XENTURION, a multidimensional resource of xenografts and tumoroids from metastatic colorectal cancer patients for population-level translational oncology

The breadth and depth at which cancer models are interrogated contribute to successful translation of drug discovery efforts to the clinic. In colorectal cancer (CRC), model availability is limited by a dearth of large-scale collections of patient-derived xenografts (PDXs) and paired tumoroids from metastatic disease, the setting where experimental therapies are typically tested. XENTURION is a unique open-science resource that combines a platform of 129 PDX models and a sister platform of 129 matched PDX-derived tumoroids (PDXTs) from patients with metastatic CRC, with accompanying multidimensional molecular and therapeutic characterization. A PDXT-based population trial with the anti-EGFR antibody cetuximab revealed variable sensitivities that were consistent with clinical response biomarkers, mirrored tumor growth changes in matched PDXs, and recapitulated the outcome of EGFR genetic deletion. Adaptive signals upregulated by EGFR blockade were computationally and functionally prioritized, and inhibition of top candidates increased the magnitude of response to cetuximab. These findings illustrate the probative value and accuracy of large ex vivo and in vivo living biobanks, highlight the importance of cross-platform and cross-methodology systematic validation, and offer avenues for molecularly informed preclinical research.

[1]  Hu Li,et al.  Culture media composition influences patient-derived organoid ability to predict therapeutic responses in gastrointestinal cancers , 2022, JCI insight.

[2]  Kristen M. Naegle,et al.  A path to translation: How 3D patient tumor avatars enable next generation precision oncology. , 2022, Cancer cell.

[3]  H. Clevers,et al.  Patient‐derived tumor organoids predict responses to irinotecan‐based neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer , 2022, International journal of cancer.

[4]  L. Trusolino,et al.  Towards precision oncology with patient-derived xenografts , 2022, Nature Reviews Clinical Oncology.

[5]  R. Jackstadt,et al.  From organoids to bedside: Advances in modeling, decoding and targeting of colorectal cancer , 2022, International journal of cancer.

[6]  M. Stratton,et al.  Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors , 2022, Nature Cancer.

[7]  Toshiro Sato,et al.  Organoid screening reveals epigenetic vulnerabilities in human colorectal cancer , 2022, Nature Chemical Biology.

[8]  Gabor T. Marth,et al.  A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology , 2022, Nature Cancer.

[9]  Bernd,et al.  The drug-induced phenotypic landscape of colorectal cancer organoids , 2021, Nature Communications.

[10]  M. Michael,et al.  Organoids as a Robust Preclinical Model for Precision Medicine in Colorectal Cancer: A Systematic Review , 2021, Annals of Surgical Oncology.

[11]  Benjamin J. Raphael,et al.  Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidates for targeted treatment , 2021, Nature Communications.

[12]  Xiaochen Bo,et al.  clusterProfiler 4.0: A universal enrichment tool for interpreting omics data , 2021, Innovation.

[13]  E. Cuppen,et al.  Prospective experimental treatment of colorectal cancer patients based on organoid drug responses , 2021, ESMO open.

[14]  L. Trusolino,et al.  Rational Treatment of Metastatic Colorectal Cancer: A Reverse Tale of Men, Mice, and Culture Dishes. , 2021, Cancer Discovery.

[15]  Hayley E. Francies,et al.  Cancer research needs a better map , 2021, Nature.

[16]  R. Tothill,et al.  CUP-AI-Dx: A tool for inferring cancer tissue of origin and molecular subtype using RNA gene-expression data and artificial intelligence , 2020, EBioMedicine.

[17]  I. Spiteri,et al.  Colorectal cancer residual disease at maximal response to EGFR blockade displays a druggable Paneth cell–like phenotype , 2020, Science Translational Medicine.

[18]  K. Neufeld,et al.  Oncogenic Serine 45-Deleted β-Catenin Remains Susceptible to Wnt Stimulation and APC Regulation in Human Colonocytes , 2020, Cancers.

[19]  R. Lothe,et al.  Patient-Derived Organoids from Multiple Colorectal Cancer Liver Metastases Reveal Moderate Intra-patient Pharmacotranscriptomic Heterogeneity , 2020, Clinical Cancer Research.

[20]  Junjie Peng,et al.  Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. , 2019, Cell stem cell.

[21]  E. Cuppen,et al.  Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients , 2019, Science Translational Medicine.

[22]  Emanuel J. V. Gonçalves,et al.  Prioritization of cancer therapeutic targets using CRISPR–Cas9 screens , 2019, Nature.

[23]  M. Boerries,et al.  Heterogeneous pathway activation and drug response modelled in colorectal-tumor-derived 3D cultures , 2019, PLoS genetics.

[24]  H. Clevers,et al.  Studying cellular heterogeneity and drug sensitivity in colorectal cancer using organoid technology. , 2018, Current opinion in genetics & development.

[25]  S. Francescon,et al.  An Update of Efficacy and Safety of Cetuximab in Metastatic Colorectal Cancer: A Narrative Review , 2018, Advances in Therapy.

[26]  Todd R. Golub,et al.  Improved estimation of cancer dependencies from large-scale RNAi screens using model-based normalization and data integration , 2018, bioRxiv.

[27]  Andrea Sottoriva,et al.  Patient-derived organoids model treatment response of metastatic gastrointestinal cancers , 2018, Science.

[28]  Ritika Kundra,et al.  Clinical Sequencing Defines the Genomic Landscape of Metastatic Colorectal Cancer. , 2018, Cancer cell.

[29]  R. Lothe,et al.  CMScaller: an R package for consensus molecular subtyping of colorectal cancer pre-clinical models , 2017, Scientific Reports.

[30]  Steven B. Neuhauser,et al.  PDX-MI: Minimal Information for Patient-Derived Tumor Xenograft Models. , 2017, Cancer research.

[31]  Byung Chun Kim,et al.  KRAS and PIK3CA mutations in colorectal adenocarcinomas correlate with aggressive histological features and behavior. , 2017, Human pathology.

[32]  B. Maček,et al.  WIPI3 and WIPI4 β-propellers are scaffolds for LKB1-AMPK-TSC signalling circuits in the control of autophagy , 2017, Nature Communications.

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

[34]  Eivind Hovig,et al.  Personal Cancer Genome Reporter: variant interpretation report for precision oncology , 2017, bioRxiv.

[35]  Catherine L. Worth,et al.  Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors , 2017, Nature Communications.

[36]  Hans Clevers,et al.  Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening , 2016, eLife.

[37]  Emanuel J. V. Gonçalves,et al.  A Landscape of Pharmacogenomic Interactions in Cancer , 2016, Cell.

[38]  Toshio Uraoka,et al.  A Colorectal Tumor Organoid Library Demonstrates Progressive Loss of Niche Factor Requirements during Tumorigenesis. , 2016, Cell stem cell.

[39]  V. Torri,et al.  Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. , 2016, The Lancet. Oncology.

[40]  Dirk Schumacher,et al.  Assay Establishment and Validation of a High-Throughput Screening Platform for Three-Dimensional Patient-Derived Colon Cancer Organoid Cultures , 2016, Journal of biomolecular screening.

[41]  Mari Mino-Kenudson,et al.  Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal Cancer. , 2016, Cancer discovery.

[42]  R. Scharpf,et al.  The Genomic Landscape of Response to EGFR Blockade in Colorectal Cancer , 2015, Nature.

[43]  Jeffrey S. Morris,et al.  The Consensus Molecular Subtypes of Colorectal Cancer , 2015, Nature Medicine.

[44]  Ron Bose,et al.  HER2 activating mutations are targets for colorectal cancer treatment. , 2015, Cancer discovery.

[45]  Hayley E. Francies,et al.  Prospective Derivation of a Living Organoid Biobank of Colorectal Cancer Patients , 2015, Cell.

[46]  G. Inghirami,et al.  Stromal contribution to the colorectal cancer transcriptome , 2015, Nature Genetics.

[47]  Takanori Kanai,et al.  Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids , 2015, Nature Medicine.

[48]  Camille Stephan-Otto Attolini,et al.  Stromal gene expression defines poor-prognosis subtypes in colorectal cancer , 2015, Nature Genetics.

[49]  Qing-Yu He,et al.  DOSE: an R/Bioconductor package for disease ontology semantic and enrichment analysis , 2015, Bioinform..

[50]  Sabine Tejpar,et al.  IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti-EGFR therapies , 2015, Science Translational Medicine.

[51]  Pieter Wesseling,et al.  DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly , 2014, Genome research.

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

[53]  Mauricio O. Carneiro,et al.  From FastQ Data to High‐Confidence Variant Calls: The Genome Analysis Toolkit Best Practices Pipeline , 2013, Current protocols in bioinformatics.

[54]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[55]  E. Van Cutsem,et al.  Massively Parallel Tumor Multigene Sequencing to Evaluate Response to Panitumumab in a Randomized Phase III Study of Metastatic Colorectal Cancer , 2013, Clinical Cancer Research.

[56]  Wei Li,et al.  RSeQC: quality control of RNA-seq experiments , 2012, Bioinform..

[57]  Steven J. M. Jones,et al.  Comprehensive molecular characterization of human colon and rectal cancer , 2012, Nature.

[58]  Johannes E. Schindelin,et al.  Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.

[59]  R. Morgan Human tumor xenografts: the good, the bad, and the ugly. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[60]  Elena Baralis,et al.  LAS: A Software Platform to Support Oncological Data Management , 2012, Journal of Medical Systems.

[61]  Davide Corà,et al.  A molecularly annotated platform of patient-derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer. , 2011, Cancer discovery.

[62]  Matko Bosnjak,et al.  REVIGO Summarizes and Visualizes Long Lists of Gene Ontology Terms , 2011, PloS one.

[63]  T. Vary,et al.  Hyperoxia inhibits protein synthesis and increases eIF2α phosphorylation in the newborn rat lung. , 2010, American journal of physiology. Lung cellular and molecular physiology.

[64]  J. Taylor,et al.  HDAC6 controls autophagosome maturation essential for ubiquitin‐selective quality‐control autophagy , 2010, The EMBO journal.

[65]  E. Van Cutsem,et al.  Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. , 2009, The New England journal of medicine.

[66]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[67]  F. Kelly Effect of hyperoxic exposure on protein synthesis in the rat. , 1988, The Biochemical journal.

[68]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[69]  Y. Agazie,et al.  The signaling and transformation potency of the overexpressed HER2 protein is dependent on the normally-expressed EGFR. , 2012, Cellular signalling.