Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Pancreatic Cancer | Research

Focal adhesion kinase inhibition synergizes with nab-paclitaxel to target pancreatic ductal adenocarcinoma

Authors: T. Y. S. Le Large, M. F. Bijlsma, B. El Hassouni, G. Mantini, T. Lagerweij, A. A. Henneman, N. Funel, B. Kok, T. V. Pham, R. de Haas, L. Morelli, J. C. Knol, S. R. Piersma, G. Kazemier, H. W. M. van Laarhoven, E. Giovannetti, C. R. Jimenez

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

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.
Appendix
Available only for authorised users
Literature
15.
go back to reference Furukawa T, Duguid W, Rosenberg L, Viallet J, Galloway D, Tsao M. Long-term culture and immortalization of epithelial cells from normal adult human pancreatic ducts transfected by the E6E7 gene of human papilloma virus 16. Am J Pathol. 1996;148(6):1763–70.PubMedPubMedCentral Furukawa T, Duguid W, Rosenberg L, Viallet J, Galloway D, Tsao M. Long-term culture and immortalization of epithelial cells from normal adult human pancreatic ducts transfected by the E6E7 gene of human papilloma virus 16. Am J Pathol. 1996;148(6):1763–70.PubMedPubMedCentral
24.
go back to reference Cavazzoni A, Monica S La, Alfieri R, et al. Enhanced efficacy of AKT and FAK kinase combined inhibition in squamous cell lung carcinomas with stable reduction in PTEN 2017;8(32):53068–53083. Cavazzoni A, Monica S La, Alfieri R, et al. Enhanced efficacy of AKT and FAK kinase combined inhibition in squamous cell lung carcinomas with stable reduction in PTEN 2017;8(32):53068–53083.
26.
go back to reference Le Large TYS, el Hassouni B, Funel N, et al. Proteomic analysis of gemcitabine- resistant pancreatic cancer cells reveals that microtubule-associated protein 2 upregulation associates with taxane treatment. Ther Adv Med Oncol. 2019;11:1–17. https://doi.org/10.1177/https.CrossRef Le Large TYS, el Hassouni B, Funel N, et al. Proteomic analysis of gemcitabine- resistant pancreatic cancer cells reveals that microtubule-associated protein 2 upregulation associates with taxane treatment. Ther Adv Med Oncol. 2019;11:1–17. https://​doi.​org/​10.​1177/​https.CrossRef
29.
Metadata
Title
Focal adhesion kinase inhibition synergizes with nab-paclitaxel to target pancreatic ductal adenocarcinoma
Authors
T. Y. S. Le Large
M. F. Bijlsma
B. El Hassouni
G. Mantini
T. Lagerweij
A. A. Henneman
N. Funel
B. Kok
T. V. Pham
R. de Haas
L. Morelli
J. C. Knol
S. R. Piersma
G. Kazemier
H. W. M. van Laarhoven
E. Giovannetti
C. R. Jimenez
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01892-z

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine