Integrated profiling of human pancreatic cancer organoids reveals chromatin accessibility features associated with drug sensitivity

Xiaohan Shi, Yunguang Li, Qiuyue Yuan, Shijie Tang, Shiwei Guo, Yehan Zhang, Juan He, Xiaoyu Zhang, Ming Han, Zhuang Liu, Yiqin Zhu, Suizhi Gao, Huan Wang, Xiongfei Xu, Kailian Zheng, Wei Jing, Luonan Chen, Yong Wang, Gang Jin, Dong Gao, Xiaohan Shi, Yunguang Li, Qiuyue Yuan, Shijie Tang, Shiwei Guo, Yehan Zhang, Juan He, Xiaoyu Zhang, Ming Han, Zhuang Liu, Yiqin Zhu, Suizhi Gao, Huan Wang, Xiongfei Xu, Kailian Zheng, Wei Jing, Luonan Chen, Yong Wang, Gang Jin, Dong Gao

Abstract

Chromatin accessibility plays an essential role in controlling cellular identity and the therapeutic response of human cancers. However, the chromatin accessibility landscape and gene regulatory network of pancreatic cancer are largely uncharacterized. Here, we integrate the chromatin accessibility profiles of 84 pancreatic cancer organoid lines with whole-genome sequencing data, transcriptomic sequencing data and the results of drug sensitivity analysis of 283 epigenetic-related chemicals and 5 chemotherapeutic drugs. We identify distinct transcription factors that distinguish molecular subtypes of pancreatic cancer, predict numerous chromatin accessibility peaks associated with gene regulatory networks, discover regulatory noncoding mutations with potential as cancer drivers, and reveal the chromatin accessibility signatures associated with drug sensitivity. These results not only provide the chromatin accessibility atlas of pancreatic cancer but also suggest a systematic approach to comprehensively understand the gene regulatory network of pancreatic cancer in order to advance diagnosis and potential personalized medicine applications.

Conflict of interest statement

The authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Establishment and histological characterization of…
Fig. 1. Establishment and histological characterization of PDPCOs.
a Overview of the study. A total of 84 PDPCO lines, including PDAC, NEN, IPMN and ACC, were established by samples obtained by surgery or EUS-FNA. Original tissues were used for histological examination and WES/panel sequencing. PDPCOs were subjected to engraftment, histological examination, WGS/WES, RNA-seq, ATAC-seq and drug screening. b Representative brightfield image of PDPCOs generated from normal pancreas, PDAC, IPMN, NEN and ACC tissue. Scale bar, 200 μm. Experiments are repeated at least three times with similar results. c, Representative H&E staining of PDAC primary tumors, organoids, and xenografts (CAS-DAC-13, CAS-DAC-29 and CAS-DAC-30). Scale bar, 50 μm. Experiments are repeated at least three times with similar results. d Representative H&E and alcian blue staining of an IPMN primary tissue, organoid and xenograft (CAS-IPMN-1). Scale bar, 50 μm. Experiments are repeated at least three times with similar results. e Representative H&E, SYP, CHGA and Ki67 staining of NEN primary tissues, organoids and xenografts (CAS-NEN-2 and CAS-NEN-4). CAS-NEN-2 was clinically diagnosed as WHO grade G2, while CAS-NEN-4 was diagnosed as NEC. Scale bar, 50 μm. Experiments are repeated at least three times with similar results. PDPCO, patient-derived pancreatic cancer organoid; PDAC, pancreatic ductal adenocarcinoma; NEN, pancreatic neuroendocrine neoplasm; IPMN, intraductal papillary mucinous neoplasm; ACC, acinar cell carcinoma; EUS-FNA, endoscopic ultrasound-guided fine needle aspiration biopsy.
Fig. 2. WGS analysis of genomic mutations…
Fig. 2. WGS analysis of genomic mutations and alterations in PDPCOs.
Genomic landscape of 70 exocrine PDPCOs, including PDACs, IPMNs, and ACCs (left), and 4 neuroendocrine PDPCOs (right). The figure shows exonic mutations; noncoding mutations in UTR, upstream/downstream, intronic, ncRNA and intergenic regions; and CNVs. CAS-DAC-23 with WES of blood DNA; CAS-NEN-2 without sequencing of blood DNA. PDPCO patient-derived pancreatic cancer organoid, PDAC pancreatic ductal adenocarcinoma, NEN pancreatic neuroendocrine neoplasm, IPMN intraductal papillary mucinous neoplasm, ACC acinar cell carcinoma, CNV copy number variation.
Fig. 3. Transcriptome analysis of PDPCOs.
Fig. 3. Transcriptome analysis of PDPCOs.
a PCA of gene expression for 45 PDPCOs in the discovery cohort: 39 CAS-DACs, 4 CAS-NENs, 1 CAS-IPMN and 1 CAS-ACC. b Heatmap of the four transcriptomic subtypes in the discovery cohort of 40 exocrine PDPCOs (39 CAS-DACs and 1 CAS-IPMN), as revealed by unsupervised NMF clustering. Samples are listed in columns, and signature genes are listed in rows as z-scores. c Identification of biological characteristics in each subtype by previously reported signatures. A P-value < 0.01 was used to determine positive or negative enrichment of published signatures in a subtype (top). Detailed GSEA results are shown at the bottom. d Bar graph showing the results of enrichment analysis of class 4 DEGs by Enrichr. Significance was computed by one-sided hypergeometric test. e Heatmap indicating the expression of glycolysis pathway-related genes across the four subtypes. Samples are listed in rows, and signature genes are listed in columns as z-scores. f External validation of the transcriptomic subtypes in the TCGA PAAD cohort. Samples are listed in rows, and signature genes are listed in columns as z-scores. g Kaplan-Meier survival analysis showing the overall survival characteristics of patients with the four transcriptomic subtypes, as identified in f. P value is determined by using log-rank test. PDPCO, patient-derived pancreatic cancer organoid; PDAC, pancreatic ductal adenocarcinoma; NEN, pancreatic neuroendocrine neoplasm; IPMN, intraductal papillary mucinous neoplasm; ACC, acinar cell carcinoma; NPO, normal pancreatic ductal organoid; PCA, principal component analysis; NMF, nonnegative matrix factorization; GSEA, gene set enrichment analysis; DEG, differentially expressed gene.
Fig. 4. Integrated analysis of RNA-seq and…
Fig. 4. Integrated analysis of RNA-seq and ATAC-seq revealing TF regulons for PDPCO subtypes and putative peak-to-gene links.
a Schematic of the process for generating subtype-specific TF regulons in PDPCOs. Comparison of enriched TF regulons between exocrine and neuroendocrine PDPCOs in b and among the four PDAC transcriptome subtypes in c. The color of each heatmap on the left represents the regulon z-score. The color of each heatmap on the right represents the -log10(P value) of the regulon score. P values associated with one-sided unpaired t test were adjusted for multiple testing using FDR. Representative subtype-specific TF regulons are listed. d Heatmap of 2,257 putative links between ATAC-seq peaks (left) and genes (right) in 41 exocrine PDPCOs (39 PDACs, 1 IPMN, and 1 ACC). Each row represents an individual link between one ATAC-seq peak and one gene. The color of each heatmap represents the z-score for ATAC-seq peak’s accessibility (left) or the z-score for gene expression (right). TF, transcription factor; PDPCO, patient-derived pancreatic cancer organoid; TG, target gene; PDAC, pancreatic ductal adenocarcinoma; FDR, false discovery rate.
Fig. 5. Integrated analysis of noncoding mutations…
Fig. 5. Integrated analysis of noncoding mutations and drug sensitivity in PDPCOs.
a Schematic for identifying potential functional noncoding mutations in 31 exocrine PDPCOs. b Proportions of noncoding mutations located within and outside ATAC peaks. c Dot plot showing fold change in accessibility at ATAC-seq peaks containing noncoding mutations across above 31 exocrine PDPCO. d Box plot showing chromatin accessibility at RIMBP2 putative enhancer and RIMBP2 gene expression across above 31 exocrine PDPCOs. Boxplot show the median (central line), the 25–75% interquartile range (box limits). e Normalized ATAC-seq tracks of RIMBP2 putative enhancer locus in 5 representative samples. The red and gray tracks represent samples with and without mutation of the RIMBP2 putative enhancer, respectively. Black dotted line indicates the position of the mutation, and the predicted enhancer region is highlighted by light blue shading. f Kaplan-Meier analysis of overall survival in the TCGA PAAD cohort stratified by high and low expression of RIMBP2 gene. P value is determined by using log-rank test. g Heatmap of 15,397 putative peak-to-drug links in 39 exocrine PDPCOs. Each row represents an individual link between one ATAC-seq peak and one drug. The color represents the z-score for chromatin accessibility (left) or the z-score for drug AUC (right). Dot plot showing a representative negative correlation in h and a representative positive correlation in i. Significance is computed by Pearson’s correlation coefficients without adjustment. j Normalized ATAC-seq tracks of BAG3 putative enhancer locus in 10 representative samples. The yellow and purple tracks represent samples with and without peaks in BAG3 putative enhancer, respectively. The peak region is highlighted by light blue shading. Comparison between peak high group with peaks in the BAG3 putative enhancer and peak low group without such peaks in the above 10 samples of BAG3 gene expression in k, AUC of 5-FU in l and AUC of PTX in m. Each group contains 5 biologically independent samples. Boxplot show the median (central line), the 25–75% interquartile range (box limits). Significance is computed by two-sided unpaired t test. PDPCO, patient-derived pancreatic cancer organoid; AUC, area under curve; 5-FU, 5-fluorouracil; PTX, paclitaxel.
Fig. 6. Clinically relevant chemosensitivity of PDPCOs…
Fig. 6. Clinically relevant chemosensitivity of PDPCOs with validation in in vivo xenograft models.
a AUCs of 5 chemotherapeutic agents in 39 exocrine PDPCOs (38 CAS-DACs and 1 CAS-IPMN). With respect to each chemotherapeutic agent, the 39 PDPCOs were divided equally into sensitive (blue, AUC lowest 1st-13th), intermediate (yellow, AUC lowest 14th–26th) and resistant (red, AUC lowest 27th–39th) groups. The gray dot indicated CAS-DAC-14. b Diagram of the clinical analysis workflow. c Kaplan-Meier survival analysis showing the recurrence-free survival outcomes of the patients corresponding to the 39 PDPCOs. P value is determined by using log-rank test. Based on the consistency between the clinical adjuvant chemotherapy regimen and chemosensitivity of the matched organoid, three groups were identified: sensitive (n = 10), intermediate (n = 13) and resistant (n = 8). d Representative radiation examination of both the surgical area and liver in the sensitive group (CAS-DAC-24, CE-CT), intermediate group (CAS-DAC-20, CE-MRI), and resistant group (CAS-DAC-22, CE-MRI) at the time of diagnosis and six months post-surgery. Yellow arrow, primary tumor; Blue arrow, hepatic cyst; Red arrow, metastasis. e Drug test of ODX-18 with GEM (n=6), using Vehicle as a control (n=6). Data are presented as mean values + SEM. Statistical Significance was computed by two-sided unpaired t test. Source data are provided as a Source Data file. f Drug test of ODX-20 with GEM (n = 6) and 5-FU (n = 6), using Vehicle as a control (n = 6). Data are presented as mean values + SEM. Statistical Significance was computed by two-sided unpaired t test. Source data are provided as a Source Data file. g Drug test of ODX-36 with GEM (n = 6), 5-FU (n = 6) and PTX (n = 6), using Vehicle as a control (n=6). Data are presented as mean values + SEM. Statistical Significance was computed by two-sided unpaired t test. Source data are provided as a Source Data file. Statistical analysis, ns P ≥ 0.05,*P < 0.05, **P < 0.01, ***P < 0.001. PDPCO, patient-derived pancreatic cancer organoid; AUC, area under curve; GEM, gemcitabine; 5-FU, 5-fluorouracil; PTX, paclitaxel; OXA, oxaliplatin; IRI, irinotecan; CE-CT: contrast-enhanced computed tomography; CE-MRI: contrast-enhanced magnetic resonance imaging.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA: a cancer J. clinicians. 2020;70:7–30.
    1. Collisson EA, Bailey P, Chang DK, Biankin AV. Molecular subtypes of pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2019;16:207–220. doi: 10.1038/s41575-019-0109-y.
    1. Bailey P, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52. doi: 10.1038/nature16965.
    1. Moffitt RA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015;47:1168–1178. doi: 10.1038/ng.3398.
    1. Scarpa A, et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543:65–71. doi: 10.1038/nature21063.
    1. Waddell N, et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495–501. doi: 10.1038/nature14169.
    1. Chan-Seng-Yue M, et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat. Genet. 2020;52:231–240. doi: 10.1038/s41588-019-0566-9.
    1. Tiriac H, et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018;8:1112–1129. doi: 10.1158/-18-0349.
    1. Buenrostro JD, Wu B, Chang HY, Greenleaf WJ. ATAC-seq: A Method for Assaying Chromatin Accessibility Genome-Wide. Curr. Protoc. Mol. Biol. 2015;109:21 29 21–21 29 29. doi: 10.1002/0471142727.mb2129s109.
    1. Brunton H, et al. HNF4A and GATA6 Loss Reveals Therapeutically Actionable Subtypes in Pancreatic Cancer. Cell Rep. 2020;31:107625. doi: 10.1016/j.celrep.2020.107625.
    1. Roe J-S, et al. Enhancer Reprogramming Promotes Pancreatic Cancer Metastasis. Cell. 2017;170:875–888.e820. doi: 10.1016/j.cell.2017.07.007.
    1. Corces MR, et al. The chromatin accessibility landscape of primary human cancers. Sci. (N. Y., N. Y.) 2018;362:eaav1898. doi: 10.1126/science.aav1898.
    1. Rossi G, Manfrin A, Lutolf MP. Progress and potential in organoid research. Nat. Rev. Genet. 2018;19:671–687. doi: 10.1038/s41576-018-0051-9.
    1. Boj SF, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324–338. doi: 10.1016/j.cell.2014.12.021.
    1. Huang L, et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat. Med. 2015;21:1364–1371. doi: 10.1038/nm.3973.
    1. Romero-Calvo I, et al. Human Organoids Share Structural and Genetic Features with Primary Pancreatic Adenocarcinoma Tumors. Mol. Cancer Res. 2019;17:70–83. doi: 10.1158/1541-7786.MCR-18-0531.
    1. Seino T, et al. Human Pancreatic Tumor Organoids Reveal Loss of Stem Cell Niche Factor Dependence during Disease Progression. cell stem cell. 2018;22:454–467.e456. doi: 10.1016/j.stem.2017.12.009.
    1. D’Agosto S, Andreani S, Scarpa A. Preclinical Modelling of PDA: Is Organoid the New Black? Int. J. Mol. Sci. 2019;20:2766. doi: 10.3390/ijms20112766.
    1. Driehuis E, et al. Pancreatic cancer organoids recapitulate disease and allow personalized drug screening. Proc. Natl. Acad. Sci. USA. 2019;116:26580–26590. doi: 10.1073/pnas.1911273116.
    1. Kawasaki K, et al. An Organoid Biobank of Neuroendocrine Neoplasms Enables Genotype-Phenotype Mapping. Cell. 2020;183:1420–1435.e1421. doi: 10.1016/j.cell.2020.10.023.
    1. Pavel M, et al. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2020;31:844–860. doi: 10.1016/j.annonc.2020.03.304.
    1. Stephens PJ, et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell. 2011;144:27–40. doi: 10.1016/j.cell.2010.11.055.
    1. Hoadley KA, et al. Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer. Cell. 2018;173:291–304 e296. doi: 10.1016/j.cell.2018.03.022.
    1. Tate JG, et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2018;47:D941–D947. doi: 10.1093/nar/gky1015.
    1. Chakravarty D, et al. OncoKB: a precision oncology knowledge base. JCO Precis. Oncol. 2017;1:1–16.
    1. Hijioka S, et al. Rb Loss and KRAS Mutation Are Predictors of the Response to Platinum-Based Chemotherapy in Pancreatic Neuroendocrine Neoplasm with Grade 3: A Japanese Multicenter Pancreatic NEN-G3 Study. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2017;23:4625–4632. doi: 10.1158/1078-0432.CCR-16-3135.
    1. Mafficini A, Scarpa A. Genetics and Epigenetics of Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr. Rev. 2019;40:506–536. doi: 10.1210/er.2018-00160.
    1. Jiao Y, et al. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Sci. (N. Y., N. Y.) 2011;331:1199–1203. doi: 10.1126/science.1200609.
    1. Marinoni I, et al. Loss of DAXX and ATRX are associated with chromosome instability and reduced survival of patients with pancreatic neuroendocrine tumors. Gastroenterology. 2014;146:453–460.e455. doi: 10.1053/j.gastro.2013.10.020.
    1. Consortium, I.T.P.-C.A.O.W.G. Pan-cancer analysis of whole genomes. Nature. 2020;578:82–93. doi: 10.1038/s41586-020-1969-6.
    1. Rheinbay E, et al. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes. Nature. 2020;578:102–111. doi: 10.1038/s41586-020-1965-x.
    1. Karasinska JM, et al. Altered Gene Expression along the Glycolysis-Cholesterol Synthesis Axis Is Associated with Outcome in Pancreatic Cancer. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2020;26:135–146. doi: 10.1158/1078-0432.CCR-19-1543.
    1. Tseng IC, Yeh MM, Yang CY, Jeng YM. NKX6-1 Is a Novel Immunohistochemical Marker for Pancreatic and Duodenal Neuroendocrine Tumors. Am. J. surgical Pathol. 2015;39:850–857. doi: 10.1097/PAS.0000000000000435.
    1. Wang, C., et al. Metformin inhibits pancreatic cancer metastasis caused by SMAD4 deficiency and consequent HNF4G upregulation. Protein & cell (2020) Online ahead of print.
    1. Kuo TL, Cheng KH, Chen LT, Hung WC. Deciphering The Potential Role of Hox Genes in Pancreatic Cancer. Cancers. 2019;11:734. doi: 10.3390/cancers11050734.
    1. Zhou Q, et al. A multipotent progenitor domain guides pancreatic organogenesis. Developmental cell. 2007;13:103–114. doi: 10.1016/j.devcel.2007.06.001.
    1. Ramsay RG, Gonda TJ. MYB function in normal and cancer cells. Nat. Rev. Cancer. 2008;8:523–534. doi: 10.1038/nrc2439.
    1. Tomic G, et al. Phospho-regulation of ATOH1 Is Required for Plasticity of Secretory Progenitors and Tissue Regeneration. cell stem cell. 2018;23:436–443.e437. doi: 10.1016/j.stem.2018.07.002.
    1. Zhang D, et al. Involvement of a Transcription factor, Nfe2, in Breast Cancer Metastasis to Bone. Cancers. 2020;12:3003. doi: 10.3390/cancers12103003.
    1. Okita Y, et al. The transcription factor MAFK induces EMT and malignant progression of triple-negative breast cancer cells through its target GPNMB. Sci. Signal. 2017;10:eaak9397. doi: 10.1126/scisignal.aak9397.
    1. Bleu M, et al. PAX8 and MECOM are interaction partners driving ovarian cancer. Nat. Commun. 2021;12:2442. doi: 10.1038/s41467-021-22708-w.
    1. Trevino AE, et al. Chromatin accessibility dynamics in a model of human forebrain development. Sci. (N. Y., N. Y.) 2020;367:eaay1645. doi: 10.1126/science.aay1645.
    1. Xu Y, et al. Multi-omics analysis at epigenomics and transcriptomics levels reveals prognostic subtypes of lung squamous cell carcinoma. Biomedicine Pharmacother. = Biomedecine pharmacotherapie. 2020;125:109859. doi: 10.1016/j.biopha.2020.109859.
    1. Chen X, et al. S100 calcium-binding protein A6 promotes epithelial-mesenchymal transition through β-catenin in pancreatic cancer cell line. PloS one. 2015;10:e0121319. doi: 10.1371/journal.pone.0121319.
    1. Ohuchida K, et al. The role of S100A6 in pancreatic cancer development and its clinical implication as a diagnostic marker and therapeutic target. Clin. Cancer Res. 2005;11:7785–7793. doi: 10.1158/1078-0432.CCR-05-0714.
    1. Crescenzo R, et al. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer cell. 2015;27:516–532. doi: 10.1016/j.ccell.2015.03.006.
    1. Tsoi H, Man EPS, Chau KM, Khoo US. Targeting the IL-6/STAT3 Signalling Cascade to Reverse Tamoxifen Resistance in Estrogen Receptor Positive Breast Cancer. Cancers. 2021;13:1511. doi: 10.3390/cancers13071511.
    1. Radhakrishnan R, et al. Histone deacetylase 10 regulates DNA mismatch repair and may involve the deacetylation of MutS homolog 2. J. Biol. Chem. 2015;290:22795–22804. doi: 10.1074/jbc.M114.612945.
    1. Das CK, et al. BAG3 Overexpression and Cytoprotective Autophagy Mediate Apoptosis Resistance in Chemoresistant Breast Cancer Cells. Neoplasia (N. Y., N. Y.) 2018;20:263–279. doi: 10.1016/j.neo.2018.01.001.
    1. Habata S, et al. BAG3-mediated Mcl-1 stabilization contributes to drug resistance via interaction with USP9X in ovarian cancer. Int. J. Oncol. 2016;49:402–410. doi: 10.3892/ijo.2016.3494.
    1. Yanagie H, et al. Improvement of sensitivity to platinum compound with siRNA knockdown of upregulated genes in platinum complex-resistant ovarian cancer cells in vitro. Biomedicine Pharmacother. = Biomedecine pharmacotherapie. 2009;63:553–560. doi: 10.1016/j.biopha.2008.04.006.
    1. Carter, B. & Zhao, K. The epigenetic basis of cellular heterogeneity. Nature reviews. Genetics (2020).
    1. Zhang S, et al. Allele-specific open chromatin in human iPSC neurons elucidates functional disease variants. Sci. (N. Y., N. Y.) 2020;369:561–565. doi: 10.1126/science.aay3983.
    1. Arruabarrena-Aristorena A, et al. FOXA1 Mutations Reveal Distinct Chromatin Profiles and Influence Therapeutic Response in Breast Cancer. Cancer cell. 2020;38:534–550 e539. doi: 10.1016/j.ccell.2020.08.003.
    1. Zhang L, Lu Q, Chang C. Epigenetics in Health and Disease. Adv. Exp. Med. Biol. 2020;1253:3–55. doi: 10.1007/978-981-15-3449-2_1.
    1. Souche R, et al. Outcome after pancreatectomy for neuroendocrine neoplams according to the WHO 2017 grading system: A retrospective multicentric analysis of 138 consecutive patients. Clin. Res. Hepatol. Gastroenterol. 2019;44:286–294. doi: 10.1016/j.clinre.2019.08.010.
    1. Pea A, et al. Genetic Analysis of Small Well-differentiated Pancreatic Neuroendocrine Tumors Identifies Subgroups With Differing Risks of Liver Metastases. Ann. Surg. 2020;271:566–573. doi: 10.1097/SLA.0000000000003022.
    1. Luley KB, et al. A Comprehensive Molecular Characterization of the Pancreatic Neuroendocrine Tumor Cell Lines BON-1 and QGP-1. Cancers. 2020;12:691. doi: 10.3390/cancers12030691.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinforma. (Oxf., Engl.) 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXivpreprint arXiv:1303.3997 (2013).
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinforma. (Oxf., Engl.) 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic acids Res. 2010;38:e164. doi: 10.1093/nar/gkq603.
    1. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: Genome-Wide Copy Number Detection and Visualization from Targeted DNA Sequencing. PLoS computational Biol. 2016;12:e1004873. doi: 10.1371/journal.pcbi.1004873.
    1. Mermel CH, et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 2011;12:R41. doi: 10.1186/gb-2011-12-4-r41.
    1. Robinson JT, et al. Integrative genomics viewer. Nat. Biotechnol. 2011;29:24–26. doi: 10.1038/nbt.1754.
    1. Zhang H, Meltzer P, Davis S. RCircos: an R package for Circos 2D track plots. BMC Bioinforma. 2013;14:244. doi: 10.1186/1471-2105-14-244.
    1. Kim D, et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36. doi: 10.1186/gb-2013-14-4-r36.
    1. Trapnell C, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 2010;28:511–515. doi: 10.1038/nbt.1621.
    1. Gaujoux R, Seoighe C. A flexible R package for nonnegative matrix factorization. BMC Bioinforma. 2010;11:367. doi: 10.1186/1471-2105-11-367.
    1. Kolde, R. Pheatmap: pretty heatmaps. R package version1 (2012).
    1. Subramanian A, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Kuleshov MV, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic acids Res. 2016;44:W90–W97. doi: 10.1093/nar/gkw377.
    1. Cerami E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404. doi: 10.1158/-12-0095.
    1. Jacobsen, A. cgdsr: R-based API for accessing the MSKCC cancer genomics data server (CGDS). R package version1 (2015).
    1. Langmead B, Trapnell C, Pop M, Salzberg SL. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 2009;10:R25. doi: 10.1186/gb-2009-10-3-r25.
    1. Li D, Hsu S, Purushotham D, Sears RL, Wang T. WashU Epigenome Browser update 2019. Nucleic acids Res. 2019;47:W158–w165. doi: 10.1093/nar/gkz348.
    1. Bolstad, B. M. preprocessCore: A collection of pre-processing functions. R package version1 (2013).
    1. Cavalcante RG, Sartor MA. annotatr: genomic regions in context. Bioinforma. (Oxf., Engl.) 2017;33:2381–2383. doi: 10.1093/bioinformatics/btx183.
    1. Aibar S, et al. SCENIC: single-cell regulatory network inference and clustering. Nat. methods. 2017;14:1083–1086. doi: 10.1038/nmeth.4463.
    1. Heinz S, et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. cell. 2010;38:576–589. doi: 10.1016/j.molcel.2010.05.004.
    1. Shi, X., et al. Integrated profiling of human pancreatic cancer organoids reveals chromatin accessibility features associated with drug sensitivity. Github10.5281/zenodo.5784804 (2021).

Source: PubMed

3
订阅