both cell types was required for the fibroblasts to enhance prostate cancer cell proliferation and migration, whereas FST knockdown in co-culture grafts decreased tumour growth in mouse xenografts. This study highlights the complexity of prostate cancer cell-fibroblast communication, demonstrates that co-culture secretomes cannot be predicted from individual cultures, and identifies FST as a tumour-microenvironment-derived secreted factor that represents a candidate therapeutic target.
    MeSH term(s) Male ; Humans ; Animals ; Mice ; Prostate/pathology ; Cell Communication ; Fibroblasts/metabolism ; Signal Transduction ; Coculture Techniques ; Prostatic Neoplasms/pathology ; Cell Line, Tumor ; Epithelial Cells/metabolism ; Tumor Microenvironment
    Language English
    Publishing date 2023-01-27
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 2415106-3
    ISSN 1878-0261 ; 1574-7891
    ISSN (online) 1878-0261
    ISSN 1574-7891
    DOI 10.1002/1878-0261.13376
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article: Targeting the human kinome for cancer therapy.

    Daly, Roger J

    Critical reviews in oncogenesis

    2012  Volume 17, Issue 1, Page(s) vi–vii

    MeSH term(s) Antineoplastic Combined Chemotherapy Protocols/administration & dosage ; Antineoplastic Combined Chemotherapy Protocols/therapeutic use ; ErbB Receptors/antagonists & inhibitors ; ErbB Receptors/genetics ; ErbB Receptors/metabolism ; Fusion Proteins, bcr-abl/antagonists & inhibitors ; Fusion Proteins, bcr-abl/genetics ; Fusion Proteins, bcr-abl/metabolism ; Humans ; Medical Oncology/methods ; Medical Oncology/trends ; Molecular Targeted Therapy/methods ; Molecular Targeted Therapy/trends ; Neoplasms/drug therapy ; Neoplasms/genetics ; Neoplasms/metabolism ; Protein Kinase Inhibitors/administration & dosage ; Protein Kinase Inhibitors/pharmacology ; Protein Kinases/genetics ; Protein Kinases/metabolism ; Proteome/analysis ; Proteome/genetics ; Proteome/metabolism ; Proto-Oncogene Proteins B-raf/antagonists & inhibitors ; Proto-Oncogene Proteins B-raf/genetics ; Proto-Oncogene Proteins B-raf/metabolism ; Receptor, ErbB-2/antagonists & inhibitors ; Receptor, ErbB-2/genetics ; Receptor, ErbB-2/metabolism
    Chemical Substances Protein Kinase Inhibitors ; Proteome ; Protein Kinases (EC 2.7.-) ; EGFR protein, human (EC 2.7.10.1) ; ERBB2 protein, human (EC 2.7.10.1) ; ErbB Receptors (EC 2.7.10.1) ; Receptor, ErbB-2 (EC 2.7.10.1) ; Fusion Proteins, bcr-abl (EC 2.7.10.2) ; BRAF protein, human (EC 2.7.11.1) ; Proto-Oncogene Proteins B-raf (EC 2.7.11.1)
    Language English
    Publishing date 2012-04-02
    Publishing country United States
    Document type Introductory Journal Article
    ZDB-ID 1036388-9
    ISSN 0893-9675
    ISSN 0893-9675
    DOI 10.1615/critrevoncog.v17.i1.10
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Distinct PEAK3 interactors and outputs expand the signaling potential of the PEAK pseudokinase family.

    Hou, Jianmei / Nguyen, Elizabeth V / Surudoi, Minglyanna / Roy, Michael J / Patel, Onisha / Lucet, Isabelle S / Ma, Xiuquan / Daly, Roger J

    Science signaling

    2022  Volume 15, Issue 722, Page(s) eabj3554

    Abstract: The pseudokinase scaffolds PEAK1 and PEAK2 are implicated in cancer cell migration and metastasis. We characterized the regulation and role of the third family member PEAK3 in cell signaling. Similar to PEAK1 and PEAK2, PEAK3 formed both homotypic and ... ...

    Abstract The pseudokinase scaffolds PEAK1 and PEAK2 are implicated in cancer cell migration and metastasis. We characterized the regulation and role of the third family member PEAK3 in cell signaling. Similar to PEAK1 and PEAK2, PEAK3 formed both homotypic and heterotypic complexes. In addition, like PEAK1, it bound to the adaptors Grb2 and CrkII. However, unlike PEAK1 and PEAK2, homodimerized PEAK3 also interacted with the ARF GTPase-activating protein ASAP1, the E3 ubiquitin ligase Cbl, and the kinase PYK2. Dimerization and subsequent phosphorylation on Tyr
    MeSH term(s) Cell Movement ; Phosphorylation ; Protein-Tyrosine Kinases/metabolism ; Signal Transduction ; src-Family Kinases/metabolism
    Chemical Substances Protein-Tyrosine Kinases (EC 2.7.10.1) ; src-Family Kinases (EC 2.7.10.2)
    Language English
    Publishing date 2022-02-22
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2417226-1
    ISSN 1937-9145 ; 1945-0877
    ISSN (online) 1937-9145
    ISSN 1945-0877
    DOI 10.1126/scisignal.abj3554
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Proteomic characterisation of prostate cancer intercellular communication reveals cell type-selective signalling and TMSB4X-dependent fibroblast reprogramming.

    Wu, Yunjian / Clark, Kimberley C / Nguyen, Elizabeth V / Niranjan, Birunthi / Horvath, Lisa G / Taylor, Renea A / Daly, Roger J

    Cellular oncology (Dordrecht)

    2022  

    Abstract: Background: In prostate cancer, the tumour microenvironment (TME) represents an important regulator of disease progression and response to treatment. In the TME, cancer-associated fibroblasts (CAFs) play a key role in tumour progression, however the ... ...

    Abstract Background: In prostate cancer, the tumour microenvironment (TME) represents an important regulator of disease progression and response to treatment. In the TME, cancer-associated fibroblasts (CAFs) play a key role in tumour progression, however the mechanisms underpinning fibroblast-cancer cell interactions are incompletely resolved. Here, we address this by applying cell type-specific labelling with amino acid precursors (CTAP) and mass spectrometry (MS)-based (phospho)proteomics to prostate cancer for the first time.
    Methods: Reciprocal interactions between PC3 prostate cancer cells co-cultured with WPMY-1 prostatic fibroblasts were characterised using CTAP-MS. Signalling network changes were determined using Metascape and Enrichr and visualised using Cytoscape. Thymosin β4 (TMSB4X) overexpression was achieved via retroviral transduction and assayed by ELISA. Cell motility was determined using Transwell and random cell migration assays and expression of CAF markers by indirect immunofluorescence.
    Results: WPMY-1 cells co-cultured with PC3s demonstrated a CAF-like phenotype, characterised by enhanced PDGFRB expression and alterations in signalling pathways regulating epithelial-mesenchymal transition, cytoskeletal organisation and cell polarisation. In contrast, co-cultured PC3 cells exhibited more modest network changes, with alterations in mTORC1 signalling and regulation of the actin cytoskeleton. The expression of the actin binding protein TMSB4X was significantly decreased in co-cultured WPMY-1 fibroblasts, and overexpression of TMSB4X in fibroblasts decreased migration of co-cultured PC3 cells, reduced fibroblast motility, and protected the fibroblasts from being educated to a CAF-like phenotype by prostate cancer cells.
    Conclusions: This study highlights the potential of CTAP-MS to characterise intercellular communication within the prostate TME and identify regulators of cellular crosstalk such as TMSB4X.
    Language English
    Publishing date 2022-09-28
    Publishing country Netherlands
    Document type Journal Article
    ZDB-ID 2595109-9
    ISSN 2211-3436 ; 1875-8606 ; 2211-3428
    ISSN (online) 2211-3436
    ISSN 1875-8606 ; 2211-3428
    DOI 10.1007/s13402-022-00719-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top