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:

Unravelling the biology of SCLC: implications for therapy

Key Points

  • Small-cell lung cancer (SCLC) is a high-grade neuroendocrine tumour associated with a poor overall survival, and limited progress has been made in the treatment of this disease over the past three decades

  • Over the past 5 years, advances in our understanding of multiple aspects of the biology of SCLC have led to the development of new therapies that are currently under clinical investigation

  • Poly [ADP-ribose] polymerase (PARP) is abundantly expressed in SCLC and is involved in DNA-damage repair; clinical trials of the PARP inhibitors veliparib, olaparib, and talazoparib are ongoing in patients with SCLC

  • Enhancer of zeste homologue 2 (EZH2) is a regulator of chromatin remodelling that can drive acquired chemoresistance; therapeutic targeting of EZH2 might augment and extend the durability of chemotherapy responses

  • Delta-like protein 3 (DLL3) is an inhibitory Notch ligand that is overexpressed in many SCLCs; rovalpituzumab tesirine (Rova-T), an anti-DLL3-antibody–drug conjugate, has shown promising activity in preclinical and early phase clinical studies

  • SCLC has a high mutational burden, raising hopes regarding immunotherapy, and immune-checkpoint blockade has shown encouraging clinical activity in patients with this disease, despite typically low tumoural expression of immune-checkpoint proteins

Abstract

Small-cell lung cancer (SCLC) is an aggressive malignancy associated with a poor prognosis. First-line treatment has remained unchanged for decades, and a paucity of effective treatment options exists for recurrent disease. Nonetheless, advances in our understanding of SCLC biology have led to the development of novel experimental therapies. Poly [ADP-ribose] polymerase (PARP) inhibitors have shown promise in preclinical models, and are under clinical investigation in combination with cytotoxic therapies and inhibitors of cell-cycle checkpoints.Preclinical data indicate that targeting of histone-lysine N-methyltransferase EZH2, a regulator of chromatin remodelling implicated in acquired therapeutic resistance, might augment and prolong chemotherapy responses. High expression of the inhibitory Notch ligand Delta-like protein 3 (DLL3) in most SCLCs has been linked to expression of Achaete-scute homologue 1 (ASCL1; also known as ASH-1), a key transcription factor driving SCLC oncogenesis; encouraging preclinical and clinical activity has been demonstrated for an anti-DLL3-antibody–drug conjugate. The immune microenvironment of SCLC seems to be distinct from that of other solid tumours, with few tumour-infiltrating lymphocytes and low levels of the immune-checkpoint protein programmed cell death 1 ligand 1 (PD-L1). Nonetheless, immunotherapy with immune-checkpoint inhibitors holds promise for patients with this disease, independent of PD-L1 status. Herein, we review the progress made in uncovering aspects of the biology of SCLC and its microenvironment that are defining new therapeutic strategies and offering renewed hope for patients.

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

Figure 1: Timeline of therapeutic advances for small-cell lung cancer (SCLC).
Figure 2: Signalling pathways and physiological domains that are the focus of experimental targeted therapies for small-cell lung cancer (SCLC).

Similar content being viewed by others

References

  1. Ferlay, J. et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 136, E359–E386 (2015).

    CAS  PubMed  Google Scholar 

  2. Torre, L. A. et al. Global cancer statistics, 2012. CA Cancer J. Clin. 65, 87–108 (2015).

    PubMed  Google Scholar 

  3. Lozano, R. et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2095–2128 (2012).

    PubMed  Google Scholar 

  4. Nicholson, A. G. et al. The International Association for the Study of Lung Cancer Lung Cancer staging project: proposals for the revision of the clinical and pathologic staging of small cell lung cancer in the forthcoming eighth edition of the TNM classification for lung cancer. J. Thorac. Oncol. 11, 300–311 (2016).

    PubMed  Google Scholar 

  5. Jemal, A. et al. Cancer Statistics, 2007. CA Cancer J. Clin. 57, 43–66 (2007).

    PubMed  Google Scholar 

  6. Govindan, R. et al. Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: analysis of the Surveillance, Epidemiologic, and End Results database. J. Clin. Oncol. 24, 4539–4544 (2006).

    PubMed  Google Scholar 

  7. American Cancer Society. Cancer Facts & Figures 2016 (American Cancer Society, 2016).

  8. Fitzmaurice, C. et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: a systematic analysis for the global burden of disease study. JAMA Oncol. 3, 524–548 (2017).

    PubMed  Google Scholar 

  9. Pesch, B. et al. Cigarette smoking and lung cancer — relative risk estimates for the major histological types from a pooled analysis of case-control studies. Int. J. Cancer 131, 1210–1219 (2012).

    CAS  PubMed  Google Scholar 

  10. Varghese, A. M. et al. Small-cell lung cancers in patients who never smoked cigarettes. J. Thorac. Oncol. 9, 892–896 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Rudin, C. M. et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat. Genet. 44, 1111–1116 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Brambilla, E., Beasley, M. B., Auerbach, O. & Kuschner, M. in WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart 4th edn (eds Travis, W. D., Brambilla, E., Burke, A. P., Marx, A. & Nicholson, G. A.) 63–68 (IARC, 2015).

    Google Scholar 

  15. Silva, M. et al. Screening with low-dose computed tomography does not improve survival of small cell lung cancer. J. Thorac. Oncol. 11, 187–193 (2016).

    PubMed  Google Scholar 

  16. Pignon, J.-P. et al. A meta-analysis of thoracic radiotherapy for small-cell lung cancer. N. Engl. J. Med. 327, 1618–1624 (1992).

    CAS  PubMed  Google Scholar 

  17. Turrisi, A. T. III et al. Twice-daily compared with once-daily thoracic radiotherapy in limited small-cell lung cancer treated concurrently with cisplatin and etoposide. N. Engl. J. Med. 340, 265–271 (1999).

    CAS  PubMed  Google Scholar 

  18. Turrisi, A. T. III, Glover, D. J. & Mason, B. A. A preliminary report: concurrent twice-daily radiotherapy plus platinum-etoposide chemotherapy for limited small cell lung cancer. Int. J. Radiat. Oncol. Biol. Phys. 15, 183–187 (1988).

    PubMed  Google Scholar 

  19. Faivre-Finn, C. et al. CONVERT: an international randomised trial of concurrent chemo-radiotherapy (cCTRT) comparing twice-daily (BD) and once-daily (OD) radiotherapy schedules in patients with limited stage small cell lung cancer (LS-SCLC) and good performance status (PS) [abstract]. J. Clin. Oncol. 34, 8504 (2016).

    Google Scholar 

  20. National Comprehensive Cancer Network. National Comprehensive Cancer Network Guidelines: Small Cell Lung Cancer Version 2.2017 (NCCN, 2016).

  21. Aupérin, A. et al. Prophylactic cranial irradiation for patients with small-cell lung cancer in complete remission. N. Engl. J. Med. 341, 476–484 (1999).

    PubMed  Google Scholar 

  22. Vallières, E. et al. The IASLC lung cancer staging project: proposals regarding the relevance of TNM in the pathologic staging of small cell lung cancer in the forthcoming (seventh) edition of the TNM classification for lung cancer. J. Thorac. Oncol. 4, 1049–1059 (2009).

    PubMed  Google Scholar 

  23. Gaspar, L. E. et al. Small-cell lung cancer: prognostic factors and changing treatment over 15 years. Clin. Lung Cancer 13, 115–122 (2012).

    PubMed  Google Scholar 

  24. Fukuoka, M. et al. Randomized trial of cyclophosphamide, doxorubicin, and vincristine versus cisplatin and etoposide versus alternation of these regimens in small-cell lung cancer. J. Natl Cancer Inst. 83, 855–861 (1991).

    CAS  PubMed  Google Scholar 

  25. Roth, B. J. et al. Randomized study of cyclophosphamide, doxorubicin, and vincristine versus etoposide and cisplatin versus alternation of these two regimens in extensive small-cell lung cancer: a phase III trial of the Southeastern Cancer Study Group. J. Clin. Oncol. 10, 282–291 (1992).

    CAS  PubMed  Google Scholar 

  26. Niell, H. B. et al. Randomized phase III intergroup trial of etoposide and cisplatin with or without paclitaxel and granulocyte colony-stimulating factor in patients with extensive-stage small-cell lung cancer: Cancer and Leukemia Group B Trial 9732. J. Clin. Oncol. 23, 3752–3759 (2005).

    CAS  PubMed  Google Scholar 

  27. Schmittel, A. et al. A randomized phase II trial of irinotecan plus carboplatin versus etoposide plus carboplatin treatment in patients with extended disease small-cell lung cancer. Ann. Oncol. 17, 663–667 (2006).

    CAS  PubMed  Google Scholar 

  28. Noda, K. et al. Irinotecan plus cisplatin compared with etoposide plus cisplatin for extensive small-cell lung cancer. N. Engl. J. Med. 346, 85–91 (2002).

    CAS  PubMed  Google Scholar 

  29. Hanna, N. et al. Randomized phase III trial comparing irinotecan/cisplatin with etoposide/cisplatin in patients with previously untreated extensive-stage disease small-cell lung cancer. J. Clin. Oncol. 24, 2038–2043 (2006).

    CAS  PubMed  Google Scholar 

  30. Rossi, A. et al. Carboplatin- or cisplatin-based chemotherapy in first-line treatment of small-cell lung cancer: the COCIS meta-analysis of individual patient data. J. Clin. Oncol. 30, 1692–1698 (2012).

    CAS  PubMed  Google Scholar 

  31. Primo, N. L. et al. Phase III trial of irinotecan/cisplatin compared with etoposide/cisplatin in extensive-stage small-cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J. Clin. Oncol. 27, 2530–2535 (2009).

    Google Scholar 

  32. Ardizzoni, A. et al. Topotecan, a new active drug in the second-line treatment of small-cell lung cancer: a phase II study in patients with refractory and sensitive disease. The European Organization for Research and Treatment of Cancer Early Clinical Studies Group and New Drug Development Office, and the Lung Cancer Cooperative Group. J. Clin. Oncol. 15, 2090–2096 (1997).

    CAS  PubMed  Google Scholar 

  33. Slotman, B. J. et al. Use of thoracic radiotherapy for extensive stage small-cell lung cancer: a phase 3 randomised controlled trial. Lancet 385, 36–42 (2015).

    PubMed  Google Scholar 

  34. Slotman, B. J. et al. Radiotherapy for extensive stage small-cell lung cancer — authors' reply. Lancet 385, 1292–1293 (2015).

    PubMed  Google Scholar 

  35. Slotman, B. et al. Prophylactic cranial irradiation in extensive small-cell lung cancer. N. Engl. J. Med. 357, 664–672 (2007).

    CAS  PubMed  Google Scholar 

  36. Takahashi, T. et al. Prophylactic cranial irradiation versus observation in patients with extensive-disease small-cell lung cancer: a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 18, 663–671 (2017).

    PubMed  Google Scholar 

  37. Giuliani, M. et al. Utilization of prophylactic cranial irradiation in patients with limited stage small cell lung carcinoma. Cancer 116, 5694–5699 (2010).

    PubMed  Google Scholar 

  38. Lok, B. H. et al. The factors influencing the utilization of prophylactic cranial irradiation in patients with limited-stage small cell lung cancer [abstract 3048]. Int. J. Radiat. Oncol. Biol. Phys. 93 (Suppl.), E420–E421 (2015).

    Google Scholar 

  39. Borromeo, M. D. et al. ASCL1 and NEUROD1 reveal heterogeneity in pulmonary neuroendocrine tumors and regulate distinct genetic programs. Cell Rep. 16, 1259–1272 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Poirier, J. T. et al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 34, 5869–5878 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Poirier, J. T. et al. Selective tropism of Seneca Valley virus for variant subtype small cell lung cancer. J. Natl Cancer Inst. 105, 1059–1065 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Gazdar, A. F., Carney, D. N., Nau, M. M. & Minna, J. D. Characterization of variant subclasses of cell lines derived from small cell lung cancer having distinctive biochemical, morphological, and growth properties. Cancer Res. 45, 2924–2930 (1985).

    CAS  PubMed  Google Scholar 

  43. Carney, D. N. et al. Establishment and identification of small cell lung cancer cell lines having classic and variant features. Cancer Res. 45, 2913–2923 (1985).

    CAS  PubMed  Google Scholar 

  44. Meuwissen, R. et al. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell 4, 181–189 (2003).

    CAS  PubMed  Google Scholar 

  45. Gazdar, A. F. et al. The comparative pathology of genetically engineered mouse models for neuroendocrine carcinomas of the lung. J. Thorac. Oncol. 10, 553–564 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Ito, T. et al. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development 127, 3913–3921 (2000).

    CAS  PubMed  Google Scholar 

  47. Jiang, T. et al. Achaete-scute complex homologue 1 regulates tumor-initiating capacity in human small cell lung cancer. Cancer Res. 69, 845–854 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Osada, H., Tatematsu, Y., Yatabe, Y., Horio, Y. & Takahashi, T. ASH1 gene is a specific therapeutic target for lung cancers with neuroendocrine features. Cancer Res. 65, 10680–10685 (2005).

    CAS  PubMed  Google Scholar 

  49. Mollaoglu, G. et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to aurora kinase inhibition. Cancer Cell 31, 270–285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Augustyn, A. et al. ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc. Natl Acad. Sci. USA 111, 14788–14793 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Osborne, J. K. et al. NeuroD1 regulates survival and migration of neuroendocrine lung carcinomas via signaling molecules TrkB and NCAM. Proc. Natl Acad. Sci. USA 110, 6524–6529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Neptune, E. R. et al. Targeted disruption of NeuroD, a proneural basic helix-loop-helix factor, impairs distal lung formation and neuroendocrine morphology in the neonatal lung. J. Biol. Chem. 283, 21160–21169 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Girard, L., Zöchbauer-Müller, S., Virmani, A. K., Gazdar, A. F. & Minna, J. D. Genome-wide allelotyping of lung cancer identifies new regions of allelic loss, differences between small cell lung cancer and non-small cell lung cancer, and loci clustering. Cancer Res. 60, 4894–4906 (2000).

    CAS  PubMed  Google Scholar 

  54. Shivapurkar, N. et al. Deletions of chromosome 4 at multiple sites are frequent in malignant mesothelioma and small cell lung carcinoma. Clin. Cancer Res. 5, 17–23 (1999).

    CAS  PubMed  Google Scholar 

  55. Petersen, I. et al. Small-cell lung cancer is characterized by a high incidence of deletions on chromosomes 3p, 4q, 5q, 10q, 13q and 17p. Br. J. Cancer 75, 79–86 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Johnson, B. E. et al. MYC family DNA amplification in small cell lung cancer patients' tumors and corresponding cell lines. Cancer Res. 48, 5163–5166 (1988).

    CAS  PubMed  Google Scholar 

  57. Brambilla, E. et al. Apoptosis-related factors p53, Bcl2, and Bax in neuroendocrine lung tumors. Am. J. Pathol. 149, 1941–1952 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Tamborini, E. et al. Detection of overexpressed and phosphorylated wild-type kit receptor in surgical specimens of small cell lung cancer. Clin. Cancer Res. 10, 8214–8219 (2004).

    CAS  PubMed  Google Scholar 

  59. Carney, D. N., Cuttitta, F., Moody, T. W. & Minna, J. D. Selective stimulation of small cell lung cancer clonal growth by bombesin and gastrin-releasing peptide. Cancer Res. 47, 821–825 (1987).

    CAS  PubMed  Google Scholar 

  60. Pedersen, N. et al. Transcriptional gene expression profiling of small cell lung cancer cells. Cancer Res. 63, 1943–1953 (2003).

    CAS  PubMed  Google Scholar 

  61. Fujino, K. et al. Insulinoma-associated protein 1 is a crucial regulator of neuroendocrine differentiation in lung cancer. Am. J. Pathol. 185, 3164–3177 (2015).

    CAS  PubMed  Google Scholar 

  62. Borges, M. et al. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 386, 852–855 (1997).

    CAS  PubMed  Google Scholar 

  63. Peifer, M. et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 44, 1104–1110 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Dowlati, A. et al. Clinical correlation of extensive-stage small-cell lung cancer genomics. Ann. Oncol. 27, 642–647 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

  66. Baylin, S. B. & Jones, P. A. A decade of exploring the cancer epigenome — biological and translational implications. Nat. Rev. Cancer 11, 726–734 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Jones, P. A. & Baylin, S. B. The fundamental role of epigenetic events in cancer. Nat. Rev. Genet. 3, 415–428 (2002).

    CAS  PubMed  Google Scholar 

  68. Esteller, M. & Herman, J. G. Cancer as an epigenetic disease: DNA methylation and chromatin alterations in human tumours. J. Pathol. 196, 1–7 (2002).

    CAS  PubMed  Google Scholar 

  69. Cedar, H. & Bergman, Y. Linking DNA methylation and histone modification: patterns and paradigms. Nat. Rev. Genet. 10, 295–304 (2009).

    CAS  PubMed  Google Scholar 

  70. Hopkins-Donaldson, S. et al. Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation. Cell Death Differ. 10, 356–364 (2003).

    CAS  PubMed  Google Scholar 

  71. Kaminskyy, V. O., Surova, O. V., Vaculova, A. & Zhivotovsky, B. Combined inhibition of DNA methyltransferase and histone deacetylase restores caspase-8 expression and sensitizes SCLC cells to TRAIL. Carcinogenesis 32, 1450–1458 (2011).

    CAS  PubMed  Google Scholar 

  72. Kalari, S., Jung, M., Kernstine, K. H., Takahashi, T. & Pfeifer, G. P. The DNA methylation landscape of small cell lung cancer suggests a differentiation defect of neuroendocrine cells. Oncogene 32, 3559–3568 (2013).

    CAS  PubMed  Google Scholar 

  73. Wang, Y. et al. Prognostic and predictive value of CpG island methylator phenotype in patients with locally advanced nonmetastatic sporadic colorectal cancer. Gastroenterol. Res. Pract. 2014, 436985 (2014).

    PubMed  PubMed Central  Google Scholar 

  74. Karlsson, A. et al. Genome-wide DNA methylation analysis of lung carcinoma reveals one neuroendocrine and four adenocarcinoma epitypes associated with patient outcome. Clin. Cancer Res. 20, 6127–6140 (2014).

    CAS  PubMed  Google Scholar 

  75. Saito, Y. et al. Prognostic significance of CpG island methylator phenotype in surgically resected small cell lung carcinoma. Cancer Sci. 107, 320–325 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Byers, L. A. et al. Proteomic profiling identifies dysregulated pathways in small cell lung cancer and novel therapeutic targets including PARP1. Cancer Discov. 2, 798–811 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Chambon, P., Weill, J. D. & Mandel, P. Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme. Biochem. Biophys. Res. Commun. 11, 39–43 (1963).

    CAS  PubMed  Google Scholar 

  78. Vyas, S., Chesarone-Cataldo, M., Todorova, T., Huang, Y.-H. & Chang, P. A systematic analysis of the PARP protein family identifies new functions critical for cell physiology. Nat. Commun. 4, 2240 (2013).

    PubMed  Google Scholar 

  79. Rouleau, M., Patel, A., Hendzel, M. J., Kaufmann, S. H. & Poirier, G. G. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer 10, 293–301 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Simbulan-Rosenthal, C. M. et al. PARP-1 binds E2F-1 independently of its DNA binding and catalytic domains, and acts as a novel coactivator of E2F-1-mediated transcription during re-entry of quiescent cells into S phase. Oncogene 22, 8460–8471 (2003).

    CAS  PubMed  Google Scholar 

  81. de Bono, J. et al. Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discov. http://dx.doi.org/10.1158/2159-8290.CD-16-1250 (2017).

  82. Audeh, M. W. et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial. Lancet 376, 245–251 (2010).

    CAS  PubMed  Google Scholar 

  83. Swisher, E. M. et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 18, 75–87 (2017).

    CAS  PubMed  Google Scholar 

  84. Bryant, H. E. et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917 (2005).

    CAS  PubMed  Google Scholar 

  85. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    CAS  PubMed  Google Scholar 

  86. Lok, B. H., Carley, A. C., Tchang, B. & Powell, S. N. RAD52 inactivation is synthetically lethal with deficiencies in BRCA1 and PALB2 in addition to BRCA2 through RAD51-mediated homologous recombination. Oncogene 32, 3552–3558 (2013).

    CAS  PubMed  Google Scholar 

  87. Lok, B. H. & Powell, S. N. Molecular pathways: understanding the role of Rad52 in homologous recombination for therapeutic advancement. Clin. Cancer Res. 18, 6400–6406 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Ceccaldi, R. et al. Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature 518, 258–262 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Mateos-Gomez, P. A. et al. Mammalian polymerase theta promotes alternative NHEJ and suppresses recombination. Nature 518, 254–257 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Watkins, J. A., Irshad, S., Grigoriadis, A. & Tutt, A. N. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 16, 211 (2014).

    PubMed  PubMed Central  Google Scholar 

  91. Mirza, M. R. et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 375, 2154–2164 (2016).

    CAS  PubMed  Google Scholar 

  92. Lok, B. H. et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin. Cancer Res. 23, 523–535 (2017).

    CAS  PubMed  Google Scholar 

  93. Murai, J. et al. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 7, 76534–76550 (2016).

    PubMed  PubMed Central  Google Scholar 

  94. Mu, Y. et al. SLFN11 inhibits checkpoint maintenance and homologous recombination repair. EMBO Rep. 17, 94–109 (2016).

    CAS  PubMed  Google Scholar 

  95. Barretina, J. et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483, 603–607 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Deng, Y. et al. High SLFN11 expression predicts better survival for patients with KRAS exon 2 wild type colorectal cancer after treated with adjuvant oxaliplatin-based treatment. BMC Cancer 15, 833 (2015).

    PubMed  PubMed Central  Google Scholar 

  97. Tang, S. W. et al. SLFN11 is a transcriptional target of EWS-FLI1 and a determinant of drug response in Ewing sarcoma. Clin. Cancer Res. 21, 4184–4193 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Nogales, V. et al. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget 7, 3084–3097 (2016).

    PubMed  Google Scholar 

  99. Byers, L. A. et al. Improved small cell lung cancer response rates with veliparib and temozolomide: results from a phase II trial [abstract MA11.07]. J. Thorac. Oncol. 12, S406–S407 (2016).

    Google Scholar 

  100. Margueron, R. & Reinberg, D. The Polycomb complex PRC2 and its mark in life. Nature 469, 343–349 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Chang, C. J. & Hung, M. C. The role of EZH2 in tumour progression. Br. J. Cancer 106, 243–247 (2012).

    CAS  PubMed  Google Scholar 

  102. Bracken, A. P. et al. EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer. EMBO J. 22, 5323–5335 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Coe, B. P. et al. Genomic deregulation of the E2F/Rb pathway leads to activation of the oncogene EZH2 in small cell lung cancer. PLoS ONE 8, e71670 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Gardner, E. E. et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell 31, 286–299 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Levine, A. J. p53, the cellular gatekeeper for growth and division. Cell 88, 323–331 (1997).

    CAS  PubMed  Google Scholar 

  106. Levine, A. J., Momand, J. & Finlay, C. A. The p53 tumour suppressor gene. Nature 351, 453–456 (1991).

    CAS  PubMed  Google Scholar 

  107. Dobbelstein, M. & Sorensen, C. S. Exploiting replicative stress to treat cancer. Nat. Rev. Drug Discov. 14, 405–423 (2015).

    CAS  PubMed  Google Scholar 

  108. Parker, L. L. & Piwnica-Worms, H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science 257, 1955–1957 (1992).

    CAS  PubMed  Google Scholar 

  109. Hirai, H. et al. Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents. Mol. Cancer Ther. 8, 2992–3000 (2009).

    CAS  PubMed  Google Scholar 

  110. Hirai, H. et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol. Ther. 9, 514–522 (2010).

    CAS  PubMed  Google Scholar 

  111. Rajeshkumar, N. V. et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin. Cancer Res. 17, 2799–2806 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Matheson, C. J., Backos, D. S. & Reigan, P. Targeting WEE1 kinase in cancer. Trends Pharmacol. Sci. 37, 872–881 (2016).

    CAS  PubMed  Google Scholar 

  113. Do, K. et al. Phase I study of single-agent AZD1775 (MK-1775), a Wee1 kinase inhibitor, in patients with refractory solid tumors. J. Clin. Oncol. 33, 3409–3415 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Guertin, A. D. et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol. Cancer Ther. 12, 1442–1452 (2013).

    CAS  PubMed  Google Scholar 

  115. Van Linden, A. A. et al. Inhibition of Wee1 sensitizes cancer cells to antimetabolite chemotherapeutics in vitro and in vivo, independent of p53 functionality. Mol. Cancer Ther. 12, 2675–2684 (2013).

    CAS  PubMed  Google Scholar 

  116. Ladi, E. et al. The divergent DSL ligand Dll3 does not activate Notch signaling but cell autonomously attenuates signaling induced by other DSL ligands. J. Cell Biol. 170, 983–992 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Loomes, K. M. et al. Dll3 and Notch1 genetic interactions model axial segmental and craniofacial malformations of human birth defects. Dev. Dyn. 236, 2943–2951 (2007).

    CAS  PubMed  Google Scholar 

  118. Morimoto, M., Nishinakamura, R., Saga, Y. & Kopan, R. Different assemblies of Notch receptors coordinate the distribution of the major bronchial Clara, ciliated and neuroendocrine cells. Development 139, 4365–4373 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Kunnimalaiyaan, M. & Chen, H. Tumor suppressor role of Notch-1 signaling in neuroendocrine tumors. Oncologist 12, 535–542 (2007).

    CAS  PubMed  Google Scholar 

  120. Chapman, G., Sparrow, D. B., Kremmer, E. & Dunwoodie, S. L. Notch inhibition by the ligand DELTA-LIKE 3 defines the mechanism of abnormal vertebral segmentation in spondylocostal dysostosis. Hum. Mol. Genet. 20, 905–916 (2011).

    CAS  PubMed  Google Scholar 

  121. Saunders, L. R. et al. A DLL3-targeted antibody–drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl Med. 7, 302ra136 (2015).

    PubMed  PubMed Central  Google Scholar 

  122. Antonow, D. & Thurston, D. E. Synthesis of DNA-interactive pyrrolo[2,1–c][1,4]benzodiazepines (PBDs). Chem. Rev. 111, 2815–2864 (2011).

    CAS  PubMed  Google Scholar 

  123. Gerratana, B. Biosynthesis, synthesis and biological activities of pyrrolobenzodiazepines. Med. Res. Rev. 32, 254–293 (2012).

    CAS  PubMed  Google Scholar 

  124. Rudin, C. M. et al. Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: a first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol. 18, 42–51 (2017).

    CAS  PubMed  Google Scholar 

  125. Nikonova, A. S., Astsaturov, I., Serebriiskii, I. G., Dunbrack, R. L. & Golemis, E. A. Aurora A kinase (AURKA) in normal and pathological cell division. Cell. Mol. Life Sci. 70, 661–687 (2013).

    CAS  PubMed  Google Scholar 

  126. Lu, Y. et al. Knocking down the expression of Aurora A gene inhibited cell proliferation and induced G2/M phase arrest in human small cell lung cancer cells. Oncol. Rep. 32, 243–249 (2014).

    CAS  PubMed  Google Scholar 

  127. Manfredi, M. G. et al. Characterization of alisertib (MLN8237), an investigational small-molecule inhibitor of aurora A kinase using novel in vivo pharmacodynamic assays. Clin. Cancer Res. 17, 7614–7624 (2011).

    CAS  PubMed  Google Scholar 

  128. Melichar, B. et al. Safety and activity of alisertib, an investigational aurora kinase A inhibitor, in patients with breast cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma: a five-arm phase 2 study. Lancet Oncol. 16, 395–405 (2015).

    CAS  PubMed  Google Scholar 

  129. Owonikoko, T. et al. Randomized phase 2 study: alisertib (MLN8237) or placebo + paclitaxel as second-line therapy for small-cell lung cancer (SCLC) [abstract OA05.05]. J. Thorac. Oncol. 12, S261–S262 (2017).

    Google Scholar 

  130. Schreiber, R. D., Old, L. J. & Smyth, M. J. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 331, 1565–1570 (2011).

    CAS  PubMed  Google Scholar 

  131. Khalil, D. N., Smith, E. L., Brentjens, R. J. & Wolchok, J. D. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat. Rev. Clin. Oncol. 13, 273–290 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    CAS  PubMed  Google Scholar 

  133. Hoos, A. et al. Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy. Semin. Oncol. 37, 533–546 (2010).

    CAS  PubMed  Google Scholar 

  134. Reck, M. et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann. Oncol. 24, 75–83 (2012).

    PubMed  Google Scholar 

  135. Wolchok, J. D. et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin. Cancer Res. 15, 7412–7420 (2009).

    CAS  PubMed  Google Scholar 

  136. Reck, M. et al. Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer. J. Clin. Oncol. 34, 3740–3748 (2016).

    CAS  PubMed  Google Scholar 

  137. Curran, M. A., Montalvo, W., Yagita, H. & Allison, J. P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl Acad. Sci. USA 107, 4275–4280 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Antonia, S. J. et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 17, 883–895 (2016).

    CAS  PubMed  Google Scholar 

  139. Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Yu, H. et al. PD-L1 expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J. Thorac. Oncol. 12, 110–120 (2017).

    PubMed  Google Scholar 

  141. Weiskopf, K. & Weissman, I. L. Macrophages are critical effectors of antibody therapies for cancer. MAbs 7, 303–310 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Matozaki, T., Murata, Y., Okazawa, H. & Ohnishi, H. Functions and molecular mechanisms of the CD47–SIRPα signalling pathway. Trends Cell Biol. 19, 72–80 (2009).

    CAS  PubMed  Google Scholar 

  143. Weiskopf, K. et al. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer. J. Clin. Invest. 126, 2610–2620 (2016).

    PubMed  PubMed Central  Google Scholar 

  144. Liu, X. et al. CD47 blockade triggers T cell-mediated destruction of immunogenic tumors. Nat. Med. 21, 1209–1215 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work of the authors is supported by funding from the National Cancer Institute (grants T32 CA009207 to J.K.S., and P30 CA008748 and R01 CA197936 to C.M.R.), and from the Conquer Cancer Foundation of ASCO, the Lung Cancer Research Foundation, and the Radiological Society of North America (to B.H.L.).

Author information

Authors and Affiliations

Authors

Contributions

J.K.S. and B.H.L. made equal contributions to the manuscript and should be considered co-first authors. J.K.S., B.H.L., J.H.L., and J.T.P. researched data for article. All authors made substantial contributions to discussion of content, writing of the Review, and review/editing of the manuscript.

Corresponding authors

Correspondence to John T. Poirier or Charles M. Rudin.

Ethics declarations

Competing interests

C.M.R. has been a paid consultant regarding oncology drug development for Bristol-Myers Squibb, Celgene, G1 Therapeutics, Harpoon Therapeutics, Medivation, and Novartis. The other authors declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sabari, J., Lok, B., Laird, J. et al. Unravelling the biology of SCLC: implications for therapy. Nat Rev Clin Oncol 14, 549–561 (2017). https://doi.org/10.1038/nrclinonc.2017.71

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrclinonc.2017.71

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