Targeting the DNA Damage Response Pathways and Replication Stress in Colorectal Cancer

Purpose Genomic instability is a hallmark of cancer and targeting DNA damage response (DDR) is emerging as a promising therapeutic strategy in different solid tumors. The effectiveness of targeting DDR in colorectal cancer has not been extensively explored. Experimental Design We challenged 112 cell models recapitulating the genomic landscape of metastatic colorectal cancer with ATM, ATR, CHK1, WEE1, and DNA-PK inhibitors, in parallel with chemotherapeutic agents. We focused then on ATR inhibitors (ATRi) and, to identify putative biomarkers of response and resistance, we analyzed at multiple levels colorectal cancer models highly sensitive or resistant to these drugs. Results We found that around 30% of colorectal cancers, including those carrying KRAS and BRAF mutations and unresponsive to targeted agents, are sensitive to at least one DDR inhibitor. By investigating potential biomarkers of response to ATRi, we found that ATRi-sensitive cells displayed reduced phospho-RPA32 foci at basal level, while ATRi-resistant cells showed increased RAD51 foci formation in response to replication stress. Lack of ATM and RAD51C expression was associated with ATRi sensitivity. Analysis of mutational signatures and HRDetect score identified a subgroup of ATRi-sensitive models. Organoids derived from patients with metastatic colorectal cancer recapitulated findings obtained in cell lines. Conclusions In conclusion, a subset of colorectal cancers refractory to current therapies could benefit from inhibitors of DDR pathways and replication stress. A composite biomarker involving phospho-RPA32 and RAD51 foci, lack of ATM and RAD51C expression, as well as analysis of mutational signatures could be used to identify colorectal cancers likely to respond to ATRi.

[1]  M. Parmar,et al.  Inhibition of WEE1 Is Effective in TP53- and RAS-Mutant Metastatic Colorectal Cancer: A Randomized Trial (FOCUS4-C) Comparing Adavosertib (AZD1775) With Active Monitoring , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[2]  M. Oberley,et al.  Homologous Recombination Deficiency Alterations in Colorectal Cancer: Clinical, Molecular, and Prognostic Implications. , 2021, Journal of the National Cancer Institute.

[3]  S. Nik-Zainal,et al.  Mutational signatures: emerging concepts, caveats and clinical applications , 2021, Nature Reviews Cancer.

[4]  J. Soria,et al.  Ceralasertib (AZD6738), an Oral ATR Kinase Inhibitor, in Combination with Carboplatin in Patients with Advanced Solid Tumors: A Phase I Study , 2021, Clinical Cancer Research.

[5]  T. Yap,et al.  Targeting the replication stress response through synthetic lethal strategies in cancer medicine. , 2021, Trends in cancer.

[6]  S. Arena,et al.  Preclinical models as patients’ avatars for precision medicine in colorectal cancer: past and future challenges , 2021, Journal of Experimental & Clinical Cancer Research.

[7]  R. Bernards,et al.  Precision oncology in metastatic colorectal cancer — from biology to medicine , 2021, Nature Reviews Clinical Oncology.

[8]  P. Loadman,et al.  Progress towards a clinically-successful ATR inhibitor for cancer therapy , 2021, Current research in pharmacology and drug discovery.

[9]  J. Schellens,et al.  A first-in-man phase 1 study of the DNA-dependent protein kinase inhibitor peposertib (formerly M3814) in patients with advanced solid tumours , 2020, British Journal of Cancer.

[10]  G. Iliakis,et al.  Replication protein A: a multifunctional protein with roles in DNA replication, repair and beyond , 2020, NAR cancer.

[11]  A. Lau,et al.  Complete loss of ATM function augments replication catastrophe induced by ATR inhibition and gemcitabine in pancreatic cancer models , 2020, British Journal of Cancer.

[12]  N. Tunariu,et al.  Phase I Trial of First-in-Class ATR Inhibitor M6620 (VX-970) as Monotherapy or in Combination With Carboplatin in Patients With Advanced Solid Tumors , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  A. Bardelli,et al.  The DNA Damage Response pathway as a land of therapeutic opportunities for colorectal cancer. , 2020, Annals of oncology : official journal of the European Society for Medical Oncology.

[14]  W. Hahn,et al.  ATM Loss Confers Greater Sensitivity to ATR Inhibition Than PARP Inhibition in Prostate Cancer , 2020, Cancer Research.

[15]  S. Morganella,et al.  A practical framework and online tool for mutational signature analyses show intertissue variation and driver dependencies , 2020, Nature Cancer.

[16]  A. Bardelli,et al.  A Subset of Colorectal Cancers with Cross-Sensitivity to Olaparib and Oxaliplatin , 2019, Clinical Cancer Research.

[17]  M. Linnebacher,et al.  Integrated Biobanking and Tumor Model Establishment of Human Colorectal Carcinoma Provides Excellent Tools for Preclinical Research , 2019, Cancers.

[18]  M. O’Connor,et al.  Pharmacology of the ATM Inhibitor AZD0156: Potentiation of Irradiation and Olaparib Responses Preclinically , 2019, Molecular Cancer Therapeutics.

[19]  L. Saal,et al.  Whole-genome-sequencing of triple negative breast cancers in a population-based clinical study , 2019, Nature Medicine.

[20]  M. Garnett,et al.  Patient-Derived Xenografts and Matched Cell Lines Identify Pharmacogenomic Vulnerabilities in Colorectal Cancer , 2019, Clinical Cancer Research.

[21]  Y. Pommier,et al.  Targeting Topoisomerase I in the Era of Precision Medicine , 2019, Clinical Cancer Research.

[22]  A. Tutt,et al.  A decade of clinical development of PARP inhibitors in perspective , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[23]  A. Bardelli,et al.  Evolving neoantigen profiles in colorectal cancers with DNA repair defects , 2019, Genome Medicine.

[24]  A. Bardelli,et al.  A Genomic Analysis Workflow for Colorectal Cancer Precision Oncology. , 2019, Clinical colorectal cancer.

[25]  R. Plummer,et al.  The DNA Damaging Revolution: PARP Inhibitors and Beyond. , 2019, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.

[26]  E. Van Cutsem,et al.  Maximising clinical benefit with adequate patient management beyond the second line in mCRC , 2019, ESMO Open.

[27]  Ville Mustonen,et al.  The repertoire of mutational signatures in human cancer , 2018, Nature.

[28]  L. K. Teixeira,et al.  DNA replication stress: oncogenes in the spotlight , 2019, Genetics and molecular biology.

[29]  M. Spies,et al.  A structural and dynamic model for the assembly of Replication Protein A on single-stranded DNA , 2018, Nature Communications.

[30]  N. Tunariu,et al.  Ataxia Telangiectasia Mutated Protein Loss and Benefit From Oxaliplatin‐based Chemotherapy in Colorectal Cancer , 2018, Clinical colorectal cancer.

[31]  Kara A. Bernstein,et al.  RAD-ical New Insights into RAD51 Regulation , 2018, Genes.

[32]  C. Caldas,et al.  A RAD51 assay feasible in routine tumor samples calls PARP inhibitor response beyond BRCA mutation , 2018, EMBO molecular medicine.

[33]  G. Mills,et al.  State-of-the-art strategies for targeting the DNA damage response in cancer , 2018, Nature Reviews Clinical Oncology.

[34]  S. Sleijfer,et al.  Pan-cancer whole genome analyses of metastatic solid tumors , 2018, bioRxiv.

[35]  C. Lord,et al.  Directing the use of DDR kinase inhibitors in cancer treatment , 2017, Expert opinion on investigational drugs.

[36]  Konrad Scheffler,et al.  Strelka2: Fast and accurate variant calling for clinical sequencing applications , 2017, bioRxiv.

[37]  Elisa Ficarra,et al.  Selective analysis of cancer-cell intrinsic transcriptional traits defines novel clinically relevant subtypes of colorectal cancer , 2017, Nature Communications.

[38]  Francesca Sperati,et al.  CHK1-targeted therapy to deplete DNA replication-stressed, p53-deficient, hyperdiploid colorectal cancer stem cells , 2017, Gut.

[39]  E. Birney,et al.  HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures , 2017, Nature Medicine.

[40]  J. Guinney,et al.  Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer , 2017, Nature Reviews Cancer.

[41]  Paul Flicek,et al.  The international Genome sample resource (IGSR): A worldwide collection of genome variation incorporating the 1000 Genomes Project data , 2016, Nucleic Acids Res..

[42]  Roland Eils,et al.  Complex heatmaps reveal patterns and correlations in multidimensional genomic data , 2016, Bioinform..

[43]  Xiaoyu Chen,et al.  Manta: rapid detection of structural variants and indels for germline and cancer sequencing applications , 2016, Bioinform..

[44]  A. Vindigni,et al.  Replication stress: getting back on track , 2016, Nature Structural &Molecular Biology.

[45]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences , 2015, F1000Research.

[46]  Saskia D. Hiltemann,et al.  Discriminating somatic and germline mutations in tumor DNA samples without matching normals , 2015, Genome research.

[47]  Marco Beccuti,et al.  The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets , 2015, Nature Communications.

[48]  Frances M. G. Pearl,et al.  Therapeutic opportunities within the DNA damage response , 2015, Nature Reviews Cancer.

[49]  Y. Pommier,et al.  ATR inhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase i inhibitors by disabling DNA replication initiation and fork elongation responses. , 2014, Cancer research.

[50]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[51]  Roland Eils,et al.  circlize implements and enhances circular visualization in R , 2014, Bioinform..

[52]  S. Opiyo,et al.  DNA-PK phosphorylation of RPA32 Ser4/Ser8 regulates replication stress checkpoint activation, fork restart, homologous recombination and mitotic catastrophe. , 2014, DNA repair.

[53]  Neville E. Sanjana,et al.  Improved vectors and genome-wide libraries for CRISPR screening , 2014, Nature Methods.

[54]  Samuel H. Wilson,et al.  Base Excision Repair Defects Invoke Hypersensitivity to PARP Inhibition , 2014, Molecular Cancer Research.

[55]  Youwei Zhang,et al.  Roles of Chk1 in cell biology and cancer therapy , 2014, International journal of cancer.

[56]  N. Mailand,et al.  ATR Prohibits Replication Catastrophe by Preventing Global Exhaustion of RPA , 2013, Cell.

[57]  Y. Bang,et al.  RAD51C-Deficient Cancer Cells Are Highly Sensitive to the PARP Inhibitor Olaparib , 2013, Molecular Cancer Therapeutics.

[58]  Heng Li Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM , 2013, 1303.3997.

[59]  Chao Chen,et al.  dbVar and DGVa: public archives for genomic structural variation , 2012, Nucleic Acids Res..

[60]  C. Cole,et al.  COSMIC: the catalogue of somatic mutations in cancer , 2011, Genome Biology.

[61]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[62]  C. Perou,et al.  Allele-specific copy number analysis of tumors , 2010, Proceedings of the National Academy of Sciences.

[63]  Yves Pommier,et al.  γH2AX and cancer , 2008, Nature Reviews Cancer.

[64]  J. Borowiec,et al.  RPA phosphorylation facilitates mitotic exit in response to mitotic DNA damage , 2008, Proceedings of the National Academy of Sciences.

[65]  Phil Quirke,et al.  Expression of DNA Double-Strand Break Repair Proteins ATM and BRCA1 Predicts Survival in Colorectal Cancer , 2006, Clinical Cancer Research.

[66]  K. Khanna,et al.  ATM, a central controller of cellular responses to DNA damage , 2001, Cell Death and Differentiation.

[67]  Elizabeth M. Smigielski,et al.  dbSNP: the NCBI database of genetic variation , 2001, Nucleic Acids Res..

[68]  S. Jackson,et al.  Regulation of p53 in response to DNA damage , 1999, Oncogene.