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.

  • Perspective
  • Published:

Non-genetic mechanisms of therapeutic resistance in cancer

Abstract

Therapeutic resistance continues to be an indominable foe in our ambition for curative cancer treatment. Recent insights into the molecular determinants of acquired treatment resistance in the clinical and experimental setting have challenged the widely held view of sequential genetic evolution as the primary cause of resistance and brought into sharp focus a range of non-genetic adaptive mechanisms. Notably, the genetic landscape of the tumour and the non-genetic mechanisms used to escape therapy are frequently linked. Remarkably, whereas some oncogenic mutations allow the cancer cells to rapidly adapt their transcriptional and/or metabolic programme to meet and survive the therapeutic pressure, other oncogenic drivers convey an inherent cellular plasticity to the cancer cell enabling lineage switching and/or the evasion of anticancer immunosurveillance. The prevalence and diverse array of non-genetic resistance mechanisms pose a new challenge to the field that requires innovative strategies to monitor and counteract these adaptive processes. In this Perspective we discuss the key principles of non-genetic therapy resistance in cancer. We provide a perspective on the emerging data from clinical studies and sophisticated cancer models that have studied various non-genetic resistance pathways and highlight promising therapeutic avenues that may be used to negate and/or counteract the non-genetic adaptive pathways.

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

Access options

Buy this article

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

Fig. 1: Interpretation of mechanisms of resistance are influenced by the resolution of the technology.
Fig. 2: Models of genetic and non-genetic therapy resistance.
Fig. 3: Mechanisms of transient and stable non-genetic resistance.
Fig. 4: Cell of origin and genetic composition influence mechanisms of resistance.
Fig. 5: Cellular plasticity as a mechanism to evade anticancer immunosurveillance.
Fig. 6: Potential therapeutic strategies to counteract non-genetic resistance mechanisms.

Similar content being viewed by others

References

  1. Scott, A. M., Wolchok, J. D. & Old, L. J. Antibody therapy of cancer. Nat. Rev. Cancer 12, 278–287 (2012).

    CAS  PubMed  Google Scholar 

  2. Mayes, P. A., Hance, K. W. & Hoos, A. The promise and challenges of immune agonist antibody development in cancer. Nat. Rev. Drug. Discov. 17, 509–527 (2018).

    CAS  PubMed  Google Scholar 

  3. Smaglo, B. G., Aldeghaither, D. & Weiner, L. M. The development of immunoconjugates for targeted cancer therapy. Nat. Rev. Clin. Oncol. 11, 637–648 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Meric-Bernstam, F. & Mills, G. B. Overcoming implementation challenges of personalized cancer therapy. Nat. Rev. Clin. Oncol. 9, 542–548 (2012).

    CAS  PubMed  Google Scholar 

  5. Fong, C. Y. et al. BET inhibitor resistance emerges from leukaemia stem cells. Nature 525, 538–542 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Kim, C. et al. Chemoresistance evolution in triple-negative breast cancer delineated by single-cell sequencing. Cell 173, 879–893.e13 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Pisco, A. O. & Huang, S. Non-genetic cancer cell plasticity and therapy-induced stemness in tumour relapse: ‘What does not kill me strengthens me’. Br. J. Cancer 112, 1725–1732 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Shlush, L. I. et al. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature 547, 104–108 (2017).

    CAS  PubMed  Google Scholar 

  9. Hugo, W. et al. Non-genomic and immune evolution of melanoma acquiring MAPKi resistance. Cell 162, 1271–1285 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Dobson, S. M. et al. Relapse-fated latent diagnosis subclones in acute B lineage leukemia are drug tolerant and possess distinct metabolic programs. Cancer Discov. 10, 568–587 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. DeNicola, G. M. et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 475, 106–109 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Nakasone, E. S. et al. Imaging tumor-stroma interactions during chemotherapy reveals contributions of the microenvironment to resistance. Cancer Cell 21, 488–503 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Rosenzweig, S. A. Acquired resistance to drugs targeting receptor tyrosine kinases. Biochem. Pharmacol. 83, 1041–1048 (2012).

    CAS  PubMed  Google Scholar 

  14. Kim, T. K., Herbst, R. S. & Chen, L. Defining and understanding adaptive resistance in cancer immunotherapy. Trends Immunol. 39, 624–631 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Beltran, H. et al. Whole-exome sequencing of metastatic cancer and biomarkers of treatment response. JAMA Oncol. 1, 466–474 (2015).

    PubMed  PubMed Central  Google Scholar 

  16. Pogrebniak, K. L. & Curtis, C. Harnessing tumor evolution to circumvent resistance. Trends Genet. 34, 639–651 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Sharma, S. V. et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 141, 69–80 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Menon, D. R. et al. A stress-induced early innate response causes multidrug tolerance in melanoma. Oncogene 34, 4545 (2015).

    CAS  PubMed  Google Scholar 

  19. Dallas, N. A. et al. Chemoresistant colorectal cancer cells, the cancer stem cell phenotype, and increased sensitivity to insulin-like growth factor-I receptor inhibition. Cancer Res. 69, 1951–1957 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Xue, Z. et al. Identification of cancer stem cells in vincristine preconditioned SGC7901 gastric cancer cell line. J. Cell Biochem. 113, 302–312 (2012).

    CAS  PubMed  Google Scholar 

  21. Pirozzi, G. et al. Epithelial to mesenchymal transition by TGFbeta-1 induction increases stemness characteristics in primary non small cell lung cancer cell line. PLoS ONE 6, e21548 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Gupta, P. B. et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 146, 633–644 (2011).

    CAS  PubMed  Google Scholar 

  23. Polyak, K. & Weinberg, R. A. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Nat. Rev. Cancer 9, 265–273 (2009).

    CAS  PubMed  Google Scholar 

  24. Singh, A. & Settleman, J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741–4751 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hoek, K. S. & Goding, C. R. Cancer stem cells versus phenotype-switching in melanoma. Pigment Cell Melanoma Res. 23, 746–759 (2010).

    CAS  PubMed  Google Scholar 

  26. Kemper, K., de Goeje, P. L., Peeper, D. S. & van Amerongen, R. Phenotype switching: tumor cell plasticity as a resistance mechanism and target for therapy. Cancer Res. 74, 5937–5941 (2014).

    CAS  PubMed  Google Scholar 

  27. Shaffer, S. M. et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature 546, 431–435 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Rambow, F. et al. Toward minimal residual disease-directed therapy in melanoma. Cell 174, 843–855 e19 (2018).

    CAS  PubMed  Google Scholar 

  29. Boshuizen, J. et al. Cooperative targeting of melanoma heterogeneity with an AXL antibody-drug conjugate and BRAF/MEK inhibitors. Nat. Med. 24, 203–212 (2018).

    CAS  PubMed  Google Scholar 

  30. Lee, J. H. et al. Transcriptional downregulation of MHC class I and melanoma de- differentiation in resistance to PD-1 inhibition. Nat. Commun. 11, 1897 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Koyama, S. et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 7, 10501 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fallahi-Sichani, M. et al. Adaptive resistance of melanoma cells to RAF inhibition via reversible induction of a slowly dividing de-differentiated state. Mol. Syst. Biol. 13, 905 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Bigger, J. Treatment of staphylococcal infections with penicillin by intermittent sterilisation. Lancet 244, 497–500 (1944).

    Google Scholar 

  34. Holden, D. W. Microbiology. Persisters unmasked. Science 347, 30–32 (2015).

    CAS  PubMed  Google Scholar 

  35. Su, Y. et al. Single-cell analysis resolves the cell state transition and signaling dynamics associated with melanoma drug-induced resistance. Proc. Natl Acad. Sci. USA 114, 13679–13684 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tsoi, J. et al. Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell 33, 890–904.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ding, L. et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature 481, 506–510 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Garg, M. et al. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood 126, 2491–2501 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, S. et al. Distinct evolution and dynamics of epigenetic and genetic heterogeneity in acute myeloid leukemia. Nat. Med. 22, 792–799 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Shiba, N. et al. Whole-exome sequencing reveals the spectrum of gene mutations and the clonal evolution patterns in paediatric acute myeloid leukaemia. Br. J. Haematol. 175, 476–489 (2016).

    CAS  PubMed  Google Scholar 

  41. Rizos, H. et al. BRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impact. Clin. Cancer Res. 20, 1965–1977 (2014).

    CAS  PubMed  Google Scholar 

  42. Shi, H. et al. A novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibition. Cancer Discov. 4, 69–79 (2014).

    CAS  PubMed  Google Scholar 

  43. Boumahdi, S. & de Sauvage, F. J. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat. Rev. Drug Discov. 19, 39–56 (2020).

    CAS  PubMed  Google Scholar 

  44. Bell, C. C. et al. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia. Nat. Commun. 10, 2723 (2019).

    PubMed  PubMed Central  Google Scholar 

  45. Chen, A. F. et al. GRHL2-dependent enhancer switching maintains a pluripotent stem cell transcriptional subnetwork after exit from naive pluripotency. Cell Stem Cell 23, 226–238.e4 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kieffer-Kwon, K. R. et al. Interactome maps of mouse gene regulatory domains reveal basic principles of transcriptional regulation. Cell 155, 1507–1520 (2013).

    CAS  PubMed  Google Scholar 

  47. Iniguez, A. B. et al. Resistance to epigenetic-targeted therapy engenders tumor cell vulnerabilities associated with enhancer remodeling. Cancer Cell 34, 922–938.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hinohara, K. et al. KDM5 histone demethylase activity links cellular transcriptomic heterogeneity to therapeutic resistance. Cancer Cell 34, 939–953.e9 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Liau, B. B. et al. Adaptive chromatin remodeling drives glioblastoma stem cell plasticity and drug tolerance. Cell Stem Cell 20, 233–246.e7 (2017).

    CAS  PubMed  Google Scholar 

  50. Zawistowski, J. S. et al. Enhancer remodeling during adaptive bypass to MEK inhibition is attenuated by pharmacologic targeting of the P-TEFb complex. Cancer Discov. 7, 302–321 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Stergachis, A. B. et al. Developmental fate and cellular maturity encoded in human regulatory DNA landscapes. Cell 154, 888–903 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Pastushenko, I. et al. Identification of the tumour transition states occurring during EMT. Nature 556, 463–468 (2018).

    CAS  PubMed  Google Scholar 

  53. Puisieux, A., Pommier, R. M., Morel, A. P. & Lavial, F. Cellular pliancy and the multistep process of tumorigenesis. Cancer Cell 33, 164–172 (2018).

    CAS  PubMed  Google Scholar 

  54. Morel, A. P. et al. A stemness-related ZEB1-MSRB3 axis governs cellular pliancy and breast cancer genome stability. Nat. Med. 23, 568–578 (2017).

    CAS  PubMed  Google Scholar 

  55. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    PubMed  PubMed Central  Google Scholar 

  56. Sanchez-Danes, A. et al. A slow-cycling LGR5 tumour population mediates basal cell carcinoma relapse after therapy. Nature 562, 434–438 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    CAS  PubMed  Google Scholar 

  58. Umkehrer, C. et al. Isolating live cell clones from barcoded populations using CRISPRa-inducible reporters. Nat. Biotechnol. https://doi.org/10.1038/s41587-020-0614-0 (2020).

    Article  PubMed  Google Scholar 

  59. Moudgil, A. et al. Self-reporting transposons enable simultaneous readout of gene expression and transcription factor binding in single cells. Cell 182, 992–1008.e21 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Weinreb, C., Rodriguez-Fraticelli, A., Camargo, F. D. & Klein, A. M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 367, eaaw3381 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Russo, M. et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science 366, 1473–1480 (2019).

    CAS  PubMed  Google Scholar 

  62. Cipponi, A. et al. MTOR signaling orchestrates stress-induced mutagenesis, facilitating adaptive evolution in cancer. Science 368, 1127–1131 (2020).

    CAS  PubMed  Google Scholar 

  63. Kadoch, C. Diverse compositions and functions of chromatin remodeling machines in cancer. Sci. Transl Med. 11, eaay1018 (2019).

    PubMed  PubMed Central  Google Scholar 

  64. Agarwal, R. et al. Dynamic molecular monitoring reveals that SWI-SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat. Med. 25, 119–129 (2019).

    CAS  PubMed  Google Scholar 

  65. Xu, G. et al. ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat. Genet. 52, 198–207 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Nagarajan, S. et al. ARID1A influences HDAC1/BRD4 activity, intrinsic proliferative capacity and breast cancer treatment response. Nat. Genet. 52, 187–197 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Quintanal-Villalonga, A. et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat. Rev. Clin. Oncol. 17, 360–371 (2020).

    PubMed  PubMed Central  Google Scholar 

  68. Oser, M. G., Niederst, M. J., Sequist, L. V. & Engelman, J. A. Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol. 16, e165–e172 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Davies, A. H., Beltran, H. & Zoubeidi, A. Cellular plasticity and the neuroendocrine phenotype in prostate cancer. Nat. Rev. Urol. 15, 271–286 (2018).

    CAS  PubMed  Google Scholar 

  70. Biehs, B. et al. A cell identity switch allows residual BCC to survive Hedgehog pathway inhibition. Nature 562, 429–433 (2018).

    CAS  PubMed  Google Scholar 

  71. Sutherland, K. D. et al. Cell of origin of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 19, 754–764 (2011).

    CAS  PubMed  Google Scholar 

  72. Sutherland, K. D. et al. Multiple cells-of-origin of mutant K-Ras-induced mouse lung adenocarcinoma. Proc. Natl Acad. Sci. USA 111, 4952–4957 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Lin, C. et al. Alveolar type II cells possess the capability of initiating lung tumor development. PLoS ONE 7, e53817 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. George, J. et al. Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47–53 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Lee, J. K. et al. Clonal history and genetic predictors of transformation into small-cell carcinomas from lung adenocarcinomas. J. Clin. Oncol. 35, 3065–3074 (2017).

    CAS  PubMed  Google Scholar 

  76. Imielinski, M. et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150, 1107–1120 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Miettinen, P. J. et al. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature 376, 337–341 (1995).

    CAS  PubMed  Google Scholar 

  78. Sequist, L. V. et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl Med. 3, 75ra26 (2011).

    PubMed  PubMed Central  Google Scholar 

  79. Takegawa, N. et al. Transformation of ALK rearrangement-positive adenocarcinoma to small-cell lung cancer in association with acquired resistance to alectinib. Ann. Oncol. 27, 953–955 (2016).

    CAS  PubMed  Google Scholar 

  80. Adelstein, D. J., Tomashefski, J. F. Jr. Snow, N. J., Horrigan, T. P. & Hines, J. D. Mixed small cell and non-small cell lung cancer. Chest 89, 699–704 (1986).

    CAS  PubMed  Google Scholar 

  81. Marcoux, N. et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J. Clin. Oncol. 37, 278–285 (2019).

    CAS  PubMed  Google Scholar 

  82. Ku, S. Y. et al. Rb1 and Trp53 cooperate to suppress prostate cancer lineage plasticity, metastasis, and antiandrogen resistance. Science 355, 78–83 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Mu, P. et al. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science 355, 84–88 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Walter, D. M. et al. RB constrains lineage fidelity and multiple stages of tumour progression and metastasis. Nature 569, 423–427 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Park, J. W. et al. Reprogramming normal human epithelial tissues to a common, lethal neuroendocrine cancer lineage. Science 362, 91–95 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Dardenne, E. et al. N-Myc induces an EZH2-mediated transcriptional program driving neuroendocrine prostate cancer. Cancer Cell 30, 563–577 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Burr, M. L. et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 36, 385–401.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Bernards, R., Dessain, S. K. & Weinberg, R. A. N-myc amplification causes down-modulation of MHC class I antigen expression in neuroblastoma. Cell 47, 667–674 (1986).

    CAS  PubMed  Google Scholar 

  89. Restifo, N. P. et al. Identification of human cancers deficient in antigen processing. J. Exp. Med. 177, 265–272 (1993).

    CAS  PubMed  Google Scholar 

  90. Paulson, K. G. et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat. Commun. 9, 3868 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Agudo, J. et al. Quiescent tissue stem cells evade immune surveillance. Immunity 48, 271–285.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Landsberg, J. et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature 490, 412–416 (2012).

    CAS  PubMed  Google Scholar 

  93. Mehta, A. et al. Immunotherapy resistance by inflammation-induced dedifferentiation. Cancer Discov. 8, 935–943 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Majzner, R. G. & Mackall, C. L. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 8, 1219–1226 (2018).

    CAS  PubMed  Google Scholar 

  95. Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).

    PubMed  PubMed Central  Google Scholar 

  96. Maude, S. L. et al. Sustained remissions with CD19-specific chimeric antigen receptor (CAR)-modified T cells in children with relapsed/refractory ALL. J. Clin. Oncol. 34, 3011–3011 (2016).

    Google Scholar 

  97. Park, J. H. et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N. Engl. J. Med. 378, 449–459 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Gardner, R. et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 127, 2406–2410 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Turtle, C. J. et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 126, 2123–2138 (2016).

    PubMed  PubMed Central  Google Scholar 

  100. Andersson, A. K. et al. The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat. Genet. 47, 330–337 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N. Engl. J. Med. 368, 2059–2074 (2013).

    PubMed Central  Google Scholar 

  102. Greaves, M. When one mutation is all it takes. Cancer Cell 27, 433–434 (2015).

    CAS  PubMed  Google Scholar 

  103. Krivtsov, A. V. et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 442, 818–822 (2006).

    CAS  PubMed  Google Scholar 

  104. Jacoby, E. et al. CD19 CAR immune pressure induces B-precursor acute lymphoblastic leukaemia lineage switch exposing inherent leukaemic plasticity. Nat. Commun. 7, 12320 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Zoghbi, A., Zur Stadt, U., Winkler, B., Muller, I. & Escherich, G. Lineage switch under blinatumomab treatment of relapsed common acute lymphoblastic leukemia without MLL rearrangement. Pediatr. Blood Cancer 64, e26594 (2017).

    Google Scholar 

  106. Rayes, A., McMasters, R. L. & O’Brien, M. M. Lineage switch in MLL-rearranged infant leukemia following CD19-directed therapy. Pediatr. Blood Cancer 63, 1113–1115 (2016).

    CAS  PubMed  Google Scholar 

  107. Xue, Y. et al. An approach to suppress the evolution of resistance in BRAF(V600E)-mutant cancer. Nat. Med. 23, 929–937 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Greaves, M. Evolutionary determinants of cancer. Cancer Discov. 5, 806–820 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. McGranahan, N. & Swanton, C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell 168, 613–628 (2017).

    CAS  PubMed  Google Scholar 

  110. Dagogo-Jack, I. & Shaw, A. T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 15, 81–94 (2018).

    CAS  PubMed  Google Scholar 

  111. Chong, C. R. & Janne, P. A. The quest to overcome resistance to EGFR-targeted therapies in cancer. Nat. Med. 19, 1389–1400 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Thress, K. S. et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat. Med. 21, 560–562 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Siravegna, G. et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 21, 827 (2015).

    CAS  PubMed  Google Scholar 

  114. Diaz, L. A. Jr. et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 486, 537–540 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Miller, T. W., Balko, J. M. & Arteaga, C. L. Phosphatidylinositol 3-kinase and antiestrogen resistance in breast cancer. J. Clin. Oncol. 29, 4452–4461 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Christie, E. L. et al. Reversion of BRCA1/2 germline mutations detected in circulating tumor DNA from patients with high-grade serous ovarian cancer. J. Clin. Oncol. 35, 1274–1280 (2017).

    CAS  PubMed  Google Scholar 

  117. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. van Galen, P. et al. Single-cell RNA-Seq reveals AML hierarchies relevant to disease progression and immunity. Cell 176, 1265–1281.e24 (2019).

    PubMed  PubMed Central  Google Scholar 

  119. Miyamoto, D. T. et al. RNA-Seq of single prostate CTCs implicates noncanonical Wnt signaling in antiandrogen resistance. Science 349, 1351–1356 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Cheng, Y. H. et al. Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat. Commun. 10, 2163 (2019).

    PubMed  PubMed Central  Google Scholar 

  121. Baryawno, N. et al. A cellular taxonomy of the bone marrow stroma in homeostasis and leukemia. Cell 177, 1915–1932.e16 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Rodriguez-Meira, A. et al. Unravelling intratumoral heterogeneity through high-sensitivity single-cell mutational analysis and parallel RNA sequencing. Mol. Cell 73, 1292–1305.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Macaulay, I. C., Ponting, C. P. & Voet, T. Single-cell multiomics: multiple measurements from single cells. Trends Genet. 33, 155–168 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Rozenblatt-Rosen, O. et al. The human tumor atlas network: charting tumor transitions across space and time at single-cell resolution. Cell 181, 236–249 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Tung, P. Y. et al. Batch effects and the effective design of single-cell gene expression studies. Sci. Rep. 7, 39921 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Stahl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    CAS  PubMed  Google Scholar 

  127. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 361, eaat5691 (2018).

    PubMed  PubMed Central  Google Scholar 

  128. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    CAS  PubMed  Google Scholar 

  129. Svedlund, J. et al. Generation of in situ sequencing based OncoMaps to spatially resolve gene expression profiles of diagnostic and prognostic markers in breast cancer. EBioMedicine 48, 212–223 (2019).

    PubMed  PubMed Central  Google Scholar 

  130. Xia, C., Fan, J., Emanuel, G., Hao, J. & Zhuang, X. Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc. Natl Acad. Sci. USA 116, 19490–19499 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Rapino, F. et al. Codon-specific translation reprogramming promotes resistance to targeted therapy. Nature 558, 605–609 (2018).

    CAS  PubMed  Google Scholar 

  132. Garcia-Jimenez, C. & Goding, C. R. Starvation and pseudo-starvation as drivers of cancer metastasis through translation reprogramming. Cell Metab. 29, 254–267 (2019).

    CAS  PubMed  Google Scholar 

  133. Jewer, M. et al. Translational control of breast cancer plasticity. Nat. Commun. 11, 2498 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Giesen, C. et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 11, 417–422 (2014).

    CAS  PubMed  Google Scholar 

  135. Jackson, H. W. et al. The single-cell pathology landscape of breast cancer. Nature 578, 615–620 (2020).

    CAS  PubMed  Google Scholar 

  136. Schulz, D. et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular resolution in breast cancer tissue samples by mass cytometry. Cell Syst. 6, 25–36.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Saez-Ayala, M. et al. Directed phenotype switching as an effective antimelanoma strategy. Cancer Cell 24, 105–119 (2013).

    CAS  PubMed  Google Scholar 

  138. Das Thakur, M. et al. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 494, 251–255 (2013).

    PubMed  PubMed Central  Google Scholar 

  139. Venkatesh, V. et al. Targeting Notch signalling pathway of cancer stem cells. Stem Cell Investig. 5, 5 (2018).

    PubMed  PubMed Central  Google Scholar 

  140. Amakye, D., Jagani, Z. & Dorsch, M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat. Med. 19, 1410–1422 (2013).

    CAS  PubMed  Google Scholar 

  141. Nusse, R. & Clevers, H. Wnt/beta-catenin signaling, disease, and emerging therapeutic modalities. Cell 169, 985–999 (2017).

    CAS  PubMed  Google Scholar 

  142. MacPherson, L. et al. HBO1 is required for the maintenance of leukaemia stem cells. Nature 577, 266–270 (2020).

    CAS  PubMed  Google Scholar 

  143. Uckelmann, H. J. et al. Therapeutic targeting of preleukemia cells in a mouse model of NPM1 mutant acute myeloid leukemia. Science 367, 586–590 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Codd, A. S., Kanaseki, T., Torigo, T. & Tabi, Z. Cancer stem cells as targets for immunotherapy. Immunology 153, 304–314 (2018).

    CAS  PubMed  Google Scholar 

  145. Shi, Y., Du, L., Lin, L. & Wang, Y. Tumour-associated mesenchymal stem/stromal cells: emerging therapeutic targets. Nat. Rev. Drug Discov. 16, 35–52 (2017).

    CAS  PubMed  Google Scholar 

  146. Friedmann Angeli, J. P., Krysko, D. V. & Conrad, M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 19, 405–414 (2019).

    CAS  PubMed  Google Scholar 

  147. Viswanathan, V. S. et al. Dependency of a therapy-resistant state of cancer cells on a lipid peroxidase pathway. Nature 547, 453–457 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Hangauer, M. J. et al. Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition. Nature 551, 247–250 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Kinsey, C. G. et al. Protective autophagy elicited by RAF–>MEK–>ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat. Med. 25, 620–627 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Roesch, A. et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 23, 811–825 (2013).

    CAS  PubMed  Google Scholar 

  151. McConkey, D. J. The integrated stress response and proteotoxicity in cancer therapy. Biochem. Biophys. Res. Commun. 482, 450–453 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Rzymski, T., Milani, M., Singleton, D. C. & Harris, A. L. Role of ATF4 in regulation of autophagy and resistance to drugs and hypoxia. Cell Cycle 8, 3838–3847 (2009).

    CAS  PubMed  Google Scholar 

  153. Falletta, P. et al. Translation reprogramming is an evolutionarily conserved driver of phenotypic plasticity and therapeutic resistance in melanoma. Genes. Dev. 31, 18–33 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272–286 (2014).

    CAS  PubMed  Google Scholar 

  155. Corces, M. R. et al. The chromatin accessibility landscape of primary human cancers. Science 362, eaav1898 (2018).

    PubMed  PubMed Central  Google Scholar 

  156. Salami, J. & Crews, C. M. Waste disposal-an attractive strategy for cancer therapy. Science 355, 1163–1167 (2017).

    CAS  PubMed  Google Scholar 

  157. Michalak, E. M., Burr, M. L., Bannister, A. J. & Dawson, M. A. The roles of DNA, RNA and histone methylation in ageing and cancer. Nat. Rev. Mol. Cell Biol. 20, 573–589 (2019).

    CAS  PubMed  Google Scholar 

  158. Tyler, D. S. et al. Click chemistry enables preclinical evaluation of targeted epigenetic therapies. Science 356, 1397–1401 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Dawson, M. A. The cancer epigenome: concepts, challenges, and therapeutic opportunities. Science 355, 1147–1152 (2017).

    CAS  PubMed  Google Scholar 

  160. Sharma, A. et al. Longitudinal single-cell RNA sequencing of patient-derived primary cells reveals drug-induced infidelity in stem cell hierarchy. Nat. Commun. 9, 4931 (2018).

    PubMed  PubMed Central  Google Scholar 

  161. Knoechel, B. et al. An epigenetic mechanism of resistance to targeted therapy in T cell acute lymphoblastic leukemia. Nat. Genet. 46, 364–370 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Wei, A. H. et al. The QUAZAR AML-001 maintenance trial: results of a phase III international, randomized, double-blind, placebo-controlled study of CC-486 (oral formulation of azacitidine) in patients with acute myeloid leukemia (AML) in first remission. Blood 134, LBA-3 (2019).

    Google Scholar 

  163. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976).

    CAS  PubMed  Google Scholar 

  164. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Morrissy, A. S. et al. Spatial heterogeneity in medulloblastoma. Nat. Genet. 49, 780–788 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    CAS  PubMed  Google Scholar 

  167. Hu, Z., Li, Z., Ma, Z. & Curtis, C. Multi-cancer analysis of clonality and the timing of systemic spread in paired primary tumors and metastases. Nat. Genet. 52, 701–708 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Patch, A. M. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015).

    CAS  PubMed  Google Scholar 

  169. Savas, P. et al. The subclonal architecture of metastatic breast cancer: results from a prospective community-based rapid autopsy program “CASCADE”. PLoS Med. 13, e1002204 (2016).

    PubMed  PubMed Central  Google Scholar 

  170. Chen, J. et al. Myelodysplastic syndrome progression to acute myeloid leukemia at the stem cell level. Nat. Med. 25, 103–110 (2019).

    CAS  PubMed  Google Scholar 

  171. Abbosh, C. et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 545, 446–451 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Murtaza, M. et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013).

    CAS  PubMed  Google Scholar 

  173. Scherer, F. et al. Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci. Transl Med. 8, 364ra155 (2016).

    PubMed  PubMed Central  Google Scholar 

  174. Yeh, P. et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat. Commun. 8, 14756 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank all the members of the J.-C.M., S.-J.D. and M.A.D laboratories for helpful discussions related to the concepts presents in this Perspective. The authors thank N. Dawson for help with figure construction and graphical illustrations. The authors thank the following funders for support: Cancer Council Victoria for a Sir Edward Dunlop Research Fellowship and Howard Hughes Medical Institute for an international research scholarship (M.A.D) and CSL for a CSL Centenary Fellowship (S.-J.D).

Author information

Authors and Affiliations

Authors

Contributions

All authors made substantial contributions to researching data for the article, the discussion of content, writing of the manuscript and editing of it before final submission.

Corresponding authors

Correspondence to Jean-Christophe Marine, Sarah-Jane Dawson or Mark A. Dawson.

Ethics declarations

Competing interests

M.A.D. has been a member of advisory boards for Cancer Therapeutics CRC, Storm Therapeutics, Celgene and Cambridge Epigenetix. S.-J.D. has been a member of the advisory board for AstraZeneca. The S.-J.D. laboratory has received research funding from Genentech. The M.A.D. and S.-J.D. laboratories receive research funding from Cancer Therapeutics CRC. J.-C.M. declares no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks J. Carroll, U. McDermott 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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Marine, JC., Dawson, SJ. & Dawson, M.A. Non-genetic mechanisms of therapeutic resistance in cancer. Nat Rev Cancer 20, 743–756 (2020). https://doi.org/10.1038/s41568-020-00302-4

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-00302-4

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