Abstract
Despite recent medical advances, colorectal cancer (CRC) remains the second-leading cause of cancer-related death worldwide. For patients with KRAS wild-type metastatic CRC, the monoclonal antibody cetuximab, which targets the epidermal growth factor receptor (EGFR), is an approved treatment option. However, therapeutic success is often limited by the emergence of drug-resistant cancer cell populations within a few months. Therefore, alternative strategies to effectively target cetuximab-refractory CRC are urgently needed. Here, we sought to identify second-line therapeutic strategies using a CRC cell line with acquired cetuximab resistance as a model. Transcriptomic profiling of the resistant cells identified the apoptosis pathway as a potential therapeutic target, which was supported by their increased susceptibility to BH3-mimetics targeting the anti-apoptotic proteins MCL-1 and BCL-xL under both 2D and 3D culture conditions. These findings were validated in organotypic CRC slice cultures generated from cetuximab-resistant patient-derived xenografts (PDXs). Multiplex immunofluorescence staining demonstrated that BCL-xL inhibition effectively triggered apoptosis in heterogeneous PDX tumor slice models, including models harboring oncogenic BRAF mutations. Our findings suggest that cetuximab-resistant CRC retains apoptotic competence, and that BCL-xL inhibition serves as a robust alternative therapeutic strategy that is largely independent of the tumor mutational profile.
Data availability
The RNA-sequencing data for LIM1215-parental and -R1/R2 cells are available on the GEO website under accession number GSE299943. The RNA-sequencing data for the PDX models are available from Charles River Laboratories (Freiburg, Germany) upon request. Furthermore, uncropped western blots performed in this study are included in this publication. All data and materials reported in this publication will be shared by the lead contact upon request.
Code availability
The codes for the RNA sequencing analysis will be shared by the lead contact upon request.
References
Biller LH, Schrag D. A Review of the Diagnosis and Treatment of Metastatic Colorectal Cancer-Reply. JAMA. 2021;325:2405.
van Emburgh BO, Sartore-Bianchi A, Di Nicolantonio F, Siena S, Bardelli A. Acquired resistance to EGFR-targeted therapies in colorectal cancer. Mol Oncol. 2014;8:1084–94.
Khan KH, Cunningham D, Werner B, Vlachogiannis G, Spiteri I, Heide T, et al. Longitudinal Liquid Biopsy and Mathematical Modeling of Clonal Evolution Forecast Time to Treatment Failure in the PROSPECT-C Phase II Colorectal Cancer Clinical Trial. Cancer Discov. 2018;8:1270–85.
Sforza V, Martinelli E, Ciardiello F, Gambardella V, Napolitano S, Martini G, et al. Mechanisms of resistance to anti-epidermal growth factor receptor inhibitors in metastatic colorectal cancer. World J Gastroenterol. 2016;22:6345–61.
Napolitano S, Martini G, Ciardiello D, Del Tufo S, Martinelli E, Troiani T, et al. Targeting the EGFR signalling pathway in metastatic colorectal cancer. Lancet Gastroenterol Hepatol. 2024;9:664–76.
Lai E, Liscia N, Donisi C, Mariani S, Tolu S, Pretta A, et al. Molecular-Biology-Driven Treatment for Metastatic Colorectal Cancer. Cancers. 2020;12:1214.
Chang Y-Y, Lin P-C, Lin H-H, Lin J-K, Chen W-S, Jiang J-K, et al. Mutation spectra of RAS gene family in colorectal cancer. Am J Surg. 2016;212:537–44.e3.
Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532–6.
Misale S, Arena S, Lamba S, Siravegna G, Lallo A, Hobor S, et al. Blockade of EGFR and MEK intercepts heterogeneous mechanisms of acquired resistance to anti-EGFR therapies in colorectal cancer. Sci Transl Med. 2014;6:224ra26.
Vogler M, Braun Y, Smith VM, Westhoff M-A, Pereira RS, Pieper NM, et al. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther. 2025;10:91.
Fu D, Pfannenstiel L, Demelash A, Phoon YP, Mayell C, Cabrera C, et al. MCL1 nuclear translocation induces chemoresistance in colorectal carcinoma. Cell Death Dis. 2022;13:63.
Scherr A-L, Mock A, Gdynia G, Schmitt N, Heilig CE, Korell F, et al. Identification of BCL-XL as highly active survival factor and promising therapeutic target in colorectal cancer. Cell Death Dis. 2020;11:875.
Zhang L, Ming L, Yu J. BH3 mimetics to improve cancer therapy; mechanisms and examples. Drug Resist Updat. 2007;10:207–17.
Le Z, Ramesh P, Steinmetz M, Medema JP. BH3 Mimetic Sensitivity of Colorectal Cancer Cell Lines in Correlation with Molecular Features Identifies Predictors of Response. Int J Mol Sci. 2021;22:3811.
Jenkins LJ, Luk IY, Chionh F, Tan T, Needham K, Ayton J, et al. BCL-XL inhibitors enhance the apoptotic efficacy of BRAF inhibitors in BRAFV600E colorectal cancer. Cell Death Dis. 2024;15:183.
Davies EJ, Dong M, Gutekunst M, Närhi K, van Zoggel HJAA, Blom S, et al. Capturing complex tumour biology in vitro: histological and molecular characterisation of precision cut slices. Sci Rep. 2015;5:17187.
Rau A, Janssen N, Kühl L, Sell T, Kalmykova S, Mürdter TE, et al. Triple Targeting of HER Receptors Overcomes Heregulin-mediated Resistance to EGFR Blockade in Colorectal Cancer. Mol Cancer Ther. 2022;21:799–809.
Balmanno K, Chell SD, Gillings AS, Hayat S, Cook SJ. Intrinsic resistance to the MEK1/2 inhibitor AZD6244 (ARRY-142886) is associated with weak ERK1/2 signalling and/or strong PI3K signalling in colorectal cancer cell lines. Int J Cancer. 2009;125:2332–41.
Kiessling MK, Curioni-Fontecedro A, Samaras P, Atrott K, Cosin-Roger J, Lang S, et al. Mutant HRAS as novel target for MEK and mTOR inhibitors. Oncotarget. 2015;6:42183–96.
Okamoto K, Zaanan A, Kawakami H, Huang S, Sinicrope FA. Reversal of Mutant KRAS-Mediated Apoptosis Resistance by Concurrent Noxa/Bik Induction and Bcl-2/Bcl-xL Antagonism in Colon Cancer Cells. Mol Cancer Res. 2015;13:659–69.
Okumura K, Huang S, Sinicrope FA. Induction of Noxa sensitizes human colorectal cancer cells expressing Mcl-1 to the small-molecule Bcl-2/Bcl-xL inhibitor, ABT-737. Clin Cancer Res. 2008;14:8132–42.
Pérez-Galán P, Roué G, Villamor N, Campo E, Colomer D. The BH3-mimetic GX15-070 synergizes with bortezomib in mantle cell lymphoma by enhancing Noxa-mediated activation of Bak. Blood. 2007;109:4441–9.
Hu Z, Ding J, Ma Z, Sun R, Seoane JA, Scott Shaffer J, et al. Quantitative evidence for early metastatic seeding in colorectal cancer. Nat Genet. 2019;51:1113–22.
Alharbi F, Almanifi E, Ashrafuzzaman M. Targeting BCL-2 family proteins using BH3 mimetic drugs for cancer therapy: A systematic review of randomized clinical trials. Med Drug Discov. 2024;24:100199. Available from: https://www.sciencedirect.com/science/article/pii/S2590098624000241.
Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
Anders S, Pyl PT, Huber W. HTSeq-a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–9.
Kim D, Paggi JM, Park C, Bennett C, Salzberg SL. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol. 2019;37:907–15.
Pertea M, Pertea GM, Antonescu CM, Chang T-C, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33:290–5.
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
Huang W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009;4:44–57.
Lê S, Josse J, Husson F. FactoMineR: an R package for multivariate analysis. J Stat Softw. 2008;25:1–18.
Blighe, Rana, and Lewis. EnhancedVolcano: Publication-ready volcano plots with enhanced colouring and labeling; 2018. https://github.com/kevinblighe/EnhancedVolcano
Bankhead P, Loughrey MB, Fernández JA, Dombrowski Y, McArt DG, Dunne PD, et al. QuPath: Open source software for digital pathology image analysis. Sci Rep. 2017;7:16878.
Acknowledgements
This work was supported by the Ministry of Science, Research and Arts Baden-Württemberg within the 3R-US project (33-7533-6-1522/2/3). MB is supported by the Deutsche Forschungsgemeinschaft (DFG) – CRC1479 (Project ID 441891347-S1), CRC 1160 (Project ID 256073931-Z02), CRC1453 (Project ID 431984000-S1), TRR167 (Project ID 259373024-Z01), TRR 359 (Project ID 491676693-Z01), TRR 353 (Project ID 471011418-SP02) and FOR 5476 UcarE (Project ID 493802833-P7) and by the German Federal Ministry of Education and Research (BMBF) within the National Decade against Cancer program PM4Onco–FKZ 01ZZ2322A (to MB and PM) and SATURN3 (01KD2206L to MB). Figures 1A and 6A were created with BioRender.com. The funders had no role in study design, data collection and analysis, publication decision, or manuscript preparation. We thank Dr. Oliver Seifert, Alexandra Kraske and Laura Böckmann (University of Stuttgart), as well as Kerstin Willecke (Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology) for their excellent technical support. We thank Kai Hirzel (University of Stuttgart) for 3D-printing the customized molds for tumor embedding, as well as Dr. Zeynab Najafova and Akshay Kanakan (Robert Bosch Center for Tumor Diseases) for providing PANC-1 and AsPC-1 cell lines.
Funding
Open Access funding enabled and organized by Projekt DEAL.
Author information
Authors and Affiliations
Contributions
SA designed and performed the experiments, analyzed the data and prepared the figures. RS and LMW assisted with the cellular assays. MD and JT provided their expertise with slice cultures and multiplex immunofluorescence staining. JS and EO provided the PDX model data and tissues. TLE and SA performed RNA-seq on the LIM1215 cells. TLE, ACB and PM analyzed the RNA-seq data from cells and tissues. MAO designed the study, acquired the funding, and supervised the work with conceptual support from REK, RT, SAJ, MB and FE. SA and MAO wrote the manuscript. RT was involved in preparing the final manuscript. All authors reviewed the manuscript and provided feedback.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics
All animal procedures were performed in an AAALAC-accredited facility and were in accordance with the Guide for the Care and Use of Laboratory Animals of the Society of Laboratory Animals (GV SOLAS). Animal experiments were approved by the Committee on the Ethics of Animal Experiments of the regional council (Regierungspräsidium Freiburg, Abt. Landwirtschaft, Ländlicher Raum, Veterinär- und Lebensmittelwesen permit-#: I-19/02 und G-23/086).
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Edited by Professor Thomas Kaufmann
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Asmanidou, S., Thiel, J., Ekstrom, T.L. et al. BCL-xL as a therapeutic target in cetuximab-refractory colorectal cancer. Cell Death Dis (2026). https://doi.org/10.1038/s41419-026-08434-5
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41419-026-08434-5