Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Hybrid sequencing-based personal full-length transcriptomic analysis implicates proteostatic stress in metastatic ovarian cancer

Abstract

Comprehensive molecular characterization of myriad somatic alterations and aberrant gene expressions at personal level is key to precision cancer therapy, yet limited by current short-read sequencing technology, individualized catalog of complete genomic and transcriptomic features is thus far elusive. Here, we integrated second- and third-generation sequencing platforms to generate a multidimensional dataset on a patient affected by metastatic epithelial ovarian cancer. Whole-genome and hybrid transcriptome dissection captured global genetic and transcriptional variants at previously unparalleled resolution. Particularly, single-molecule mRNA sequencing identified a vast array of unannotated transcripts, novel long noncoding RNAs and gene chimeras, permitting accurate determination of transcription start, splice, polyadenylation and fusion sites. Phylogenetic and enrichment inference of isoform-level measurements implicated early functional divergence and cytosolic proteostatic stress in shaping ovarian tumorigenesis. A complementary imaging-based high-throughput drug screen was performed and subsequently validated, which consistently pinpointed proteasome inhibitors as an effective therapeutic regime by inducing protein aggregates in ovarian cancer cells. Therefore, our study suggests that clinical application of the emerging long-read full-length analysis for improving molecular diagnostics is feasible and informative. An in-depth understanding of the tumor transcriptome complexity allowed by leveraging the hybrid sequencing approach lays the basis to reveal novel and valid therapeutic vulnerabilities in advanced ovarian malignancies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

References

  1. Mills K, Fuh K. Recent advances in understanding, diagnosing, and treating ovarian cancer. F1000Res. 2017;6:84.

    PubMed  PubMed Central  Google Scholar 

  2. Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J, Karlan BY. Ovarian cancer. Nat Rev Dis Prim. 2016;2:16061.

    PubMed  Google Scholar 

  3. Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–15.

    Google Scholar 

  4. Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521:489–94.

    CAS  PubMed  Google Scholar 

  5. Wang YK, Bashashati A, Anglesio MS, Cochrane DR, Grewal DS, Ha G, et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat Genet. 2017;49:856–65.

    CAS  PubMed  Google Scholar 

  6. Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N, Sander C. Emerging landscape of oncogenic signatures across human cancers. Nat Genet. 2013;45:1127–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhang Z, Ma P, Jing Y, Yan Y, Cai MC, Zhang M, et al. BET bromodomain inhibition as a therapeutic strategy in ovarian cancer by downregulating FoxM1. Theranostics. 2016;6:219–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhang Z, Peng H, Wang X, Yin X, Ma P, Jing Y, et al. Preclinical efficacy and molecular mechanism of targeting CDK7-dependent transcriptional addiction in ovarian cancer. Mol Cancer Ther. 2017;16:1739–50.

    CAS  PubMed  Google Scholar 

  9. Kumar-Sinha C, Chinnaiyan AM. Precision oncology in the age of integrative genomics. Nat Biotechnol. 2018;36:46–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Alkan C, Sajjadian S, Eichler EE. Limitations of next-generation genome sequence assembly. Nat Methods. 2011;8:61–65.

    CAS  PubMed  Google Scholar 

  11. Niu B, Fu L, Sun S, Li W. Artificial and natural duplicates in pyrosequencing reads of metagenomic data. BMC Bioinforma. 2010;11:187.

    Google Scholar 

  12. Steijger T, Abril JF, Engstrom PG, Kokocinski F, Consortium R, Hubbard TJ, et al. Assessment of transcript reconstruction methods for RNA-seq. Nat Methods. 2013;10:1177–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Cieslik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet. 2018;19:93–109.

    CAS  PubMed  Google Scholar 

  14. Ozsolak F, Milos PM. RNA sequencing: advances, challenges and opportunities. Nat Rev Genet. 2011;12:87–98.

    CAS  PubMed  Google Scholar 

  15. Eid J, Fehr A, Gray J, Luong K, Lyle J, Otto G, et al. Real-time DNA sequencing from single polymerase molecules. Science. 2009;323:133–8.

    Article  CAS  PubMed  Google Scholar 

  16. Rhoads A, Au KF. PacBio sequencing and its applications. Genom Proteom Bioinforma. 2015;13:278–89.

    Google Scholar 

  17. Sharon D, Tilgner H, Grubert F, Snyder M. A single-molecule long-read survey of the human transcriptome. Nat Biotechnol. 2013;31:1009–14.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Roberts RJ, Carneiro MO, Schatz MC. The advantages of SMRT sequencing. Genome Biol. 2013;14:405.

    PubMed  PubMed Central  Google Scholar 

  19. Travers KJ, Chin CS, Rank DR, Eid JS, Turner SW. A flexible and efficient template format for circular consensus sequencing and SNP detection. Nucleic Acids Res. 2010;38:e159.

    PubMed  PubMed Central  Google Scholar 

  20. Koren S, Schatz MC, Walenz BP, Martin J, Howard JT, Ganapathy G, et al. Hybrid error correction and de novo assembly of single-molecule sequencing reads. Nat Biotechnol. 2012;30:693–700.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Abdel-Ghany SE, Hamilton M, Jacobi JL, Ngam P, Devitt N, Schilkey F, et al. A survey of the sorghum transcriptome using single-molecule long reads. Nat Commun. 2016;7:11706.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Au KF, Sebastiano V, Afshar PT, Durruthy JD, Lee L, Williams BA, et al. Characterization of the human ESC transcriptome by hybrid sequencing. Proc Natl Acad Sci USA. 2013;110:E4821–4830.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Kuang Z, Boeke JD, Canzar S. The dynamic landscape of fission yeast meiosis alternative-splice isoforms. Genome Res. 2017;27:145–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Li S, Yamada M, Han X, Ohler U, Benfey PN. High-resolution expression map of the Arabidopsis root reveals alternative splicing and lincRNA regulation. Dev Cell. 2016;39:508–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Tian B, Manley JL. Alternative polyadenylation of mRNA precursors. Nat Rev Mol Cell Biol. 2017;18:18–30.

    CAS  PubMed  Google Scholar 

  26. Wang B, Tseng E, Regulski M, Clark TA, Hon T, Jiao Y, et al. Unveiling the complexity of the maize transcriptome by single-molecule long-read sequencing. Nat Commun. 2016;7:11708.

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Weirather JL, Afshar PT, Clark TA, Tseng E, Powers LS, Underwood JG, et al. Characterization of fusion genes and the significantly expressed fusion isoforms in breast cancer by hybrid sequencing. Nucleic Acids Res. 2015;43:e116.

    PubMed  PubMed Central  Google Scholar 

  28. Lengyel E. Ovarian cancer development and metastasis. Am J Pathol. 2010;177:1053–64.

    PubMed  PubMed Central  Google Scholar 

  29. Consortium F, the RP, CLST, Forrest AR, Kawaji H, Rehli M, et al. A promoter-level mammalian expression atlas. Nature. 2014;507:462–70.

  30. de Klerk E, t Hoen PA. Alternative mRNA transcription, processing, and translation: insights from RNA sequencing. Trends Genet. 2015;31:128–39.

    PubMed  Google Scholar 

  31. Kornblihtt AR, Schor IE, Allo M, Dujardin G, Petrillo E, Munoz MJ. Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat Rev Mol Cell Biol. 2013;14:153–65.

    CAS  PubMed  Google Scholar 

  32. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, et al. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456:470–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cui T, Zhang L, Huang Y, Yi Y, Tan P, Zhao Y, et al. MNDRv2.0: an updated resource of ncRNA-disease associations in mammals. Nucleic Acids Res. 2018;46:D371–D374.

    CAS  PubMed  Google Scholar 

  34. Yi Y, Zhao Y, Li C, Zhang L, Huang H, Li Y, et al. RAID v2.0: an updated resource of RNA-associated interactions across organisms. Nucleic Acids Res. 2017;45:D115–D118.

    CAS  PubMed  Google Scholar 

  35. Zhang T, Tan P, Wang L, Jin N, Li Y, Zhang L, et al. RNALocate: a resource for RNA subcellular localizations. Nucleic Acids Res. 2017;45:D135–D138.

    CAS  PubMed  Google Scholar 

  36. Mertens F, Johansson B, Fioretos T, Mitelman F. The emerging complexity of gene fusions in cancer. Nat Rev Cancer. 2015;15:371–81.

    CAS  PubMed  Google Scholar 

  37. Schram AM, Chang MT, Jonsson P, Drilon A. Fusions in solid tumours: diagnostic strategies, targeted therapy, and acquired resistance. Nat Rev Clin Oncol. 2017;14:735–48.

    CAS  PubMed  Google Scholar 

  38. Engstrom PG, Steijger T, Sipos B, Grant GR, Kahles A, Ratsch G, et al. Systematic evaluation of spliced alignment programs for RNA-seq data. Nat Methods. 2013;10:1185–91.

    PubMed  PubMed Central  Google Scholar 

  39. Bashashati A, Ha G, Tone A, Ding J, Prentice LM, Roth A, et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J Pathol. 2013;231:21–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Choi YJ, Rhee JK, Hur SY, Kim MS, Lee SH, Chung YJ, et al. Intraindividual genomic heterogeneity of high-grade serous carcinoma of the ovary and clinical utility of ascitic cancer cells for mutation profiling. J Pathol. 2017;241:57–66.

    CAS  PubMed  Google Scholar 

  41. Hoogstraat M, de Pagter MS, Cirkel GA, van Roosmalen MJ, Harkins TT, Duran K, et al. Genomic and transcriptomic plasticity in treatment-naive ovarian cancer. Genome Res. 2014;24:200–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. McPherson A, Roth A, Laks E, Masud T, Bashashati A, Zhang AW, et al. Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer. Nat Genet. 2016;48:758–67.

    CAS  PubMed  Google Scholar 

  43. Yin X, Jing Y, Cai MC, Ma P, Zhang Y, Xu C, et al. Clonality, heterogeneity, and evolution of synchronous bilateral ovarian cancer. Cancer Res. 2017;77:6551–61.

    CAS  PubMed  Google Scholar 

  44. Vilasi S, Bulone D, Caruso Bavisotto C, Campanella C, Marino Gammazza A, San Biagio PL, et al. Chaperonin of group I: oligomeric spectrum and biochemical and biological implications. Front Mol Biosci. 2017;4:99.

    PubMed  Google Scholar 

  45. Chatterjee S, Burns TF. Targeting Heat Shock Proteins in Cancer: A Promising Therapeutic Approach. Int J Mol Sci. 2017;18:1978

    PubMed Central  Google Scholar 

  46. Fulda S, Rajalingam K, Dikic I. Ubiquitylation in immune disorders and cancer: from molecular mechanisms to therapeutic implications. EMBO Mol Med. 2012;4:545–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Nandi D, Tahiliani P, Kumar A, Chandu D. The ubiquitin-proteasome system. J Biosci. 2006;31:137–55.

    CAS  PubMed  Google Scholar 

  48. Faoro R, Bassu M, Mejia YX, Stephan T, Dudani N, Boeker C, et al. Aberration-corrected cryoimmersion light microscopy. Proc Natl Acad Sci USA. 2018;115:1204–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Rubio-Perez C, Tamborero D, Schroeder MP, Antolin AA, Deu-Pons J, Perez-Llamas C, et al. In silico prescription of anticancer drugs to cohorts of 28 tumor types reveals targeting opportunities. Cancer Cell. 2015;27:382–96.

    CAS  PubMed  Google Scholar 

  50. Manasanch EE, Orlowski RZ. Proteasome inhibitors in cancer therapy. Nat Rev Clin Oncol. 2017;14:417–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Tang Z, Dai S, He Y, Doty RA, Shultz LD, Sampson SB, et al. MEK guards proteome stability and inhibits tumor-suppressive amyloidogenesis via HSF1. Cell. 2015;160:729–44.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Tilgner H, Grubert F, Sharon D, Snyder MP. Defining a personal, allele-specific, and single-molecule long-read transcriptome. Proc Natl Acad Sci USA. 2014;111:9869–74.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Turajlic S, Swanton C. Metastasis as an evolutionary process. Science. 2016;352:169–75.

    CAS  PubMed  Google Scholar 

  54. Bazzaro M, Lee MK, Zoso A, Stirling WL, Santillan A, Shih Ie M, et al. Ubiquitin-proteasome system stress sensitizes ovarian cancer to proteasome inhibitor-induced apoptosis. Cancer Res. 2006;66:3754–63.

    CAS  PubMed  Google Scholar 

  55. Kumar SK. New treatment options for the management of multiple myeloma. J Natl Compr Canc Netw. 2017;15:709–12.

    PubMed  Google Scholar 

  56. Jandial DA, Brady WE, Howell SB, Lankes HA, Schilder RJ, Beumer JH, et al. A phase I pharmacokinetic study of intraperitoneal bortezomib and carboplatin in patients with persistent or recurrent ovarian cancer: an NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol. 2017;145:236–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Ramirez PT, Landen CN Jr., Coleman RL, Milam MR, Levenback C, Johnston TA, et al. Phase I trial of the proteasome inhibitor bortezomib in combination with carboplatin in patients with platinum- and taxane-resistant ovarian cancer. Gynecol Oncol. 2008;108:68–71.

    CAS  PubMed  Google Scholar 

  58. Li H, Durbin R. Fast and accurate short read alignment with Burrows−Wheeler transform. Bioinformatics. 2009;25:1754–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–9.

    PubMed  PubMed Central  Google Scholar 

  60. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–303.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Cibulskis K, Lawrence MS, Carter SL, Sivachenko A, Jaffe D, Sougnez C, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang K, Li M, Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164.

    PubMed  PubMed Central  Google Scholar 

  63. Ng PC, Henikoff S. SIFT: predicting amino acid changes that affect protein function. Nucleic Acids Res. 2003;31:3812–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Adzhubei I, Jordan DM, Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen-2. Curr Protoc Human Genet. 2013;Chapter 7:Unit7 20.

    Google Scholar 

  65. Boeva V, Popova T, Bleakley K, Chiche P, Cappo J, Schleiermacher G, et al. Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics. 2012;28:423–5.

    CAS  PubMed  Google Scholar 

  66. Chen K, Wallis JW, McLellan MD, Larson DE, Kalicki JM, Pohl CS, et al. BreakDancer: an algorithm for high-resolution mapping of genomic structural variation. Nat Methods. 2009;6:677–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36.

    PubMed  PubMed Central  Google Scholar 

  68. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014;30:923–30.

    CAS  PubMed  Google Scholar 

  70. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.

    CAS  PubMed  Google Scholar 

  71. Jia W, Qiu K, He M, Song P, Zhou Q, Zhou F, et al. SOAPfuse: an algorithm for identifying fusion transcripts from paired-end RNA-Seq data. Genome Biol. 2013;14:R12.

    PubMed  PubMed Central  Google Scholar 

  72. Trincado JL, Entizne JC, Hysenaj G, Singh B, Skalic M, Elliott DJ, et al. SUPPA2: fast, accurate, and uncertainty-aware differential splicing analysis across multiple conditions. Genome Biol. 2018;19:40.

    PubMed  PubMed Central  Google Scholar 

  73. Xia Z, Donehower LA, Cooper TA, Neilson JR, Wheeler DA, Wagner EJ, et al. Dynamic analyses of alternative polyadenylation from RNA-seq reveal a 3’-UTR landscape across seven tumour types. Nat Commun. 2014;5:5274.

    CAS  PubMed  Google Scholar 

  74. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Hackl T, Hedrich R, Schultz J, Forster F. proovread: large-scale high-accuracy PacBio correction through iterative short read consensus. Bioinformatics. 2014;30:3004–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Wu TD, Reeder J, Lawrence M, Becker G, Brauer MJ. GMAP and GSNAP for genomic sequence alignment: enhancements to speed, accuracy, and functionality. Methods Mol Biol. 2016;1418:283–334.

    PubMed  Google Scholar 

  77. Sun L, Luo H, Bu D, Zhao G, Yu K, Zhang C, et al. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 2013;41:e166.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Kong L, Zhang Y, Ye ZQ, Liu XQ, Zhao SQ, Wei L, et al. CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res. 2007;35:W345–349.

    PubMed  PubMed Central  Google Scholar 

  79. Finn RD, Coggill P, Eberhardt RY, Eddy SR, Mistry J, Mitchell AL, et al. The Pfam protein families database: towards a more sustainable future. Nucleic Acids Res. 2016;44:D279–285.

    CAS  PubMed  Google Scholar 

  80. Li A, Zhang J, Zhou Z. PLEK: a tool for predicting long non-coding RNAs and messenger RNAs based on an improved k-mer scheme. BMC Bioinforma. 2014;15:311.

    Google Scholar 

  81. Rosenthal R, McGranahan N, Herrero J, Taylor BS, Swanton C. DeconstructSigs: delineating mutational processes in single tumors distinguishes DNA repair deficiencies and patterns of carcinoma evolution. Genome Biol. 2016;17:31.

    PubMed  PubMed Central  Google Scholar 

  82. Miller CA, White BS, Dees ND, Griffith M, Welch JS, Griffith OL, et al. SciClone: inferring clonal architecture and tracking the spatial and temporal patterns of tumor evolution. PLoS Comput Biol. 2014;10:e1003665.

    PubMed  PubMed Central  Google Scholar 

  83. Roth A, Khattra J, Yap D, Wan A, Laks E, Biele J, et al. PyClone: statistical inference of clonal population structure in cancer. Nat Methods. 2014;11:396–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack T, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 2012;30:413–21.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Natural Science Foundation of China (81472537 and 81672714 to GZ; 81502597 to YJ; 81472426 to WD), the Grants from the State Key Laboratory of Oncogenes and Related Genes (SB17-06 to M-CC), the grants from Shanghai Jiao Tong University School of Medicine (DLY201505 to WD; YG2016MS51 to XY), Shanghai Municipal Education Commission-Gaofeng Clinical Medicine Grant Support (20161313 to GZ), the Shanghai Institutions of Higher Learning (Eastern Scholar to GZ), Shanghai Rising-Star Program (16QA1403600 to GZ), Shanghai Municipal Commission of Health and Family Planning (2013ZYJB0202 and 15GWZK0701 to WD; 20174Y0189 to YJ; 20174Y0043 to M-CC), the grant from Shanghai Key Laboratory of Gynecologic Oncology (FKZL-2017-01 to YJ), and the grant from Science and Technology Commission of Shanghai Municipality (16140904401 to XY).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Xiaojie Wang or Guanglei Zhuang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jing, Y., Zhang, Y., Zhu, H. et al. Hybrid sequencing-based personal full-length transcriptomic analysis implicates proteostatic stress in metastatic ovarian cancer. Oncogene 38, 3047–3060 (2019). https://doi.org/10.1038/s41388-018-0644-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-018-0644-y

This article is cited by

Search

Quick links