37
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Drug screening and genome editing in human pancreatic cancer organoids identifies drug-gene interactions and candidates for off-label therapy

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Summary

          Pancreatic cancer (PDAC) is a highly aggressive malignancy for which the identification of novel therapies is urgently needed. Here, we establish a human PDAC organoid biobank from 31 genetically distinct lines, covering a representative range of tumor subtypes, and demonstrate that these reflect the molecular and phenotypic heterogeneity of primary PDAC tissue. We use CRISPR-Cas9 genome editing and drug screening to characterize drug-gene interactions with ARID1A and BRCA2. We find that missense, but not frameshift, mutations in the PDAC driver gene ARID1A are associated with increased sensitivity to the kinase inhibitors dasatinib (p < 0.0001) and VE-821 (p < 0.0001). We further conduct an automated drug-repurposing screen with 1,172 FDA-approved compounds, identifying 26 compounds that effectively kill PDAC organoids, including 19 chemotherapy drugs currently approved for other cancer types. We validate the activity of these compounds in vitro and in vivo. The in vivo validated hits include emetine and ouabain, compounds that are approved for non-cancer indications and that perturb the ability of PDAC organoids to respond to hypoxia. Our study provides proof-of-concept for advancing precision oncology and for identifying candidates for drug repurposing via genome editing and drug screening in tumor organoid biobanks.

          Graphical abstract

          Highlights

          • Established biobank from 31 genetically distinct human PDAC organoid lines

          • Missense mutations in ARID1A increase sensitivity of PDAC to dastinib and VE-821

          • Automated drug screening identifies compounds that effectively kill PDAC organoids

          • Emetine and ouabain perturb the ability of PDAC cells to respond to hypoxia

          Abstract

          Hirt et al. established a human PDAC organoid biobank to study drug-gene interactions and perform high-throughput drug screening. Using a fully automated organoid screening pipeline, they conduct a drug repurposing screen including 1,172 drugs. The in vivo validated hits include emetine and ouabain, small molecules approved for non-cancer indications. Functional studies demonstrated that both compounds kill PDAC cells by interfering with their ability to respond to hypoxia.

          Related collections

          Most cited references65

          • Record: found
          • Abstract: found
          • Article: not found

          FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer.

          Data are lacking on the efficacy and safety of a combination chemotherapy regimen consisting of oxaliplatin, irinotecan, fluorouracil, and leucovorin (FOLFIRINOX) as compared with gemcitabine as first-line therapy in patients with metastatic pancreatic cancer. We randomly assigned 342 patients with an Eastern Cooperative Oncology Group performance status score of 0 or 1 (on a scale of 0 to 5, with higher scores indicating a greater severity of illness) to receive FOLFIRINOX (oxaliplatin, 85 mg per square meter of body-surface area; irinotecan, 180 mg per square meter; leucovorin, 400 mg per square meter; and fluorouracil, 400 mg per square meter given as a bolus followed by 2400 mg per square meter given as a 46-hour continuous infusion, every 2 weeks) or gemcitabine at a dose of 1000 mg per square meter weekly for 7 of 8 weeks and then weekly for 3 of 4 weeks. Six months of chemotherapy were recommended in both groups in patients who had a response. The primary end point was overall survival. The median overall survival was 11.1 months in the FOLFIRINOX group as compared with 6.8 months in the gemcitabine group (hazard ratio for death, 0.57; 95% confidence interval [CI], 0.45 to 0.73; P<0.001). Median progression-free survival was 6.4 months in the FOLFIRINOX group and 3.3 months in the gemcitabine group (hazard ratio for disease progression, 0.47; 95% CI, 0.37 to 0.59; P<0.001). The objective response rate was 31.6% in the FOLFIRINOX group versus 9.4% in the gemcitabine group (P<0.001). More adverse events were noted in the FOLFIRINOX group; 5.4% of patients in this group had febrile neutropenia. At 6 months, 31% of the patients in the FOLFIRINOX group had a definitive degradation of the quality of life versus 66% in the gemcitabine group (hazard ratio, 0.47; 95% CI, 0.30 to 0.70; P<0.001). As compared with gemcitabine, FOLFIRINOX was associated with a survival advantage and had increased toxicity. FOLFIRINOX is an option for the treatment of patients with metastatic pancreatic cancer and good performance status. (Funded by the French government and others; ClinicalTrials.gov number, NCT00112658.).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            A simple practice guide for dose conversion between animals and human

            Understanding the concept of extrapolation of dose between species is important for pharmaceutical researchers when initiating new animal or human experiments. Interspecies allometric scaling for dose conversion from animal to human studies is one of the most controversial areas in clinical pharmacology. Allometric approach considers the differences in body surface area, which is associated with animal weight while extrapolating the doses of therapeutic agents among the species. This review provides basic information about translation of doses between species and estimation of starting dose for clinical trials using allometric scaling. The method of calculation of injection volume for parenteral formulation based on human equivalent dose is also briefed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Genomic analyses identify molecular subtypes of pancreatic cancer.

              Integrated genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-β, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. Expression analysis defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) aberrantly differentiated endocrine exocrine (ADEX) that correlate with histopathological characteristics. Squamous tumours are enriched for TP53 and KDM6A mutations, upregulation of the TP63∆N transcriptional network, hypermethylation of pancreatic endodermal cell-fate determining genes and have a poor prognosis. Pancreatic progenitor tumours preferentially express genes involved in early pancreatic development (FOXA2/3, PDX1 and MNX1). ADEX tumours displayed upregulation of genes that regulate networks involved in KRAS activation, exocrine (NR5A2 and RBPJL), and endocrine differentiation (NEUROD1 and NKX2-2). Immunogenic tumours contained upregulated immune networks including pathways involved in acquired immune suppression. These data infer differences in the molecular evolution of pancreatic cancer subtypes and identify opportunities for therapeutic development.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Genom
                Cell Genom
                Cell Genomics
                Elsevier
                2666-979X
                09 February 2022
                09 February 2022
                09 February 2022
                : 2
                : 2
                : 100095
                Affiliations
                [1 ]Institute of Molecular Health Sciences, ETH Zurich, Switzerland
                [2 ]Institute of Pharmacology and Toxicology, University Zurich, Switzerland
                [3 ]NEXUS Personalized Health Technologies, ETH Zurich, Switzerland
                [4 ]SIB Swiss Institute of Bioinformatics, Zurich, Switzerland
                [5 ]Department of Pathology and Molecular Pathology, University Hospital Zurich, Switzerland
                [6 ]Comprehensive Cancer Center, University Hospital Zurich, Switzerland
                [7 ]Department for BioMedical Research, Urology Research laboratory, University Bern, Switzerland
                [8 ]Department of Urology, Inselspital, Bern University Hospital, Switzerland
                [9 ]Discovery Services, Oncotest, Charles River, Freiburg, Germany
                Author notes
                []Corresponding author schwank@ 123456pharma.uzh.ch
                [10]

                Lead contact

                Article
                S2666-979X(22)00012-X 100095
                10.1016/j.xgen.2022.100095
                7612395
                35187519
                dec457cf-d56e-464c-96af-d65dd0ef37f3
                © 2022 The Author(s)

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 30 July 2020
                : 27 January 2021
                : 19 January 2022
                Categories
                Article

                pancreatic cancer,organoids,drug screening,pdx,brca2,arid1a,crispr,hif-1α

                Comments

                Comment on this article