Skip to main content
Top
Published in: Molecular Cancer 1/2017

Open Access 01-12-2017 | Letter to the Editor

Generation of stable PDX derived cell lines using conditional reprogramming

Authors: Alexandra Borodovsky, Travis J. McQuiston, Daniel Stetson, Ambar Ahmed, David Whitston, Jingwen Zhang, Michael Grondine, Deborah Lawson, Sharon S. Challberg, Michael Zinda, Brian A. Pollok, Brian A. Dougherty, Celina M. D’Cruz

Published in: Molecular Cancer | Issue 1/2017

Login to get access

Abstract

Efforts to develop effective cancer therapeutics have been hindered by a lack of clinically predictive preclinical models which recapitulate this complex disease. Patient derived xenograft (PDX) models have emerged as valuable tools for translational research but have several practical limitations including lack of sustained growth in vitro. In this study, we utilized Conditional Reprogramming (CR) cell technology- a novel cell culture system facilitating the generation of stable cultures from patient biopsies- to establish PDX-derived cell lines which maintain the characteristics of the parental PDX tumor. Human lung and ovarian PDX tumors were successfully propagated using CR technology to create stable explant cell lines (CR-PDX). These CR-PDX cell lines maintained parental driver mutations and allele frequency without clonal drift. Purified CR-PDX cell lines were amenable to high throughput chemosensitivity screening and in vitro genetic knockdown studies. Additionally, re-implanted CR-PDX cells proliferated to form tumors that retained the growth kinetics, histology, and drug responses of the parental PDX tumor. CR technology can be used to generate and expand stable cell lines from PDX tumors without compromising fundamental biological properties of the model. It offers the ability to expand PDX cells in vitro for subsequent 2D screening assays as well as for use in vivo to reduce variability, animal usage and study costs. The methods and data detailed here provide a platform to generate physiologically relevant and predictive preclinical models to enhance drug discovery efforts.
Appendix
Available only for authorised users
Literature
2.
go back to reference Liu X, Ory V, Chapman S, Yuan H, Albanese C, Kallakury B, Timofeeva OA, Nealon C, Dakic A, Simic V, et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol. 2012;180(2):599–607.CrossRefPubMedPubMedCentral Liu X, Ory V, Chapman S, Yuan H, Albanese C, Kallakury B, Timofeeva OA, Nealon C, Dakic A, Simic V, et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol. 2012;180(2):599–607.CrossRefPubMedPubMedCentral
3.
go back to reference Suprynowicz FA, Upadhyay G, Krawczyk E, Kramer SC, Hebert JD, Liu X, Yuan H, Cheluvaraju C, Clapp PW, Boucher RC Jr, et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc Natl Acad Sci U S A. 2012;109(49):20035–40.CrossRefPubMedPubMedCentral Suprynowicz FA, Upadhyay G, Krawczyk E, Kramer SC, Hebert JD, Liu X, Yuan H, Cheluvaraju C, Clapp PW, Boucher RC Jr, et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc Natl Acad Sci U S A. 2012;109(49):20035–40.CrossRefPubMedPubMedCentral
4.
go back to reference Panaccione A, Chang MT, Carbone BE, Guo Y, Moskaluk CA, Virk RK, Chiriboga L, Prasad ML, Judson B, Mehra S, et al. NOTCH1 and SOX10 are essential for proliferation and radiation resistance of cancer stem-like cells in adenoid cystic carcinoma. Clin Cancer Res. 2016;22(8):2083–95.CrossRefPubMedPubMedCentral Panaccione A, Chang MT, Carbone BE, Guo Y, Moskaluk CA, Virk RK, Chiriboga L, Prasad ML, Judson B, Mehra S, et al. NOTCH1 and SOX10 are essential for proliferation and radiation resistance of cancer stem-like cells in adenoid cystic carcinoma. Clin Cancer Res. 2016;22(8):2083–95.CrossRefPubMedPubMedCentral
5.
go back to reference Yuan H, Myers S, Wang J, Zhou D, Woo JA, Kallakury B, Ju A, Bazylewicz M, Carter YM, Albanese C, et al. Use of reprogrammed cells to identify therapy for respiratory papillomatosis. N Engl J Med. 2012;367(13):1220–7.CrossRefPubMedPubMedCentral Yuan H, Myers S, Wang J, Zhou D, Woo JA, Kallakury B, Ju A, Bazylewicz M, Carter YM, Albanese C, et al. Use of reprogrammed cells to identify therapy for respiratory papillomatosis. N Engl J Med. 2012;367(13):1220–7.CrossRefPubMedPubMedCentral
6.
go back to reference Rhyasen GW, Hattersley MM, Yao Y, Dulak A, Wang W, Petteruti P, Dale IL, Boiko S, Cheung T, Zhang J, et al. AZD5153: a novel bivalent BET Bromodomain inhibitor highly active against hematologic malignancies. Mol Cancer Ther. 2016;15(11):2563–74.CrossRefPubMed Rhyasen GW, Hattersley MM, Yao Y, Dulak A, Wang W, Petteruti P, Dale IL, Boiko S, Cheung T, Zhang J, et al. AZD5153: a novel bivalent BET Bromodomain inhibitor highly active against hematologic malignancies. Mol Cancer Ther. 2016;15(11):2563–74.CrossRefPubMed
7.
go back to reference Henry RE, Barry ER, Castriotta L, Ladd B, Markovets A, Beran G, Ren Y, Zhou F, Adam A, Zinda M, et al. Acquired savolitinib resistance in non-small cell lung cancer arises via multiple mechanisms that converge on MET-independent mTOR and MYC activation. Oncotarget. 2016;7(36):57651–70.CrossRefPubMedPubMedCentral Henry RE, Barry ER, Castriotta L, Ladd B, Markovets A, Beran G, Ren Y, Zhou F, Adam A, Zinda M, et al. Acquired savolitinib resistance in non-small cell lung cancer arises via multiple mechanisms that converge on MET-independent mTOR and MYC activation. Oncotarget. 2016;7(36):57651–70.CrossRefPubMedPubMedCentral
8.
go back to reference Davies BR, Logie A, McKay JS, Martin P, Steele S, Jenkins R, Cockerill M, Cartlidge S, Smith PD. AZD6244 (ARRY-142886), a potent inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 kinases: mechanism of action in vivo, pharmacokinetic/pharmacodynamic relationship, and potential for combination in preclinical models. Mol Cancer Ther. 2007;6(8):2209–19.CrossRefPubMed Davies BR, Logie A, McKay JS, Martin P, Steele S, Jenkins R, Cockerill M, Cartlidge S, Smith PD. AZD6244 (ARRY-142886), a potent inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 kinases: mechanism of action in vivo, pharmacokinetic/pharmacodynamic relationship, and potential for combination in preclinical models. Mol Cancer Ther. 2007;6(8):2209–19.CrossRefPubMed
9.
go back to reference Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487(7408):500–4.CrossRefPubMedPubMedCentral Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487(7408):500–4.CrossRefPubMedPubMedCentral
10.
go back to reference Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via Stromal reciprocation. Cell. 2016;165(7):1818.CrossRefPubMedPubMedCentral Tape CJ, Ling S, Dimitriadi M, McMahon KM, Worboys JD, Leong HS, Norrie IC, Miller CJ, Poulogiannis G, Lauffenburger DA, et al. Oncogenic KRAS regulates tumor cell signaling via Stromal reciprocation. Cell. 2016;165(7):1818.CrossRefPubMedPubMedCentral
Metadata
Title
Generation of stable PDX derived cell lines using conditional reprogramming
Authors
Alexandra Borodovsky
Travis J. McQuiston
Daniel Stetson
Ambar Ahmed
David Whitston
Jingwen Zhang
Michael Grondine
Deborah Lawson
Sharon S. Challberg
Michael Zinda
Brian A. Pollok
Brian A. Dougherty
Celina M. D’Cruz
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2017
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-017-0745-1

Other articles of this Issue 1/2017

Molecular Cancer 1/2017 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