Skip to main content

Advertisement

Log in

Notch pathway in small-cell lung cancer: from preclinical evidence to therapeutic challenges

  • Review
  • Published:
Cellular Oncology Aims and scope Submit manuscript

Abstract

Background

Small-cell lung cancer (SCLC) is an aggressive disease with still limited therapeutic options. Despite being both a chemo- and radiation-sensitive malignancy, SCLC recurrence occurs in most cases and negatively impacts patients’ prognosis. Over the last few years, a deeper understanding of SCLC molecular aberrations has led to the identification of Notch pathway deregulation as a crucial event in SCLC tumorigenesis, disease progression and chemoresistance. In particular, the delta-like protein 3 (DLL3), a Notch inhibitory ligand whose expression is directly related to the key neuroendocrine transcription factor ASCL1, was found to be expressed in ~85% of SCLCs, while it exhibits minimal to absent surface expression in normal lungs. DLL3 thus represents an appealing novel biomarker as well as a potential target in SCLC.

Conclusions

The first DLL3-targeted antibody-drug conjugate rovalpituzumab tesirine (Rova-T, SC16LD6.5) has shown promising results in terms of efficacy and safety for the management of extensive SCLC, supporting further studies on this novel therapeutic approach that combines specific SCLC targeting with the cell-killing ability of a pyrrolobenzodiazepine dimer. In the present review, we discuss currently available evidence on the biological role of Notch signaling in SCLC from early preclinical findings to current and future clinical implications.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. M. Reck, K.F. Rabe, Advanced non-small-cell lung cancer. N. Eng. J. Med. 377, 849 (2017)

  2. J.K. Sabari, B.H. Lok, J.H. Laird, J.T. Poirier, C.M. Rudin, Unravelling the biology of SCLC: implications for therapy. Nat. Rev. Clin. Oncol. 14, 549–561 (2017)

  3. A.G. Nicholson, K. Chansky, J. Crowley, R. Beyruti, K. Kubota, A. Turrisi, W.E.E. Eberhardt, J. van Meerbeeck, R. Rami-Porta, P. Goldstraw, R. Rami-Porta, H. Asamura, D. Ball, D.G. Beer, R. Beyruti, V. Bolejack, K. Chansky, J. Crowley, F. Detterbeck, W.E.E. Eberhardt, J. Edwards, F. Galateau-Sallé, D. Giroux, F. Gleeson, P. Groome, J. Huang, C. Kennedy, J. Kim, Y.T. Kim, L. Kingsbury, H. Kondo, M. Krasnik, K. Kubota, T. Lerut, G. Lyons, M. Marino, E.M. Marom, J. van Meerbeeck, A. Mitchell, T. Nakano, A.G. Nicholson, A. Nowak, M. Peake, T. Rice, K. Rosenzweig, E. Ruffini, V. Rusch, N. Saijo, P. Van Schil, J.-P. Sculier, L. Shemanski, K. Stratton, K. Suzuki, Y. Tachimori, C.F. Thomas, W. Travis, M.S. Tsao, A. Turrisi, J. Vansteenkiste, H. Watanabe, Y.-L. Wu, P. Baas, J. Erasmus, S. Hasegawa, K. Inai, K. Kernstine, H. Kindler, L. Krug, K. Nackaerts, H. Pass, D. Rice, C. Falkson, P.L. Filosso, G. Giaccone, K. Kondo, M. Lucchi, M. Okumura, E. Blackstone, F.A. Cavaco, E.A. Barrera, J.A. Arca, I.P. Lamelas, A.A. Obrer, R.G. Jorge, D. Ball, G.K. Bascom, A.I.B. Orozco, M.A.G. Castro, M.G. Blum, D. Chimondeguy, V. Cvijanovic, S. Defranchi, B. de Olaiz Navarro, I.E. Campuzano, I.M. Vidueira, E.F. Araujo, F.A. García, K.M. Fong, G.F. Corral, S.C. González, J.F. Gilart, L.G. Arangüena, S.G. Barajas, P. Girard, T. Goksel, M.T.G. Budiño, G.G. Casaurrán, J.A.G. Blanco, J.H. Hernández, H.H. Rodríguez, J.H. Collantes, M.I. Heras, J.M.I. Elena, E. Jakobsen, S. Kostas, P.L. Atance, A.N. Ares, M. Liao, M. Losanovscky, G. Lyons, R. Magaroles, L.D.E. Júlvez, M.M. Gorospe, B. McCaughan, C. Kennedy, R.M. Íñiguez, L.M. Sorribes, S.N. Gozalo, C.Á. de Arriba, M.N. Delgado, J.P. Alarcón, J.C.P. Cuesta, J.S. Park, H. Pass, M.J.P. Fernández, M. Rosenberg, V. Rusch, J.S. de Cos Escuín, A.S. Vinuesa, M.S. Mitjans, T.E. Strand, D. Subotic, S. Swisher, R. Terra, C. Thomas, K. Tournoy, P. Van Schil, M. Velasquez, Y.L. Wu, K. Yokoi, J. 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. Thorac. Oncol. 11, 300 (2016)

  4. B. Oronsky, T.R. Reid, A. Oronsky, C.A. Carter, What's new in SCLC? A review. Neoplasia 19, 842 (2017)

  5. S.J. Antonia, J.A. López-Martin, J. Bendell, P.A. Ott, M. Taylor, J.P. Eder, D. Jäger, M.C. Pietanza, D.T. Le, F. de Braud, M.A. Morse, P.A. Ascierto, L. Horn, A. Amin, R.N. Pillai, J. Evans, I. Chau, P. Bono, A. Atmaca, P. Sharma, C.T. Harbison, C.S. Lin, O. Christensen, E. Calvo, 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)

    Article  CAS  PubMed  Google Scholar 

  6. M.D. Hellmann, M.K. Callahan, M.M. Awad, E. Calvo, P.A. Ascierto, A. Atmaca, N.A. Rizvi, F.R. Hirsch, G. Selvaggi, J.D. Szustakowski, A. Sasson, R. Golhar, P. Vitazka, H. Chang, W.J. Geese, S.J. Antonia, Tumor mutational burden and efficacy of Nivolumab monotherapy and in combination with Ipilimumab in small-cell lung cancer. Cancer Cell 33, 853–861.e4 (2018)

  7. C.M. Rudin, M.C. Pietanza, T.M. Bauer, N. Ready, D. Morgensztern, B.S. Glisson, L.A. Byers, M.L. Johnson, H.A. Burris, F. Robert, T.H. Han, S. Bheddah, N. Theiss, S. Watson, D. Mathur, B. Vennapusa, H. Zayed, S. Lally, D.K. Strickland, R. Govindan, S.J. Dylla, S.L. Peng, D.R. Spigel, SCRX16–001 investigators, 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)

  8. D.P. Carbone, D. Morgensztern, S. Le Moulec, R. Santana-Davila, N. Ready, C.L. Hann, B.S. Glisson, A. Dowlati, C.M. Rudin, S. Lally, S. Yalamanchili, J. Wolf, R. Govindan, B. Besse, Efficacy and safety of rovalpituzumab tesirine in patients With DLL3-expressing, ≥ 3rdline small cell lung cancer: Results from the phase 2 TRINITY study. J. Clin. Oncol. 36, 8507 (2018)

  9. R.J. Fleming, Structural conservation of Notch receptors and ligands. Semin. Cell Dev. Biol. 9, 599–607 (1998)

  10. L. Meder, R. Büttner, M. Odenthal, Notch signaling triggers the tumor heterogeneity of small cell lung cancer. J. Thorac. Dis. 9, 4884–4888 (2017)

    Article  PubMed  PubMed Central  Google Scholar 

  11. E. Ladi, J.T. Nichols, W. Ge, A. Miyamoto, C. Yao, L.T. Yang, J. Boulter, Y.E. Sun, C. Kintner, G. Weinmaster, 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)

  12. G. Chapman, D.B. Sparrow, E. Kremmer, S.L. Dunwoodie, 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)

  13. J.S. Crabtree, C.S. Singleton, L. Miele, Notch signaling in neuroendocrine tumors. Front. Oncol. 6, 94 (2016)

  14. J.C. Aster, W.S. Pear, S.C. Blacklow, The varied roles of Notch in cancer. Annu. Rev. Pathol. Mech. Dis 12, 245–275 (2017)

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

  16. M. Ishibashi, S.L. Ang, K. Shiota, S. Nakanishi, R. Kageyama, F. Guillemot, Targeted disruption of mammalian hairy and Enhancer of split homolog-1 (HES-1) leads to up-regulation of neural helix-loop-helix factors, premature neurogenesis, and severe neural tube defects. Genes Dev. 9, 3136–3148 (1995)

  17. M. Borges, R.I. Linnoila, H.J.K. van de Velde, H. Chen, B.D. Nelkin, M. Mabry, S.B. Baylin, D.W. Ball, An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 386, 852–855 (1997)

    Article  CAS  PubMed  Google Scholar 

  18. H. Osada, S. Tomida, Y. Yatabe, Y. Tatematsu, T. Takeuchi, H. Murakami, Y. Kondo, Y. Sekido, T. Takahashi, Roles of Achaete-Scute Homologue 1 in DKK1 and E-cadherin repression and neuroendocrine differentiation in lung cancer. Cancer Res. 68, 1647–1655 (2008)

  19. K.-S. Park, M.-C. Liang, D.M. Raiser, R. Zamponi, R.R. Roach, S.J. Curtis, Z. Walton, B.E. Schaffer, C.M. Roake, A.-F. Zmoos, C. Kriegel, K.-K. Wong, J. Sage, C.F. Kim, Characterization of the cell of origin for small cell lung cancer. Cell Cycle 10, 2806–2815 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. M. Brzozowa-Zasada, A. Piecuch, A. Dittfeld, Ł. Mielańczyk, M. Michalski, G. Wyrobiec, M. Harabin-Słowińska, J. Kurek, R. Wojnicz, Notch signalling pathway as an oncogenic factor involved in cancer development. Contemp. Oncol. 20, 267 (2016)

  21. M. Brzozowa-Zasada, A. Piecuch, M. Michalski, O. Segiet, J. Kurek, M. Harabin-Słowińska, R. Wojnicz, Notch and its oncogenic activity in human malignancies. Eur. Surg. 49, 199–209 (2017)

  22. J. George, J.S. Lim, S.J. Jang, Y. Cun, L. Ozretia, G. Kong, F. Leenders, X. Lu, L. Fernández-Cuesta, G. Bosco, C. Müller, I. Dahmen, N.S. Jahchan, K.-S.S. Park, D. Yang, A.N. Karnezis, D. Vaka, A. Torres, M.S. Wang, J.O. Korbel, R. Menon, S.-M.M. Chun, D. Kim, M. Wilkerson, N. Hayes, D. Engelmann, B. Pützer, M. Bos, S. Michels, I. Vlasic, D. Seidel, B. Pinther, P. Schaub, C. Becker, J. Altmüller, J. Yokota, T. Kohno, R. Iwakawa, K. Tsuta, M. Noguchi, T. Muley, H. Hoffmann, P.A. Schnabel, I. Petersen, Y. Chen, A. Soltermann, V. Tischler, C.M. Choi, Y.-H.H. Kim, P.P. Massion, Y. Zou, D. Jovanovic, M. Kontic, G.M. Wright, P.A. Russell, B. Solomon, I. Koch, M. Lindner, L.A. Muscarella, A. la Torre, J.K. Field, M. Jakopovic, J. Knezevic, E. Castaños-Vélez, L. Roz, U. Pastorino, O.-T.T. Brustugun, M. Lund-Iversen, E. Thunnissen, J. Köhler, M. Schuler, J. Botling, M. Sandelin, M. Sanchez-Cespedes, H.B. Salvesen, V. Achter, U. Lang, M. Bogus, P.M. Schneider, T. Zander, S. Ansén, M. Hallek, J. Wolf, M. Vingron, Y. Yatabe, W.D. Travis, P. Nürnberg, C. Reinhardt, S. Perner, L. Heukamp, R. Büttner, S.A. Haas, E. Brambilla, M. Peifer, J. Sage, R.K. Thomas, L. Ozretić, G. Kong, F. Leenders, X. Lu, L. Fernández-Cuesta, G. Bosco, C. Müller, I. Dahmen, N.S. Jahchan, K.-S.S. Park, D. Yang, A.N. Karnezis, D. Vaka, A. Torres, M.S. Wang, J.O. Korbel, R. Menon, S.-M.M. Chun, D. Kim, M. Wilkerson, N. Hayes, D. Engelmann, B. Pützer, M. Bos, S. Michels, I. Vlasic, D. Seidel, B. Pinther, P. Schaub, C. Becker, J. Altmüller, J. Yokota, T. Kohno, R. Iwakawa, K. Tsuta, M. Noguchi, T. Muley, H. Hoffmann, P.A. Schnabel, I. Petersen, Y. Chen, A. Soltermann, V. Tischler, C.M. Choi, Y.-H.H. Kim, P.P. Massion, Y. Zou, D. Jovanovic, M. Kontic, G.M. Wright, P.A. Russell, B. Solomon, I. Koch, M. Lindner, L.A. Muscarella, A. la Torre, J.K. Field, M. Jakopovic, J. Knezevic, E. Castaños-Vélez, L. Roz, U. Pastorino, O.-T.T. Brustugun, M. Lund-Iversen, E. Thunnissen, J. Köhler, M. Schuler, J. Botling, M. Sandelin, M. Sanchez-Cespedes, H.B. Salvesen, V. Achter, U. Lang, M. Bogus, P.M. Schneider, T. Zander, S. Ansén, M. Hallek, J. Wolf, M. Vingron, Y. Yatabe, W.D. Travis, P. Nürnberg, C. Reinhardt, S. Perner, L. Heukamp, R. Büttner, S.A. Haas, E. Brambilla, M. Peifer, J. Sage, R.K. Thomas, Comprehensive genomic profiles of small cell lung cancer. Nature 524, 47 (2015)

  23. F. Ardeshir-Larijani, G. Wildey, P. Fu, P. Bhateja, A. Dowlati, Frequency of NOTCH pathway mutation in primary tumor of SCLC compared to metastatic biopsies and association with better survival. J. Clin. Oncol. 36, e20574 (2018)

  24. J. Nong, Y. Gong, Y. Guan, X. Yi, Y. Yi, L. Chang, L. Yang, J. Lv, Z. Guo, H. Jia, Y. Chu, T. Liu, M. Chen, L. Byers, E. Roarty, V.K. Lam, V.A. Papadimitrakopoulou, I. Wistuba, J.V. Heymach, B. Glisson, Z. Liao, J.J. Lee, P.A. Futreal, S. Zhang, X. Xia, J. Zhang, J. Wang, Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer. Nat. Commun. 9, 3114 (2018)

  25. V. Sriuranpong, M.W. Borges, R.K. Ravi, D.R. Arnold, B.D. Nelkin, S.B. Baylin, D.W. Ball, Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res 61, 3200–3205 (2001)

  26. H. Wael, R. Yoshida, S. Kudoh, K. Hasegawa, K. Niimori-Kita, T. Ito, Notch1 signaling controls cell proliferation, apoptosis and differentiation in lung carcinoma. Lung Cancer 85, 131–140 (2014)

    Article  PubMed  Google Scholar 

  27. W.A. Hassan, R. Yoshida, S. Kudoh, K. Hasegawa, K. Niimori-Kita, T. Ito, Notch1 controls cell invasion and metastasis in small cell lung carcinoma cell lines. Lung Cancer 86, 304–310 (2014)

    Article  PubMed  Google Scholar 

  28. W.A. Hassan, R. Yoshida, S. Kudoh, Y. Motooka, T. Ito, Evaluation of role of Notch3 signaling pathway in human lung cancer cells. J. Cancer Res. Clin. Oncol. 142, 981–993 (2016)

  29. L. Meder, K. König, L. Ozretic, A.M. Schultheis, F. Ueckeroth, C.P. Ade, K. Albus, D. Boehm, U. Rommerscheidt-Fuss, A. Florin, T. Buhl, W. Hartmann, J. Wolf, S. Merkelbach-Bruse, M. Eilers, S. Perner, L.C. Heukamp, R. Buettner, NOTCH, ASCL1, p53 and RB alterations define an alternative pathway driving neuroendocrine and small cell lung carcinomas. Int. J. Cancer 138, 927–938 (2016)

  30. T. Jiang, B.J. Collins, N. Jin, D.N. Watkins, M.V. Brock, W. Matsui, B.D. Nelkin, D.W. Ball, Achaete-Scute Complex Homologue 1 regulates tumor-initiating capacity in human small cell lung cancer. Cancer Res. 69, 845–854 (2009)

  31. J.S. Lim, A. Ibaseta, M.M. Fischer, B. Cancilla, G. O’Young, S. Cristea, V.C. Luca, N.S.J. Di Yang, C. Hamard, M. Antoine, M. Wislez, C. Kong, J. Cain, Y.W. Liu, A.M. Kapoun, K.C. Garcia, T. Hoey, C.L. Murriel, J. Sage, Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 545, 360-364 (2017)

  32. W.A. Hassan, S. Takebayashi, M.O.A. Abdalla, K. Fujino, S. Kudoh, Y. Motooka, Y. Sato, Y. Naito, K. Higaki, J. Wakimoto, S. Okada, M. Nakao, Y. Ishikawa, T. Ito, Correlation between histone acetylation and expression of Notch1 in human lung carcinoma and its possible role in combined small-cell lung carcinoma. Lab. Invest. 97, 913–921 (2017)

  33. W. Zhang, L. Girard, Y.-A. Zhang, T. Haruki, M. Papari-Zareei, V. Stastny, H.K. Ghayee, K. Pacak, T.G. Oliver, J.D. Minna, A.F. Gazdar, Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl. Lung Cancer Res. 7, 32–49 (2018)

  34. W.A. Hassan, R. Yoshida, S. Kudoh, H. Kameyama, K. Hasegawa, K. Niimori-Kita, T. Ito, Notch1 controls cell chemoresistance in small cell lung carcinoma cells. Thorac. Cancer 7, 123–128 (2016)

    Article  CAS  PubMed  Google Scholar 

  35. J. Calbó, R. Meuwissen, E. Van Montfort, O. Van Tellingen, A. Berns, Genotype-phenotype relationships in a mouse model for human small-cell lung cancer. Cold Spring Harb. Symp. Quant. Biol. 70, 225–232 (2005)

  36. T. Kuramoto, H. Goto, A. Mitsuhashi, S. Tabata, H. Ogawa, H. Uehara, A. Saijo, S. Kakiuchi, Y. Maekawa, K. Yasutomo, M. Hanibuchi, S. Akiyama, S. Sone, Y. Nishioka, Dll4-Fc, an inhibitor of Dll4-notch signaling, suppresses liver metastasis of small cell lung cancer cells through the downregulation of the NF-κB activity. Mol. Cancer Ther. 11, 2578–2587 (2012)

  37. C. Massard, A. Azaro, J.-C. Soria, U. Lassen, C. Le Tourneau, D. Sarker, C. Smith, U. Ohnmacht, G. Oakley, B.K.R. Patel, E.S.M. Yuen, K.A. Benhadji, J. Rodon, First-in-human study of LY3039478, an oral Notch signaling inhibitor in advanced or metastatic cancer. Ann. Oncol. 29, 1911–1917 (2018)

  38. L. Sun, Q. He, C. Tsai, J. Lei, J. Chen, L. Vienna Makcey, D.H. Coy, HDAC inhibitors suppressed small cell lung cancer cell growth and enhanced the suppressive effects of receptor-targeting cytotoxins via upregulating somatostatin receptor II. Am. J. Transl. Res. 10, 545–553 (2018)

  39. M.C. Pietanza, A.I. Spira, R.M. Jotte, S.M. Gadgeel, A.C. Mita, L.L. Hart, W.L. Gluck, A.C. Chiang, S.V. Liu, A.M. Kapoun, L. Xu, D. Hill, J. Dupont, D.R. Spigel, L. Zhou, J. Clin. Oncol. 33, 7508 (2015)

  40. OncoMed's Phase 2 Trial of Tarextumab in Small Cell Lung Cancer Does Not Meet Endpoints (2017). https://globenewswire.com/news-release/2017/04/17/961251/0/en/OncoMed-s-Phase-2-Trial-of-Tarextumab-in-Small-Cell-Lung-Cancer-Does-Not-Meet-Endpoints.html. Accessed 14 Feb 2019

  41. H. Kikuchi, J. Sakakibara-Konishi, M. Furuta, H. Yokouchi, H. Nishihara, S. Yamazaki, H. Uramoto, F. Tanaka, M. Harada, K. Akie, F. Sugaya, Y. Fujita, K. Takamura, T. Kojima, T. Harada, M. Higuchi, O. Honjo, Y. Minami, N. Watanabe, S. Oizumi, H. Suzuki, T. Ishida, H. Dosaka-Akita, H. Isobe, M. Munakata, M. Nishimura, Expression of Notch1 and Numb in small cell lung cancer. Oncotarget 8, 10348–10358 (2017)

  42. L.R. Saunders, A.J. Bankovich, W.C. Anderson, M.A. Aujay, S. Bheddah, K. Black, R. Desai, P.A. Escarpe, J. Hampl, A. Laysang, D. Liu, J. Lopez-Molina, M. Milton, A. Park, M.A. Pysz, H. Shao, B. Slingerland, M. Torgov, S.A. Williams, O. Foord, P. Howard, J. Jassem, A. Badzio, P. Czapiewski, D.H. Harpole, A. Dowlati, P.P. Massion, W.D. Travis, M.C. Pietanza, J.T. Poirier, C.M. Rudin, R.A. Stull, S.J. Dylla, A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl. Med. 7, 302ra136 (2015)

  43. I. Geffers, K. Serth, G. Chapman, R. Jaekel, K. Schuster-Gossler, R. Cordes, D.B. Sparrow, E. Kremmer, S.L. Dunwoodie, T. Klein, A. Gossler, Divergent functions and distinct localization of the Notch ligands DLL1 and DLL3 in vivo. J. Cell Biol. 178, 465–476 (2007)

  44. M. Saito, K. Saito, K. Shiraishi, D. Maeda, H. Suzuki, Y. Minamiya, K. Kono, T. Kohno, A. Goto, Identification of candidate responders for anti-PD-L1/PD-1 immunotherapy, Rova-T therapy, or EZH2 inhibitory therapy in small-cell lung cancer. Mol. Clin. Oncol. 8, 310–314 (2017)

  45. Y. Liu, L. Yan, J. He, D. Luo, JCES 01.17 The correlation of DLL3 expression with high-grade pulmonary neuroendocrine carcinoma clinicopathologic features and prognose. J. Thorac. Oncol. 12, S1736 (2017)

  46. K. Tanaka, K. Isse, T. Fujihira, M. Takenoyama, L. Saunders, S. Bheddah, Y. Nakanishi, I. Okamoto, Prevalence of Delta-like protein 3 expression in patients with small cell lung cancer. Lung Cancer 115, 116–120 (2018)

    Article  PubMed  Google Scholar 

  47. D. Simos, G. Sajjady, M. Sergi, M.S. Liew, R. Califano, C. Ho, N. Leighl, S. White, Y. Summers, W. Petrcich, P. Wheatley-Price, Third-line chemotherapy in small-cell lung cancer: An international analysis. Clin. Lung Cancer 15, 110–118 (2014)

  48. Study Comparing Rovalpituzumab Tesirine Versus Topotecan in Subjects With Advanced or Metastatic Small Cell Lung Cancer With High Levels of Delta-like Protein 3 (DLL3) and Who Have First Disease Progression During or Following Front-line Platinum-based Chemotherapy (TAHOE). https://clinicaltrials.gov/ct2/show/NCT03061812. Accessed 14 Feb 2019

  49. S.K. Sharma, J. Pourat, D. Abdel-Atti, S.D. Carlin, A. Piersigilli, A.J. Bankovich, E.E. Gardner, O. Hamdy, K. Isse, S. Bheddah, J. Sandoval, K.M. Cunanan, E.B. Johansen, V. Allaj, V. Sisodiya, D. Liu, B.M. Zeglis, C.M. Rudin, S.J. Dylla, J.T. Poirier, J.S. Lewis, Noninvasive interrogation of DLL3 expression in metastatic small cell lung cancer. Cancer Res, 77, 3931–3941 (2017)

  50. R.J. Cardnell, L. Li, T. Sen, R. Bara, P. Tong, J. Fujimoto, A.S. Ireland, M.R. Guthrie, S. Bheddah, U. Banerjee, N.N. Kalu, Y.-H. Fan, S.J. Dylla, F.M. Johnson, I.I. Wistuba, T.G. Oliver, J.V. Heymach, B.S. Glisson, J. Wang, L.A. Byers, Protein expression of TTF1 and cMYC define distinct molecular subgroups of small cell lung cancer with unique vulnerabilities to aurora kinase inhibition, DLL3 targeting, and other targeted therapies. Oncotarget 8, 73419–73432 (2017)

  51. Phase 3 Trial of Rova-T as Second-line Therapy for Advanced Small-Cell Lung Cancer (TAHOE Study) Halted. https://news.abbvie.com/news/phase-3-trial-rova-t-as-second-line-therapy-for-advanced-small-cell-lung-cancer-tahoe-study-halted.htm. Accessed 14 February 2019

  52. A Study of Rovalpituzumab Tesirine as Maintenance Therapy Following First- Line Platinum-Based Chemotherapy in Participants With Extensive Stage Small Cell Lung Cancer (MERU). https://clinicaltrials.gov/ct2/show/NCT03033511. Accessed 14 Feb 2019

  53. A Study of Rovalpituzumab Tesirine Administered in Combination With Nivolumab and With or Without Ipilimumab for Adults With Extensive-Stage Small Cell Lung Cancer. https://clinicaltrials.gov/ct2/show/NCT03026166. Accessed 14 Feb 2019

  54. Study Evaluating the Safety of Rovalpituzumab Tesirine for Third-Line and Later Treatment of Subjects With Relapsed or Refractory Small Cell Lung Cancer. https://clinicaltrials.gov/ct2/show/NCT03334487. Accessed 14 Feb 2019

  55. Rovalpituzumab Tesirine in Delta-Like Protein 3-Expressing Advanced Solid Tumors. https://clinicaltrials.gov/ct2/show/NCT02709889. Accessed 14 Feb 2019

  56. A Study of Rovalpituzumab Tesirine (SC16LD6.5) in the Frontline Treatment of Patients With Delta-Like Protein 3-Expressing Extensive Stage Small Cell Lung Cancer. https://clinicaltrials.gov/ct2/show/NCT02819999. Accessed 14 Feb 2019

  57. A. Rossi, M.C. Garassino, M. Cinquini, P. Sburlati, M. Di Maio, G. Farina, C. Gridelli, V. Torri, Maintenance or consolidation therapy in small-cell lung cancer: A systematic review and meta-analysis. Lung Cancer 70, 119–128 (2010)

    Article  PubMed  Google Scholar 

  58. G. Roviello, L. Zanotti, M.R. Cappelletti, A. Gobbi, C. Senti, A. Bottini, D. Generali, No advantage in survival with targeted therapies as maintenance in patients with limited and extensive-stage small cell lung ccancer: A literature-based meta-analysis of randomized trials. Clin. Lung Cancer 17, 334–340 (2016)

  59. Bristol-Myers Squibb Announces CheckMate −451 Study Did Not Meet Primary Endpoint of Overall Survival with Opdivo Plus Yervoy Vs. Placebo as A Maintenance Therapy in Patients with Extensive-Stage Small Cell Lung Cancer After Completion of First-Line. https://news.bms.com/press-release/corporatefinancial-news/bristol-myers-squibb-announces-checkmate-451-study-did-not-mee. Accessed 14 Feb 2019

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alessandro Leonetti.

Ethics declarations

Conflicts of interests

Tiseo M: Advisory board for Abbvie. Other contributing authors have no conflicts of interest, including specific financial interests or relationship and affiliations relevant to the subject matter or materials discussed in the manuscript.

Additional information

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

Leonetti, A., Facchinetti, F., Minari, R. et al. Notch pathway in small-cell lung cancer: from preclinical evidence to therapeutic challenges. Cell Oncol. 42, 261–273 (2019). https://doi.org/10.1007/s13402-019-00441-3

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13402-019-00441-3

Keywords

Navigation