Your browser doesn't support javascript.
loading
Kinase activities in pancreatic ductal adenocarcinoma with prognostic and therapeutic avenues.
Vallés-Martí, Andrea; de Goeij-de Haas, Richard R; Henneman, Alex A; Piersma, Sander R; Pham, Thang V; Knol, Jaco C; Verheij, Joanne; Dijk, Frederike; Halfwerk, Hans; Giovannetti, Elisa; Jiménez, Connie R; Bijlsma, Maarten F.
Affiliation
  • Vallés-Martí A; Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands.
  • de Goeij-de Haas RR; OncoProteomics Laboratory, Cancer Center Amsterdam, The Netherlands.
  • Henneman AA; Cancer Biology, Cancer Center Amsterdam, The Netherlands.
  • Piersma SR; Pharmacology Laboratory, Cancer Center Amsterdam, The Netherlands.
  • Pham TV; Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands.
  • Knol JC; OncoProteomics Laboratory, Cancer Center Amsterdam, The Netherlands.
  • Verheij J; Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands.
  • Dijk F; OncoProteomics Laboratory, Cancer Center Amsterdam, The Netherlands.
  • Halfwerk H; Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands.
  • Giovannetti E; OncoProteomics Laboratory, Cancer Center Amsterdam, The Netherlands.
  • Jiménez CR; Department of Medical Oncology, Amsterdam University Medical Center, VU University, Amsterdam, The Netherlands.
  • Bijlsma MF; OncoProteomics Laboratory, Cancer Center Amsterdam, The Netherlands.
Mol Oncol ; 2024 Apr 22.
Article in En | MEDLINE | ID: mdl-38650175
ABSTRACT
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with a limited number of known driver mutations but considerable cancer cell heterogeneity. Phosphoproteomics provides a direct read-out of aberrant signaling and the resultant clinically relevant phenotype. Mass spectrometry (MS)-based proteomics and phosphoproteomics were applied to 42 PDAC tumors. Data encompassed over 19 936 phosphoserine or phosphothreonine (pS/T; in 5412 phosphoproteins) and 1208 phosphotyrosine (pY; in 501 phosphoproteins) sites and a total of 3756 proteins. Proteome data identified three distinct subtypes with tumor intrinsic and stromal features. Subsequently, three phospho-subtypes were apparent two tumor intrinsic (Phos1/2) and one stromal (Phos3), resembling known PDAC molecular subtypes. Kinase activity was analyzed by the Integrative iNferred Kinase Activity (INKA) scoring. Phospho-subtypes displayed differential phosphorylation signals and kinase activity, such as FGR and GSK3 activation in Phos1, SRC kinase family and EPHA2 in Phos2, and EGFR, INSR, MET, ABL1, HIPK1, JAK, and PRKCD in Phos3. Kinase activity analysis of an external PDAC cohort supported our findings and underscored the importance of PI3K/AKT and ERK pathways, among others. Interestingly, unfavorable patient prognosis correlated with higher RTK, PAK2, STK10, and CDK7 activity and high proliferation, whereas long survival was associated with MYLK and PTK6 activity, which was previously unknown. Subtype-associated activity profiles can guide therapeutic combination approaches in tumor and stroma-enriched tissues, and emphasize the critical role of parallel signaling pathways. In addition, kinase activity profiling identifies potential disease markers with prognostic significance.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Mol Oncol Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2024 Document type: Article Affiliation country: Netherlands

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Mol Oncol Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2024 Document type: Article Affiliation country: Netherlands
...