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.

  • Review Article
  • Published:

Discovering and validating cancer genetic dependencies: approaches and pitfalls

Abstract

Cancer ‘genetic dependencies’ — genes whose products are essential for cancer cell fitness — are promising targets for therapeutic development. However, recent evidence has cast doubt on the validity of several putative dependencies that are currently being targeted in cancer clinical trials, underscoring the challenges inherent in correctly identifying cancer-essential genes. Here we review several common techniques and platforms for discovering and characterizing cancer dependencies. We discuss the strengths and drawbacks of different gene-perturbation approaches, and we highlight the use of poorly validated genetic and pharmacological agents as a common cause of target misidentification. A careful consideration of the limitations of current technologies and cancer models will improve our ability to correctly uncover cancer genetic dependencies and will facilitate the development of improved therapeutic agents.

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

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

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

Fig. 1: Potential pitfalls when using RNAi or CRISPR–Cas9 technology.
Fig. 2: Comparison of different preclinical models for identifying cancer dependencies.

Similar content being viewed by others

References

  1. Luo, J., Solimini, N. L. & Elledge, S. J. Principles of cancer therapy: oncogene and non-oncogene addiction. Cell 136, 823–837 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Ben-Neriah, Y., Daley, G. Q., Mes-Masson, A. M., Witte, O. N. & Baltimore, D. The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene. Science 233, 212–214 (1986).

    CAS  PubMed  Google Scholar 

  3. Nowell, P. C. & Hungerford, D. A. Chromosome studies on normal and leukemic human leukocytes. J. Natl Cancer Inst. 25, 85–109 (1960).

    CAS  PubMed  Google Scholar 

  4. Lugo, T. G., Pendergast, A. M., Muller, A. J. & Witte, O. N. Tyrosine kinase activity and transformation potency of bcr-abl oncogene products. Science 247, 1079–1082 (1990).

    CAS  PubMed  Google Scholar 

  5. Huettner, C. S., Zhang, P., Van Etten, R. A. & Tenen, D. G. Reversibility of acute B-cell leukaemia induced by BCR-ABL1. Nat. Genet. 24, 57–60 (2000).

    CAS  PubMed  Google Scholar 

  6. Druker, B. J. & Lydon, N. B. Lessons learned from the development of an Abl tyrosine kinase inhibitor for chronic myelogenous leukemia. J. Clin. Invest. 105, 3–7 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Druker, B. J. et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N. Engl. J. Med. 344, 1031–1037 (2001).

    CAS  PubMed  Google Scholar 

  8. Gorre, M. E. et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 293, 876–880 (2001). This landmark article reports that patients with CML who relapsed following treatment with imatinib developed point mutations in BCRABL that restored kinase activity, thereby proving that the efficacy of imatinib resulted from an ‘on-target’ effect.

    CAS  PubMed  Google Scholar 

  9. Roumiantsev, S. et al. Clinical resistance to the kinase inhibitor STI-571 in chronic myeloid leukemia by mutation of Tyr-253 in the Abl kinase domain P-loop. Proc. Natl Acad. Sci. USA 99, 10700–10705 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Azam, M., Latek, R. R. & Daley, G. Q. Mechanisms of autoinhibition and STI-571/Imatinib resistance revealed by mutagenesis of BCR-ABL. Cell 112, 831–843 (2003).

    CAS  PubMed  Google Scholar 

  11. Druker, B. J. et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N. Engl. J. Med. 355, 2408–2417 (2006).

    CAS  PubMed  Google Scholar 

  12. Kantarjian, H. et al. Improved survival in chronic myeloid leukemia since the introduction of imatinib therapy: a single-institution historical experience. Blood 119, 1981–1987 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Luke, J. J. & Hodi, F. S. Vemurafenib and BRAF inhibition: a new class of treatment for metastatic melanoma. Clin. Cancer Res. 18, 9–14 (2012).

    CAS  PubMed  Google Scholar 

  14. Seshacharyulu, P. et al. Targeting the EGFR signaling pathway in cancer therapy. Expert. Opin. Ther. Targets 16, 15–31 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wong, C. H., Siah, K. W. & Lo, A. W. Estimation of clinical trial success rates and related parameters. Biostatistics 20, 273–286 (2019).

    PubMed  Google Scholar 

  16. Hay, M., Thomas, D. W., Craighead, J. L., Economides, C. & Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 32, 40–51 (2014).

    CAS  PubMed  Google Scholar 

  17. Waring, M. J. et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat. Rev. Drug. Discov. 14, 475–486 (2015).

    CAS  PubMed  Google Scholar 

  18. Settleman, J., Sawyers, C. L. & Hunter, T. Challenges in validating candidate therapeutic targets in cancer. eLife 7, e32402 (2018).

    PubMed  PubMed Central  Google Scholar 

  19. Giuliano, C. J., Lin, A., Smith, J. C., Palladino, A. C. & Sheltzer, J. M. MELK expression correlates with tumor mitotic activity but is not required for cancer growth. eLife 7, e32838 (2018).

    PubMed  PubMed Central  Google Scholar 

  20. Huang, H.-T. et al. MELK is not necessary for the proliferation of basal-like breast cancer cells. eLife 6, e26693 (2017).

    PubMed  PubMed Central  Google Scholar 

  21. Ji, W. et al. OTSSP167 abrogates mitotic checkpoint through inhibiting multiple mitotic kinases. PLoS One 11, e0153518 (2016).

    PubMed  PubMed Central  Google Scholar 

  22. Lin, A., Giuliano, C. J., Sayles, N. M. & Sheltzer, J. M. CRISPR/Cas9 mutagenesis invalidates a putative cancer dependency targeted in on-going clinical trials. eLife 6, e24179 (2017). To our knowledge, this study represents the first demonstration that a protein targeted in clinical trials is fully dispensable for cancer cell fitness, and that a small molecule studied in clinical trials kills specifically through an off-target mechanism.

    PubMed  PubMed Central  Google Scholar 

  23. Lin, A. et al. Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials. Sci. Transl Med. 11, eaaw8412 (2019). This study uses CRISPR-mediated gene editing to establish that many cancer drugs studied in clinical trials have been designed to inhibit non-essential targets and kill cancer cells only through off-target effects.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Tsherniak, A. et al. Defining a cancer dependency map. Cell 170, 564–576.e16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Bradley, C. A. Targeted therapies: understanding tumour drug addiction. Nat. Rev. Cancer 17, 634–635 (2017).

    CAS  PubMed  Google Scholar 

  26. Feng, F. Y. & Gilbert, L. A. Lethal clues to cancer-cell vulnerability. Nature 568, 463–464 (2019).

    CAS  PubMed  Google Scholar 

  27. Asghar, U., Witkiewicz, A. K., Turner, N. C. & Knudsen, E. S. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat. Rev. Drug. Discov. 14, 130–146 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Paplomata, E., Nahta, R. & O’Regan, R. M. Systemic therapy for early-stage HER2-positive breast cancers: time for a less-is-more approach? Cancer 121, 517–526 (2015).

    CAS  PubMed  Google Scholar 

  29. Girdler, F. et al. Validating Aurora B as an anti-cancer drug target. J. Cell. Sci. 119, 3664–3675 (2006).

    CAS  PubMed  Google Scholar 

  30. Settleman, J. Oncogene addiction. Curr. Biol. 22, R43–R44 (2012).

    CAS  PubMed  Google Scholar 

  31. Felsher, D. W. & Bishop, J. M. Reversible tumorigenesis by MYC in hematopoietic lineages. Mol. Cell 4, 199–207 (1999).

    CAS  PubMed  Google Scholar 

  32. Nagel, R., Semenova, E. A. & Berns, A. Drugging the addict: non-oncogene addiction as a target for cancer therapy. EMBO Rep. 17, 1516–1531 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Boutros, M. & Ahringer, J. The art and design of genetic screens: RNA interference. Nat. Rev. Genet. 9, 554–566 (2008).

    CAS  PubMed  Google Scholar 

  34. Agrawal, N. et al. RNA interference: biology, mechanism, and applications. Microbiol. Mol. Biol. Rev. 67, 657–685 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Paddison, P. J., Caudy, A. A., Bernstein, E., Hannon, G. J. & Conklin, D. S. Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev. 16, 948–958 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Paddison, P. J. et al. A resource for large-scale RNA-interference-based screens in mammals. Nature 428, 427–431 (2004).

    CAS  PubMed  Google Scholar 

  37. Silva, J. M. et al. Profiling essential genes in human mammary cells by multiplex RNAi screening. Science 319, 617–620 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Dominguez, A. A., Lim, W. A. & Qi, L. S. Beyond editing: repurposing CRISPR–Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol. 17, 5–15 (2016).

    CAS  PubMed  Google Scholar 

  39. Hsu, P. D., Lander, E. S. & Zhang, F. Development and applications of CRISPR-Cas9 for genome engineering. Cell 157, 1262–1278 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Adli, M. The CRISPR tool kit for genome editing and beyond. Nat. Commun. 9, 1–13 (2018).

    CAS  Google Scholar 

  41. Hart, T., Brown, K. R., Sircoulomb, F., Rottapel, R. & Moffat, J. Measuring error rates in genomic perturbation screens: gold standards for human functional genomics. Mol. Syst. Biol. 10, 733 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Hart, T. et al. High-resolution CRISPR screens reveal fitness genes and genotype-specific cancer liabilities. Cell 163, 1515–1526 (2015). This elegant study conducts multiple genome-wide CRISPR screens, introduces a Bayesian framework to analyse them and substantially expands the number of known essential genes in human cells.

    CAS  PubMed  Google Scholar 

  43. Morgens, D. W., Deans, R. M., Li, A. & Bassik, M. C. Systematic comparison of CRISPR/Cas9 and RNAi screens for essential genes. Nat. Biotechnol. 34, 634–636 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Vu, V. et al. Natural variation in gene expression modulates the severity of mutant phenotypes. Cell 162, 391–402 (2015).

    CAS  PubMed  Google Scholar 

  45. Chang, H. H. Y., Pannunzio, N. R., Adachi, N. & Lieber, M. R. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat. Rev. Mol. Cell Biol. 18, 495–506 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Ferreira da Silva, J. et al. Genome-scale CRISPR screens are efficient in non-homologous end-joining deficient cells. Sci. Rep. 9, 1–10 (2019).

    CAS  Google Scholar 

  47. Kass, E. M. & Jasin, M. Collaboration and competition between DNA double-strand break repair pathways. FEBS Lett. 584, 3703–3708 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Aguirre, A. J. et al. Genomic copy number dictates a gene-independent cell response to CRISPR-Cas9 targeting. Cancer Discov. 6, 914–929 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Munoz, D. M. et al. CRISPR screens provide a comprehensive assessment of cancer vulnerabilities but generate false-positive hits for highly amplified genomic regions. Cancer Discov. 6, 900–913 (2016).

    CAS  PubMed  Google Scholar 

  50. Meyers, R. M. et al. Computational correction of copy number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells. Nat. Genet. 49, 1779–1784 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response. Nat. Med. 24, 927 (2018).

    CAS  PubMed  Google Scholar 

  52. Enache, O. M. et al. Cas9 activates the p53 pathway and selects for p53-inactivating mutations. Nat. Genet. https://doi.org/10.1038/s41588-020-0623-4 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Brown, K. R., Mair, B., Soste, M. & Moffat, J. CRISPR screens are feasible in TP53 wild-type cells. Mol. Syst. Biol. 15, e8679 (2019).

    PubMed  PubMed Central  Google Scholar 

  54. Haapaniemi, E., Botla, S., Persson, J., Schmierer, B. & Taipale, J. Reply to “CRISPR screens are feasible in TP53 wild-type cells”. Mol Syst Biol 15, e9059 (2019).

    PubMed  PubMed Central  Google Scholar 

  55. Jackson, A. L. & Linsley, P. S. Recognizing and avoiding siRNA off-target effects for target identification and therapeutic application. Nat. Rev. Drug Discov. 9, 57–67 (2010).

    CAS  PubMed  Google Scholar 

  56. Krueger, U. et al. Insights into effective RNAi gained from large-scale siRNA validation screening. Oligonucleotides 17, 237–250 (2007).

    CAS  PubMed  Google Scholar 

  57. Sigoillot, F. D. et al. A bioinformatics method identifies prominent off-targeted transcripts in RNAi screens. Nat. Methods 9, 363–366 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Aagaard, L. & Rossi, J. J. RNAi therapeutics: principles, prospects and challenges. Adv. Drug Deliv. Rev. 59, 75–86 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kamola, P. J., Nakano, Y., Takahashi, T., Wilson, P. A. & Ui-Tei, K. The siRNA non-seed region and its target sequences are auxiliary determinants of off-target effects. PLoS Comput. Biol. 11, e1004656 (2015).

    PubMed  PubMed Central  Google Scholar 

  61. Putzbach, W. et al. Many si/shRNAs can kill cancer cells by targeting multiple survival genes through an off-target mechanism. eLife 6, e29702 (2017).

    PubMed  PubMed Central  Google Scholar 

  62. Putzbach, W. et al. CD95/Fas ligand mRNA is toxic to cells. eLife 7, e38621 (2018).

    PubMed  PubMed Central  Google Scholar 

  63. Grimm, D. et al. Fatality in mice due to oversaturation of cellular microRNA/short hairpin RNA pathways. Nature 441, 537–541 (2006).

    CAS  PubMed  Google Scholar 

  64. Khan, A. A. et al. Transfection of small RNAs globally perturbs gene regulation by endogenous microRNAs. Nat. Biotechnol. 27, 549–555 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Scholl, C. et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 137, 821–834 (2009).

    CAS  PubMed  Google Scholar 

  66. Luo, T. et al. STK33 kinase inhibitor BRD-8899 has no effect on KRAS-dependent cancer cell viability. Proc. Natl Acad. Sci. USA 109, 2860–2865 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Weïwer, M. et al. A potent and selective quinoxalinone-based stk33 inhibitor does not show synthetic lethality in KRAS-dependent cells. ACS Med. Chem. Lett. 3, 1034–1038 (2012).

    PubMed  PubMed Central  Google Scholar 

  68. Babij, C. et al. STK33 kinase activity is nonessential in KRAS-dependent cancer cells. Cancer Res. 71, 5818–5826 (2011).

    CAS  PubMed  Google Scholar 

  69. Fu, Y. et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 31, 822–826 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Pattanayak, V. et al. High-throughput profiling of off-target DNA cleavage reveals RNA-programmed Cas9 nuclease specificity. Nat. Biotechnol. 31, 839–843 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Boettcher, M. & McManus, M. T. Choosing the right tool for the job: RNAi, TALEN or CRISPR. Mol. Cell 58, 575–585 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Shalem, O., Sanjana, N. E. & Zhang, F. High-throughput functional genomics using CRISPR-Cas9. Nat. Rev. Genet. 16, 299–311 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Smith, I. et al. Evaluation of RNAi and CRISPR technologies by large-scale gene expression profiling in the connectivity map. PLoS Biol. 15, e2003213 (2017).

    PubMed  PubMed Central  Google Scholar 

  74. Kleinstiver, B. P. et al. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 529, 490–495 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Popp, M. W. & Maquat, L. E. Leveraging rules of nonsense-mediated mRNA decay for genome engineering and personalized medicine. Cell 165, 1319–1322 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Chen, D. et al. CRISPR/Cas9-mediated genome editing induces exon skipping by complete or stochastic altering splicing in the migratory locust. BMC Biotechnol. 18, 60 (2018).

    PubMed  PubMed Central  Google Scholar 

  77. Tang, J.-X. et al. CRISPR/Cas9-mediated genome editing induces gene knockdown by altering the pre-mRNA splicing in mice. BMC Biotechnol. https://doi.org/10.1186/s12896-018-0472-8 (2018).

  78. Tuladhar, R. et al. CRISPR/Cas9-based mutagenesis frequently provokes on-target mRNA misregulation. bioRxiv https://doi.org/10.1101/583138 (2019).

    Article  Google Scholar 

  79. Smits, A. H. et al. Biological plasticity rescues target activity in CRISPR knock outs. Nat. Methods 16, 1087–1093 (2019). Tuladhar et al. (2020) and Smits et al. (2019) reveal that some CRISPR manipulations fail to generate true gene knockouts, as target expression is rescued through alternative transcriptional regulation.

    CAS  PubMed  Google Scholar 

  80. El-Brolosy, M. A. et al. Genetic compensation triggered by mutant mRNA degradation. Nature 568, 193–197 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Ajina, R. et al. SpCas9-expression by tumor cells can cause T cell-dependent tumor rejection in immunocompetent mice. Oncoimmunology 8, e1577127 (2019).

    PubMed  PubMed Central  Google Scholar 

  82. Crudele, J. M. & Chamberlain, J. S. Cas9 immunity creates challenges for CRISPR gene editing therapies. Nat. Commun. 9, 3497 (2018).

    PubMed  PubMed Central  Google Scholar 

  83. Mullenders, J. & Bernards, R. Loss-of-function genetic screens as a tool to improve the diagnosis and treatment of cancer. Oncogene 28, 4409–4420 (2009).

    CAS  PubMed  Google Scholar 

  84. Gupta, S., Schoer, R. A., Egan, J. E., Hannon, G. J. & Mittal, V. Inducible, reversible, and stable RNA interference in mammalian cells. Proc. Natl Acad. Sci. USA 101, 1927–1932 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Giuliano, C. J., Lin, A., Girish, V. & Sheltzer, J. M. Generating single cell–derived knockout clones in mammalian cells with CRISPR/Cas9. Curr. Protoc. Mol. Biol. 128, e100 (2019).

    PubMed  PubMed Central  Google Scholar 

  86. Zou, X. et al. Validating the concept of mutational signatures with isogenic cell models. Nat. Commun. 9, 1744 (2018).

    PubMed  PubMed Central  Google Scholar 

  87. Depetter, Y. et al. Selective pharmacological inhibitors of HDAC6 reveal biochemical activity but functional tolerance in cancer models. Int. J. Cancer 145, 735–747 (2019).

    CAS  PubMed  Google Scholar 

  88. Kasap, C., Elemento, O. & Kapoor, T. M. DrugTargetSeqR: a genomics- and CRISPR-Cas9-based method to analyze drug targets. Nat. Chem. Biol. 10, 626–628 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Evers, B. et al. CRISPR knockout screening outperforms shRNA and CRISPRi in identifying essential genes. Nat. Biotechnol. 34, 631–633 (2016).

    CAS  PubMed  Google Scholar 

  90. Knott, S. R. V. et al. A computational algorithm to predict shRNA potency. Mol. Cell 56, 796–807 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Sanson, K. R. et al. Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities. Nat. Commun. 9, 5416 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Shi, J. et al. Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains. Nat. Biotechnol. 33, 661–667 (2015). This article demonstrates how domain-specific CRISPR screening can be used to identify druggable cancer dependencies.

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Stegmeier, F., Hu, G., Rickles, R. J., Hannon, G. J. & Elledge, S. J. A lentiviral microRNA-based system for single-copy polymerase II-regulated RNA interference in mammalian cells. Proc. Natl Acad. Sci. USA 102, 13212–13217 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Ma, H. T., On, K. F., Tsang, Y. H. & Poon, R. Y. C. An inducible system for expression and validation of the specificity of short hairpin RNA in mammalian cells. Nucleic Acids Res. 35, e22 (2007).

    PubMed  PubMed Central  Google Scholar 

  95. Buehler, E., Chen, Y.-C. & Martin, S. C911: a bench-level control for sequence specific siRNA off-target effects. PLoS One 7, e51942 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Gilbert, L. A. et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 154, 442–451 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Larson, M. H. et al. CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat. Protoc. 8, 2180–2196 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Qi, L. S. et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 152, 1173–1183 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Liu, S. J. et al. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells. Science 355, eaah7111 (2017).

    Google Scholar 

  100. Zetsche, B. et al. Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163, 759–771 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Sanson, K. R. et al. Optimization of AsCas12a for combinatorial genetic screens in human cells. bioRxiv https://doi.org/10.1101/747170 (2019).

    Article  Google Scholar 

  102. Zetsche, B. et al. Multiplex gene editing by CRISPR–Cpf1 using a single crRNA array. Nat. Biotechnol. 35, 31–34 (2017).

    CAS  PubMed  Google Scholar 

  103. Dede, M., Kim, E. & Hart, T. Biases and blind-spots in genome-wide CRISPR knockout screens. bioRxiv https://doi.org/10.1101/2020.01.16.909606 (2020).

    Article  Google Scholar 

  104. Konermann, S. et al. Transcriptome engineering with RNA-targeting type VI-D CRISPR effectors. Cell 173, 665–676.e14 (2018). This article describes a new CRISPR system — CasRx/Cas13d — that can be used to eliminate target mRNA molecules with greater specificity than RNAi-mediated approaches.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Wessels, H.-H. et al. Massively parallel Cas13 screens reveal principles for guide RNA design. Nat. Biotechnol. https://doi.org/10.1038/s41587-020-0456-9 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Bondeson, D. P. et al. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat. Chem. Biol. 11, 611–617 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Zhang, X. et al. Protein targeting chimeric molecules specific for bromodomain and extra-terminal motif family proteins are active against pre-clinical models of multiple myeloma. Leukemia 32, 2224–2239 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Pei, H., Peng, Y., Zhao, Q. & Chen, Y. Small molecule PROTACs: an emerging technology for targeted therapy in drug discovery. RSC Adv. 9, 16967–16976 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Wilmington, S. R. & Matouschek, A. An Inducible system for rapid degradation of specific cellular proteins using proteasome adaptors. PLoS One 11, e0152679 (2016).

    PubMed  PubMed Central  Google Scholar 

  110. Gillet, J.-P. et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc. Natl Acad. Sci. USA 108, 18708–18713 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Hidalgo, M. et al. Patient derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 4, 998–1013 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Lampreht Tratar, U., Horvat, S. & Cemazar, M. Transgenic mouse models in cancer research. Front. Oncol. 8, 268 (2018).

    PubMed  PubMed Central  Google Scholar 

  113. Ertel, A., Verghese, A., Byers, S. W., Ochs, M. & Tozeren, A. Pathway-specific differences between tumor cell lines and normal and tumor tissue cells. Mol. Cancer 5, 55 (2006).

    PubMed  PubMed Central  Google Scholar 

  114. Goodspeed, A., Heiser, L. M., Gray, J. W. & Costello, J. C. Tumor-derived cell lines as molecular models of cancer pharmacogenomics. Mol. Cancer Res. 14, 3–13 (2016).

    CAS  PubMed  Google Scholar 

  115. Friberg, S. & Mattson, S. On the growth rates of human malignant tumors: implications for medical decision making. J. Surg. Oncol. 65, 284–297 (1997).

    CAS  PubMed  Google Scholar 

  116. Tubiana, M. Tumor cell proliferation kinetics and tumor growth rate. Acta Oncol. 28, 113–121 (1989).

    CAS  PubMed  Google Scholar 

  117. Ben-David, U. et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature 560, 325–330 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Imamura, Y. et al. Comparison of 2D- and 3D-culture models as drug-testing platforms in breast cancer. Oncol. Rep. 33, 1837–1843 (2015).

    CAS  PubMed  Google Scholar 

  119. Unger, C. et al. Modeling human carcinomas: physiologically relevant 3D models to improve anti-cancer drug development. Adv. Drug. Deliv. Rev. 79–80, 50–67 (2014).

    PubMed  Google Scholar 

  120. Yamada, K. M. & Cukierman, E. Modeling tissue morphogenesis and cancer in 3D. Cell 130, 601–610 (2007).

    CAS  PubMed  Google Scholar 

  121. Han, K. et al. CRISPR screens in cancer spheroids identify 3D growth-specific vulnerabilities. Nature 580, 136–141 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Birgersdotter, A., Sandberg, R. & Ernberg, I. Gene expression perturbation in vitro—a growing case for three-dimensional (3D) culture systems. Semin. Cancer Biol. 15, 405–412 (2005).

    PubMed  Google Scholar 

  123. Li, C. et al. Cell type and culture condition-dependent alternative splicing in human breast cancer cells revealed by splicing-sensitive microarrays. Cancer Res. 66, 1990–1999 (2006).

    CAS  PubMed  Google Scholar 

  124. Amann, A. et al. Development of a 3D angiogenesis model to study tumour – endothelial cell interactions and the effects of anti-angiogenic drugs. Sci. Rep. 7, 2963 (2017).

    PubMed  PubMed Central  Google Scholar 

  125. Day, C.-P., Merlino, G. & Van Dyke, T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell 163, 39–53 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Cooper, L. A. D. et al. The tumor microenvironment strongly impacts master transcriptional regulators and gene expression class of glioblastoma. Am. J. Pathol. 180, 2108–2119 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Gerashchenko, G. V. et al. Expression pattern of genes associated with tumor microenvironment in prostate cancer. Exp. Oncol. 40, 315–322 (2018).

    CAS  PubMed  Google Scholar 

  128. Kondou, R. et al. Classification of tumor microenvironment immune types based on immune response-associated gene expression. Int. J. Oncol. 54, 219–228 (2019).

    CAS  PubMed  Google Scholar 

  129. Wang, M. et al. Humanized mice in studying efficacy and mechanisms of PD-1-targeted cancer immunotherapy. FASEB J. 32, 1537–1549 (2018).

    CAS  PubMed  Google Scholar 

  130. Ben-David, U. et al. Patient-derived xenografts undergo mouse-specific tumor evolution. Nat. Genet. 49, 1567–1575 (2017). Although PDXs have sometimes been described as the gold standard model for faithfully capturing human cancer biology, this article demonstrates that passaging PDXs in mice can cause significant evolutionary divergence from their initial state.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Sachs, N. et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell 172, 373–386.e10 (2018).

    CAS  PubMed  Google Scholar 

  132. Boj, S. F. et al. Organoid models of human and mouse ductal pancreatic cancer. Cell 160, 324–338 (2015).

    CAS  PubMed  Google Scholar 

  133. Yang, H., Sun, L., Liu, M. & Mao, Y. Patient-derived organoids: a promising model for personalized cancer treatment. Gastroenterol. Rep. 6, 243–245 (2018).

    Google Scholar 

  134. Vlachogiannis, G. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359, 920–926 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Verissimo, C. S. et al. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. eLife 5, e18489 (2016).

    PubMed  PubMed Central  Google Scholar 

  136. Driehuis, E. & Clevers, H. CRISPR/Cas 9 genome editing and its applications in organoids. Am. J. Physiol. Gastrointest. Liver Physiol. 312, G257–G265 (2017).

    PubMed  Google Scholar 

  137. Kondo, J. & Inoue, M. Application of cancer organoid model for drug screening and personalized therapy. Cells 8, 470 (2019).

    CAS  PubMed Central  Google Scholar 

  138. Holderfield, M., Deuker, M. M., McCormick, F. & McMahon, M. Targeting RAF kinases for cancer therapy: BRAF mutated melanoma and beyond. Nat. Rev. Cancer 14, 455–467 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Jorda, R. et al. How selective are pharmacological inhibitors of cell-cycle-regulating cyclin-dependent kinases? J. Med. Chem. 61, 9105–9120 (2018).

    CAS  PubMed  Google Scholar 

  140. Cho, Y.-S., Kang, Y., Kim, K., Cha, Y.-J. & Cho, H.-S. The crystal structure of MPK38 in complex with OTSSP167, an orally administrative MELK selective inhibitor. Biochem. Biophys. Res. Commun. 447, 7–11 (2014).

    CAS  PubMed  Google Scholar 

  141. Gad, H. et al. MTH1 inhibition eradicates cancer by preventing sanitation of the dNTP pool. Nature 508, 215–221 (2014).

    CAS  PubMed  Google Scholar 

  142. Kettle, J. G. et al. Potent and selective inhibitors of MTH1 probe its role in cancer cell survival. J. Med. Chem. 59, 2346–2361 (2016).

    CAS  PubMed  Google Scholar 

  143. Kawamura, T. et al. Proteomic profiling of small-molecule inhibitors reveals dispensability of MTH1 for cancer cell survival. Sci. Rep. 6, 26521 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Patterson, J. C. et al. VISAGE reveals a targetable mitotic spindle vulnerability in cancer cells. Cell Syst. 9, 74–92.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Wacker, S. A., Houghtaling, B. R., Elemento, O. & Kapoor, T. M. Using transcriptome sequencing to identify mechanisms of drug action and resistance. Nat. Chem. Biol. 8, 235–237 (2012). This article describes a powerful approach that takes advantage of microsatellite-unstable cancer cells to identify mutations that confer resistance to drugs with unknown targets.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Szlachta, K. et al. CRISPR knockout screening identifies combinatorial drug targets in pancreatic cancer and models cellular drug response. Nat. Commun. 9, 4275 (2018).

    PubMed  PubMed Central  Google Scholar 

  147. Hess, G. T. et al. Directed evolution using dCas9-targeted somatic hypermutation in mammalian cells. Nat. Methods 13, 1036–1042 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Neggers, J. E. et al. Target identification of small molecules using large-scale CRISPR-Cas mutagenesis scanning of essential genes. Nat. Commun. 9, 502 (2018).

    PubMed  PubMed Central  Google Scholar 

  149. Ipsaro, J. J. et al. Rapid generation of drug-resistance alleles at endogenous loci using CRISPR-Cas9 indel mutagenesis. PLoS One 12, e0172177 (2017).

    PubMed  PubMed Central  Google Scholar 

  150. Gurden, M. D. et al. Naturally occurring mutations in the mps1 gene predispose cells to kinase inhibitor drug resistance. Cancer Res. 75, 3340–3354 (2015).

    CAS  PubMed  Google Scholar 

  151. Murai, J. et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 72, 5588–5599 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Antolin, A. A. et al. Objective, quantitative, data-driven assessment of chemical probes. Cell Chem. Biol. 25, 194–205.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Arrowsmith, C. H. et al. The promise and peril of chemical probes. Nat. Chem. Biol. 11, 536–541 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Blagg, J. & Workman, P. Choose and use your chemical probe wisely to explore cancer biology. Cancer Cell 32, 9–25 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Workman, P. & Collins, I. Probing the probes: fitness factors for small molecule tools. Chem. Biol. 17, 561–577 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Frye, S. V. The art of the chemical probe. Nat. Chem. Biol. 6, 159–161 (2010).

    CAS  PubMed  Google Scholar 

  157. Hunter, P. The reproducibility “crisis”. EMBO Rep. 18, 1493–1496 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Kaelin, W. G. Common pitfalls in preclinical cancer target validation. Nat. Rev. Cancer 17, 441–450 (2017). This is a highly instructive review that describes how flaws in assay design and experimental interpretation can affect the reproducibility of preclinical research.

    Google Scholar 

  159. Plesser, H. E. Reproducibility vs. replicability: a brief history of a confused terminology. Front. Neuroinform. 11, 76 (2018).

    PubMed  PubMed Central  Google Scholar 

  160. Behan, F. M. et al. Prioritization of cancer therapeutic targets using CRISPR–Cas9 screens. Nature 568, 511–516 (2019).

    CAS  PubMed  Google Scholar 

  161. Ghandi, M. et al. Next-generation characterization of the cancer cell line encyclopedia. Nature 569, 503–508 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. McDonald, E. R. et al. Project DRIVE: a compendium of cancer dependencies and synthetic lethal relationships uncovered by large-scale, deep RNAi screening. Cell 170, 577–592.e10 (2017).

    CAS  PubMed  Google Scholar 

  163. Dempster, J. M. et al. Agreement between two large pan-cancer CRISPR-Cas9 gene dependency datasets. bioRxiv https://doi.org/10.1101/604447 (2019).

    Article  Google Scholar 

  164. Rosenbluh, J. et al. Complementary information derived from CRISPR Cas9 mediated gene deletion and suppression. Nat. Commun. 8, 15403 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. El-Brolosy, M. A. & Stainier, D. Y. R. Genetic compensation: a phenomenon in search of mechanisms. PLoS Genet. 13, e1006780 (2017).

    PubMed  PubMed Central  Google Scholar 

  166. Hoelder, S., Clarke, P. A. & Workman, P. Discovery of small molecule cancer drugs: Successes, challenges and opportunities. Mol. Oncol. 6, 155–176 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. von Bubnoff, N., Peschel, C. & Duyster, J. Resistance of Philadelphia-chromosome positive leukemia towards the kinase inhibitor imatinib (STI571, Glivec): a targeted oncoprotein strikes back. Leukemia 17, 829–838 (2003).

    Google Scholar 

  168. Chandrasekhar, C., Kumar, P. S. & Sarma, P. V. G. K. Novel mutations in the kinase domain of BCR-ABL gene causing imatinib resistance in chronic myeloid leukemia patients. Sci. Rep. 9, 2412 (2019).

    PubMed  PubMed Central  Google Scholar 

  169. Yamamoto, M., Kurosu, T., Kakihana, K., Mizuchi, D. & Miura, O. The two major imatinib resistance mutations E255K and T315I enhance the activity of BCR/ABL fusion kinase. Biochem. Biophys. Res. Commun. 319, 1272–1275 (2004).

    CAS  PubMed  Google Scholar 

  170. Gorre, M. E., Ellwood-Yen, K., Chiosis, G., Rosen, N. & Sawyers, C. L. BCR-ABL point mutants isolated from patients with imatinib mesylate–resistant chronic myeloid leukemia remain sensitive to inhibitors of the BCR-ABL chaperone heat shock protein 90. Blood 100, 3041–3044 (2002).

    CAS  PubMed  Google Scholar 

  171. Gottesman, M. M. Mechanisms of cancer drug resistance. Annu. Rev. Med. 53, 615–627 (2002).

    CAS  PubMed  Google Scholar 

  172. Phillips, R. E. et al. Target identification reveals lanosterol synthase as a vulnerability in glioma. Proc. Natl Acad. Sci. USA 116, 7957–7962 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Thomenius, M. J. et al. Small molecule inhibitors and CRISPR/Cas9 mutagenesis demonstrate that SMYD2 and SMYD3 activity are dispensable for autonomous cancer cell proliferation. PLoS One 13, e0197372 (2018).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the members of the Sheltzer laboratory for their helpful comments on the manuscript. Research in the Sheltzer laboratory is supported by an NIH Early Independence award (1DP5OD021385), a Breast Cancer Alliance Young Investigator award, a Damon Runyon-Rachleff Innovation award, a Gates Foundation Innovative Technology Solutions grant and a CSHL–Northwell Health Translational Cancer Research grant. A.L. is supported by an NSF Graduate Research Fellowship and a Gabilan Stanford Graduate Fellowship. The figures in this Review were made with Biorender.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Jason M. Sheltzer.

Ethics declarations

Competing interests

A.L. and J.M.S. are co-founders of Meliora Therapeutics. J.M.S. has received consulting fees from Ono Pharmaceutical Co. and is a member of the advisory board of Tyra Biosciences.

Additional information

Peer review information

Nature Reviews Genetics thanks T. Hart and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Chemical Probes Portal: http://www.chemicalprobes.org/

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lin, A., Sheltzer, J.M. Discovering and validating cancer genetic dependencies: approaches and pitfalls. Nat Rev Genet 21, 671–682 (2020). https://doi.org/10.1038/s41576-020-0247-7

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41576-020-0247-7

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer