Progress in the knowledge on the transformation of lung adenocarcinoma to small-cell lung cancer : Journal of Cancer Research and Therapeutics

Secondary Logo

Journal Logo

Review Article

Progress in the knowledge on the transformation of lung adenocarcinoma to small-cell lung cancer

Wang, Aiguang; Han, Cuiping; Zhao, Hui1; Zheng, Zhaomin; Ye, Xin; Shan, Rong1,

Author Information
Journal of Cancer Research and Therapeutics 19(1):p 14-19, March 2023. | DOI: 10.4103/jcrt.jcrt_1842_22
  • Open

Abstract

INTRODUCTION

Lung cancer is a common type of carcinoma and is the leading cause of cancer-related deaths worldwide. The two broad histological subtypes of lung cancer are non-small-cell lung cancer (NSCLC), which accounts for 85% of cases and includes adenocarcinoma and squamous-cell carcinoma, and small-cell lung cancer (SCLC), which accounts for 15% of cases.[1–3] Substantial improvements and refinements in the treatment have led to remarkable progress and changed outcomes for many patients. However, with an increase in the awareness of repeat biopsy, more and more patients with lung cancer have been found to undergo a histological transformation during treatment, with lung adenocarcinoma (LAdC) to SCLC transformation being the most frequent.[4–7]

MECHANISM OF THE TRANSFORMATION OF LUNG ADENOCARCINOMA TO SMALL-CELL LUNG CANCER

Acquired drug resistance in the transformation of EGFR-mutant LAdC to SCLC

Somatic activating mutations in the epidermal growth factor receptor (EGFR) tyrosine kinase domain are present in 15%–50% of patients with NSCLC.[8–11] Treatment with EGFR tyrosine kinase inhibitors (EGFR–TKIs) extend the progression-free survival (PFS) compared with that observed with chemotherapy.[12–16] Thus, EGFR–TKIs are the standard therapy for patients with EGFR mutations. However, acquired resistance inevitably develops 10–12 months after treatment when detecting the T790M mutation.[17–22] Repeat biopsy showed that several mechanisms might account for the acquired resistance including the rare (approximately 3%–10%) LAdC to SCLC histological transformation, which affects the prognosis of patients with lung cancer.[23–25]

SCLC develops from neuroendocrine cells in the central airway, whereas LAdC derives from alveolar-type II cells located on the alveolar surface. Alveolar-type II cells may be common precursors of LAdC and SCLC.[26–29] Therefore, LAdC cells with EGFR mutations originating from alveolar-type II cells may transdifferentiate into SCLC under the selective pressure of treatment.[23,28–35]

However, there are different findings about the LAdC to SCLC conversion against TKI therapy. Norkowski et al.[36] found that SCLC transformation is not associated with TKIs but might be related to carrying an EGFR mutation because only two out of six patients developing SCLC after adenocarcinoma were treated with TKIs after adenocarcinoma. Chu et al.[37] demonstrated the occurrence of neuroendocrine transformation in NSCLC in the absence of TKI targets or other treatments.

Correlations between tumor protein 53 (TP53), RB transcriptional corepressor 1 (RB1), and transformed SCLC

The TP53 gene is located on chromosome 17 short arm (17p13). Mutated or inactivated TP53 loses its tumor suppressor function.[38–41]The antiproliferative role of the p53 protein makes it a primary target for inactivation in cancer. However, it may also drive oncogenes.[42] RB1 is an important tumor suppressor gene. RB1 mutations or deletions cause the upregulation of pluripotent stem cell reprogramming factors and promote cell proliferation.[43,44] Studies have evaluated the role of the loss of TP53 and RB1 expression in neuroendocrine and alveolar-type cells and found that this loss led to SCLC.[45]

Nearly 50% of NSCLC patients have EGFR/TP53 co-mutations, which are an independent risk factor for poor PFS and overall survival (OS) in patients treated with EGFR–TKIs.[46–48]

Most lung cancers with EGFR/RB1 co-mutants are associated with TP53 mutations. TP53 is the most frequently mutated gene in lung cancer cells, with similar mutation frequencies in SCLC and NSCLC.[49,50]

The ontogeny and molecular pathogenesis of SCLC transformation with TP53 and/or RB1 mutation remain poorly understood. A high incidence of aggressive lung tumors similar to SCLC was found in mice with conditional inactivation of RB1 and TP53 in lung epithelial cells, suggesting that RB1 and TP53 inactivation is a prerequisite for SCLC pathogenesis.[51]Additionally, the EGFR/TP53/RB1 triple mutation had a six times higher risk of conversion to SCLC than that of patients with EGFR without TP53 or RB1 co-mutation,[46] and other researchers found this rate even 42.8 times higher.[52] However, not all NSCLCs with triple mutations undergo SCLC transformation. Additionally, reduced RB1 expression in EGFR-mutant LAdC is not enough to cause SCLC transformation.[46]

LAdC and SCLC resistant to EGFR–TKIs have a common clonal origin.[49] Indeed, resistant SCLCs differentiate early from LAdC clones with fully inactivated RB1 and TP53. The transformation of LAdC with EGFR mutations to SCLC is associated with the inactivation of RB1 and TP53 genes.

NOTCH signaling dysregulation is crucial for SCLC tumorigenesis, disease progression, and chemoresistance.[53–57] neurogenic locus notch homolog (NOTCH) and achaete-scute homolog 1 (ASCL1) pathways are master regulators of the neuroendocrine differentiation in small-cell lung carcinoma.[58–63] Koba proposed that the NOTCH–ASCL1 axis might also play a role in the transformation of SCLC with inactivated TP53 and RB1.[56] Meder identified a novel pathway driving the pathogenesis of secondary SCLC. It involves NOTCH inactivation mutations, the activation of the NOTCH target ASCL1, and the canonical wingless-type (WNT) signaling in the context of mutual bi-allelic RB1 and TP53 lesions. The authors experimentally verified the involvement of the NOTCH–ASCL1–RB–p53 signaling axis in vitro and validated its activation by genetic alterations in vivo.[64] Moreover, ASCL1 activation may cooperate with the biallelic RB/TP53 loss in neuroendocrine precursors in primary SCLC and contribute to secondary SCLC arising from NSCLC upon cancer therapy.[65–67]

Other hypotheses of the research

Telomerase reverse transcriptase amplification is another common genetic mechanism of SCLC transformation in patients with EGFR-mutant LAdC.[68]

A whole-genome sequencing study of LAdC to SCLC transformation revealed that the somatic mutation profile changes in SCLC after transformation, with fewer C > T and more C > A mutations. Additionally, the copy number variants (CNVs) burden of the transformed SCLC is increased compared with that in the initial LAdC (61.1 in SCLC vs. 39.0 in LAdC, Wilcoxon P = 0.4). A greater CNV burden in LAdC is associated with a shorter time to SCLC transformation, and a greater CNV burden in the transformed SCLC results in a shorter OS after transformation. A clonal evolution analysis showed different clonal components between the initial LAdC and transformed SCLC.[69]

CLINICAL CHARACTERISTICS OF THE LADC TO SCLC TRANSFORMATION

A systematic review and pooled analysis of reported cases of SCLC diagnosed in patients with EGFR-mutated (37/39) or ALK-rearranged (2/39) LAdC treated with TKIs were performed. Changes in EGFR exon 19 were observed in 19 cases and exon 21 in 9 cases. The median time to transform to SCLC was 19 months, and the median survival after transformation was 6 months. All SCLC cases had the same EGFR mutation as that present in the former adenocarcinoma.[70]

Marcoux et al.[71] retrospectively analyzed 67 patients with EGFR-mutant SCLC and other high-grade neuroendocrine carcinomas treated with one or more EGFR–TKIs before transformation to SCLC. The transformation can occur at any time point in the course of the disease. Typically, the time to conversion ranged from 2 months to 5 years, with a median time of 17.8 months. The median OS was 31.5 months (95% confidence interval [CI], 24.8–41.3 months) since the initial diagnosis and 10.9 months (95% CI, 8.0–13.7 months) since lung cancer transformation. Tissues from 59 patients were genotyped as the first evidence of SCLC. The initial EGFR mutation was maintained in all patients, and 15 out of the 19 EGFR T790M-positive mutants were converted to wild-type. Re-emergence of NSCLC clones was also found in some cases.

Ferrer et al.[72] analyzed 48 cases of EGFR-mutated NSCLC and 13 cases of non-EGFR-mutated NSCLC that were converted to SCLC. Most EGFR-mutated tumors retained the EGFR mutation after transformation. The median time to SCLC transformation in the EGFR-mutant group was shorter than that in the non-EFGR-mutant group (16 months vs. 26 months). The median OS rate was 28 months in the EGFR-mutant group and 37 months in the non-EFGR-mutant group. After transformation, the median OS was 10 months in the EGFR-mutant group and 9 months in the non-EGFR-mutant group.

Central nervous system (CNS) metastases are frequent after transformation. Among 59 patients, 38 (64%) experienced CNS progression at some point after the SCLC diagnosis. The median follow-up time after conversion to SCLC was 8.1 months (range of 0–26.9 months), and 45 deaths (67%) occurred. The median OS of patients with metastatic lung cancer was 31.5 months since the first diagnosis and 10.9 months since SCLC formation.[71]

THERAPEUTIC STRATEGIES AGAINST TRANSFORMED SCLC

The molecular characterization of NSCLC biomarkers significantly improved global outcomes for patients. Despite the recent understanding of SCLC biology, effective biomarkers for SCLC are lacking.[52,73] The clinical treatment of NSCLC after histological transformation remains challenging, and little data are available to guide clinical decision-making.

Platinum–etoposide is the most commonly used regimen after transformation to SCLC or de novo diagnosis of SCLC.[74] The clinical response rate in patients treated with platinum–etoposide is 54%. A high response rate (eight [80%] out of 10 patients) was achieved in patients previously receiving platinum chemotherapy for adenocarcinoma. Estimated median PFS with platinum–etoposide treatment was 3.4 months (95% CI, 2.4 to 5.4 months). No responses are observed among patients receiving checkpoint inhibitors, such as programmed death-1 or programmed death-ligand 1 inhibitor, either as a single agent or in combination with the ipilimumab–nivolumab regimen.[71] Accordingly, we speculated that NSCLC cells sensitive to taxanes exist in patients presenting transformed SCLC with an EGFR mutation. Additionally, transformed SCLC with EGFR mutations might be more sensitive to taxanes than de novo-formed SCLC.[71] Another group proposed that, although EGFR mutations are found in SCLC after transformation, the EGFR protein expression is significantly reduced, rendering the transformed tumors unresponsive to EGFR–TKIs.[47]

A multicenter retrospective study reported that anlotinib is effective in patients with small-cell transformation after third-generation TKI treatment. These patients have a better prognosis than patients with transformation after first or second-generation treatment (49.4 months vs. 20.0 months, P = 0.013).[75]

Furthermore, the C797S and T790MEGFR mutations were shown to guide the selection of TKIs for the treatment of transformed SCLC. Indeed, cells with C797S and T790M mutations in trans are resistant to third-generation EGFR–TKIs but sensitive to a combination of first- and third-generation TKIs. In contrast, the activity of EGFR with cis mutations cannot be inhibited by EGFR–TKIs alone or in combination. T790M wild-type cells with C797S mutation (for which third-generation TKIs are the preferred treatment) are resistant to third-generation TKIs but remain sensitive to first-generation TKIs.[76]

PREDICTORS OF SCLC TRANSFORMATION

Neuron-specific enolase (NSE) and pro-gastrin-releasing peptide (ProGRP) serum levels are increased at the time of the second biopsy and decreased following EGFR–TKI treatment. Thus, ProGRP and NSE may be useful for the early detection of SCLC transformation in patients with resistance to EGFR–TKIs and in whom secondary biopsy should be performed.[77]

The EGFR/TP53/RB1 triple mutation may increase the risk of SCLC transformation. Patients with EGFR/RB1/TP53-mutant lung cancer have a largely higher incidence of whole-genome doubling compared with that in NSCLC and SCLC. They also show further enrichment in triple-mutant cancers with eventual small-cell histology. A mutation in activation-induced cytidine deaminase/apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like is enriched in transformed triple-mutant lung cancers. Thus, activation-induced cytidine deaminase hypermutation might be an early predictor of SCLC transformation.[46,78]

However, not all LAdC-transformed SCLCs harbor the EGFR mutation. Fujita et al.[6] described SCLC transformation in an ALK-positive patient treated with alectinib for 9 months. Another case of SCLC transformation was reported by Toyokawa in a 70-year-old male patient smoker with advanced rectal cancer who received 5-fluorouracil-based chemotherapy.[79]

LAdC to SCLC transformation might be a mechanism of TKI treatment resistance. However, ALK-positive LAdC treated with the targeted drug or LAdC without mutations but subjected to chemotherapy might also be converted into SCLC. We hypothesize that LAdC to SCLC transformation is a common event resulting from the breakthrough in drug development against LAdC that significantly improves patients’ survival. When LAdC progresses or has a poor prognosis, repeat biopsies are often performed for choosing a new therapeutic regimen, and are very likely to reveal altered histopathology. It can also be inferred that LAdC and SCLS have a common origin, and even that SCLC might be a worse differentiation stage of LAdC.

Financial support and sponsorship

The study was funded by the National Natural Science Foundation of China (Nos. 81901851 and 82072028).

Conflicts of interest

There are no conflicts of interest.

Acknowledgments

The Institutional Review Board of the First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Public Health Clinical Center has approved this study. Informed consent was obtained from all individuals included in the study. The work has not been published elsewhere. All the authors listed have seen and approved the manuscript that is enclosed, and contributed significantly to the work.

REFERENCES

1. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet 2021;398:535–54.
2. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022;72:7–33.
3. Dela Cruz CS, Tanoue LT, Matthay RA. Lung cancer:Epidemiology, etiology, and prevention. Clin Chest Med 2011;32:605–44.
4. Hirakawa H, Komiya K, Nakashima C, Ogusu S, Nakamura T, Tanaka M, et al. A case of osimertinib-resistant lung adenocarcinoma responded effectively to alternating therapy. Ann Transl Med 2018;6:464.
5. Taniguchi Y, Horiuchi H, Morikawa T, Usui K. Small-cell carcinoma transformation of pulmonary adenocarcinoma after osimertinib treatment:A case report. Case Rep Oncol 2018;11:323–9.
6. Fujita S, Masago K, Katakami N, Yatabe Y. Transformation to SCLC after treatment with the ALK inhibitor alectinib. J Thorac Oncol 2016;11:e67–72.
7. Ahmed T, Vial MR, Ost D, Stewart J, Hasan MA, Grosu HB. Non-small cell lung cancer transdifferentiation into small cell lung cancer:A case series. Lung Cancer 2018;122:220–3.
8. Rosell R, Moran T, Queralt C, Porta R, Cardenal F, Camps C, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med 2009;361:958–67.
9. Shi Y, Au JS, Thongprasert S, Srinivasan S, Tsai CM, Khoa MT, et al. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J Thorac Oncol 2014;9:154–62.
10. Roh MS, Yoon NB, Lee S, Kang BH, Um SJ, Lee DH, et al. Correlation of epidermal growth factor receptor mutation status in plasma and tissue samples of patients with non-small cell lung cancer. J Cancer Res Ther 2020;16:843–9.
11. Gaur P, Bhattacharya S, Kant S, Kushwaha RAS, Singh G, Pandey S. Association of cytokines levels with epidermal growth factor receptor mutation in lung cancer patients. J Cancer Res Ther 2020;16:811–5.
12. Minari R, Bordi P, Del Re M, Facchinetti F, Mazzoni F, Barbieri F, et al. Primary resistance to osimertinib due to SCLC transformation:Issue of T790M determination on liquid re-biopsy. Lung Cancer 2018;115:21–7.
13. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947–57.
14. Zhu CM, Lian XY, Zhang HY, Bai L, Yun WJ, Zhao RH, et al. EGFR tyrosine kinase inhibitors alone or in combination with chemotherapy for non-small-cell lung cancer with EGFR mutations:A meta-analysis of randomized controlled trials. J Cancer Res Ther 2021;17:664–70.
15. Reck M, Rabe KF. Precision diagnosis and treatment for advanced non-small-cell lung cancer. N Engl J Med 2017;377:849–61.
16. Popat S, Wotherspoon A, Nutting CM, Gonzalez D, Nicholson AG, O'Brien M. Transformation to “high grade”neuroendocrine carcinoma as an acquired drug resistance mechanism in EGFR-mutant lung adenocarcinoma. Lung Cancer 2013;80:1–4.
17. Engelman JA, Jänne PA. Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin Cancer Res 2008;14:2895–9.
18. He J, Huang Z, Han L, Gong Y, Xie C. Mechanisms and management of 3rd generation EGFR TKI resistance in advanced non small cell lung cancer (Review). Int J Oncol 2021;59:90.
19. Tang ZH, Lu JJ. Osimertinib resistance in non-small cell lung cancer:Mechanisms and therapeutic strategies. Cancer Lett 2018;420:242–6.
20. Westover D, Zugazagoitia J, Cho BC, Lovly CM, Paz-Ares L. Mechanisms of acquired resistance to first- and second-generation EGFR tyrosine kinase inhibitors. Ann Oncol 2018;29:10–9.
21. Reita D, Pabst L, Pencreach E, Guérin E, Dano L, Rimelen V, et al. Molecular mechanism of EGFR-TKI resistance in EGFR-mutated non-small cell lung cancer:Application to biological diagnostic and monitoring. Cancers (Basel) 2021;13:4926.
22. Mu Y, Hao X, Xing P, Hu X, Wang Y, Li T, et al. Acquired resistance to osimertinib in patients with non-small-cell lung cancer:Mechanisms and clinical outcomes. J Cancer Res Clin Oncol 2020;146:2427–33.
23. Shaurova T, Zhang L, Goodrich DW, Hershberger PA. Understanding lineage plasticity as a path to targeted therapy failure in EGFR-mutant non-small cell lung cancer. Front Genet 2020;11:281.
24. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;3:75ra26.
25. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res 2013;19:2240–7.
26. Oser MG, Niederst MJ, Sequist LV, Engelman JA. Transformation from non-small-cell lung cancer to small-cell lung cancer:Molecular drivers and cells of origin. Lancet Oncol 2015;16:e165–72.
27. Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem cells in lung development, renewal and cancer. Natur 2014;507:190–4.
28. Park KS, Liang MC, Raiser DM, Zamponi R, Roach RR, Curtis SJ, et al. Characterization of the cell of origin for small cell lung cancer. Cell Cycle 2011;10:2806–15.
29. Lin C, Song H, Huang C, Yao E, Gacayan R, Xu SM, et al. Alveolar type II cells possess the capability of initiating lung tumor development. PLoS One 2012;7:e53817.
30. Shi X, Duan H, Liu X, Zhou L, Liang Z. Genetic alterations and protein expression in combined small cell lung cancers and small cell lung cancers arising from lung adenocarcinomas after therapy with tyrosine kinase inhibitors. Oncotarget 2016;7:34240–9.
31. Jiang Y, Shou L, Guo Q, Bao Y, Xu X, An S, et al. Small-cell lung cancer transformation from EGFR-mutant adenocarcinoma after EGFR-TKIs resistance:A case report. Medicine (Baltimore) 2021;100:e26911.
32. Hwang S, Hong TH, Park S, Jung HA, Sun JM, Ahn JS, et al. Molecular subtypes of small cell lung cancer transformed from adenocarcinoma after EGFR tyrosine kinase inhibitor treatment. Transl Lung Cancer Res 2021;10:4209–20.
33. Jin CB, Yang L. Histological transformation of non-small cell lung cancer:Clinical analysis of nine cases. World J Clin Cases 2021;9:4617–26.
34. Yang H, Liu L, Zhou C, Xiong Y, Hu Y, Yang N, et al. The clinicopathologic of pulmonary adenocarcinoma transformation to small cell lung cancer. Medicine (Baltimore) 2019;98:e14893.
35. Zhang C, Lin L, Guo X, Chen P. Significance of genetic sequencing in patients with lung adenocarcinoma with transformation to small cell lung cancer:A case report and systematic review. Transl Cancer Res 2020;9:3725–33.
36. Norkowski E, Ghigna MR, Lacroix L, Le Chevalier T, Fadel É, Dartevelle P, et al. Small-cell carcinoma in the setting of pulmonary adenocarcinoma:New insights in the era of molecular pathology. J Thorac Oncol 2013;8:1265–71.
37. Chu X, Xu Y, Li Y, Zhou Y, Chu L, Yang X, et al. Neuroendocrine transformation from EGFR/ALK-wild type or TKI-naïve non-small cell lung cancer:An under-recognized phenomenon. Lung Cancer 2022;169:22–30.
38. Olivier M, Hollstein M, Hainaut P. TP53 mutations in human cancers:Origins, consequences, and clinical use. Cold Spring HarbPerspect Biol 2010;2:a001008.
39. Muller PA, Vousden KH. p53 mutations in cancer. Nat Cell Biol 2013;15:2–8.
40. Brosh R, Rotter V. When mutants gain new powers:News from the mutant p53 field. Nat Rev Cancer 2009;9:701–13.
41. George J, Lim JS, Jang SJ, Cun Y, Ozretić L, Kong G, et al. Comprehensive genomic profiles of small cell lung cancer. Nature 2015;524:47–53.
42. Wilkie M, Lau A, Vlatkovic N, Jones T, Boyd M. Tumour metabolism in squamous cell carcinoma of the head and neck:An in-vitro study of the consequences of TP53 mutation and therapeutic implications. Lancet 2015;385;Suppl 1;S101.
43. Kareta MS, Gorges LL, Hafeez S, Benayoun BA, Marro S, Zmoos AF, et al. Inhibition of pluripotency networks by the Rb tumor suppressor restricts reprogramming and tumorigenesis. Cell Stem Cell 2015;16:39–50.
44. Peifer M, Fernández-Cuesta L, Sos ML, George J, Seidel D, Kasper LH, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat Genet 2012;44:1104–10.
45. Sutherland KD, Proost N, Brouns I, Adriaensen D, Song JY, Berns A. Cell of origin of small cell lung cancer:Inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 2011;19:754–64.
46. Offin M, Chan JM, Tenet M, Rizvi HA, Shen R, Riely GJ, et al. Concurrent RB1 and TP53 alterations define a subset of EGFR-mutant lung cancers at risk for histologic transformation and inferior clinical outcomes. J Thorac Oncol 2019;14:1784–93.
47. Molina-Vila MA, Bertran-Alamillo J, Gascó A, Mayo-de-las-Casas C, Sánchez-Ronco M, Pujantell-Pastor L, et al. Nondisruptive p53 mutations are associated with shorter survival in patients with advanced non-small cell lung cancer. Clin Cancer Res 2014;20:4647–59.
48. Niederst MJ, Sequist LV, Poirier JT, Mermel CH, Lockerman EL, Garcia AR, et al. RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer. Nat Commun 2015;6:6377.
49. Lin MW, Su KY, Su TJ, Chang CC, Lin JW, Lee YH, et al. Clinicopathological and genomic comparisons between different histologic components in combined small cell lung cancer and non-small cell lung cancer. Lung Cancer 2018;125:282–90.
50. Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Primers 2021;7:3.
51. Meuwissen R, Linn SC, Linnoila RI, Zevenhoven J, Mooi WJ, Berns A. Induction of small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a conditional mouse model. Cancer Cell 2003;4:181–9.
52. Lee JK, Lee J, Kim S, Kim S, Youk J, Park S, et al. Clonal history and genetic predictors of transformation into small-cell carcinomas from lung adenocarcinomas. J Clin Oncol 2017;35:3065–74.
53. Leonetti A, Facchinetti F, Minari R, Cortellini A, Rolfo CD, Giovannetti E, et al. Notch pathway in small-cell lung cancer:From preclinical evidence to therapeutic challenges. Cell Oncol (Dordr) 2019;42:261–73.
54. Wael H, Yoshida R, Kudoh S, Hasegawa K, Niimori-Kita K, Ito T. Notch1 signaling controls cell proliferation, apoptosis and differentiation in lung carcinoma. Lung Cancer 2014;85:131–40.
55. Hassan WA, Yoshida R, Kudoh S, Hasegawa K, Niimori-Kita K, Ito T. Notch1 controls cell invasion and metastasis in small cell lung carcinoma cell lines. Lung Cancer 2014;86:304–10.
56. Collins BJ, Kleeberger W, Ball DW. Notch in lung development and lung cancer. Semin Cancer Biol 2004;14:357–64.
57. Koba H, Kimura H, Yoneda T, Ogawa N, Tanimura K, Tambo Y, et al. NOTCH alteration in EGFR-mutated lung adenocarcinoma leads to histological small-cell carcinoma transformation under EGFR-TKI treatment. Transl Lung Cancer Res 2021;10:4161–73.
58. Westhoff B, Colaluca IN, D'Ario G, Donzelli M, Tosoni D, Volorio S, et al. Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci U S A 2009;106:22293–8.
59. Borges M, Linnoila RI, van de Velde HJ, Chen H, Nelkin BD, Mabry M, et al. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 1997;386:852–5.
60. Linnoila RI, Zhao B, DeMayo JL, Nelkin BD, Baylin SB, DeMayo FJ, et al. Constitutive achaete-scute homologue-1 promotes airway dysplasia and lung neuroendocrine tumors in transgenic mice. Cancer Res 2000;60:4005–9.
61. Ouadah Y, Rojas ER, Riordan DP, Capostagno S, Kuo CS, Krasnow MA. Rare pulmonary neuroendocrine cells are stem cells regulated by Rb, p53, and Notch. Cell 2019;179:403–16 e23.
62. Lim JS, Ibaseta A, Fischer MM, Cancilla B, O'Young G, Cristea S, et al. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature 2017;545:360–4.
63. Augert A, Eastwood E, Ibrahim AH, Wu N, Grunblatt E, Basom R, et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci Signal 2019;12:eaau2922.
64. Meder L, König K, Ozretić L, Schultheis AM, Ueckeroth F, Ade CP, et al. NOTCH, ASCL1, p53 and RB alterations define an alternative pathway driving neuroendocrine and small cell lung carcinomas. Int J Cancer 2016;138:927–38.
65. Osada H, Tomida S, Yatabe Y, Tatematsu Y, Takeuchi T, Murakami H, et al. Roles of achaete-scute homologue 1 in DKK1 and E-cadherin repression and neuroendocrine differentiation in lung cancer. Cancer Res 2008;68:1647–55.
66. Jiang T, Collins BJ, Jin N, Watkins DN, Brock MV, Matsui W, et al. Achaete-scute complex homologue 1 regulates tumor-initiating capacity in human small cell lung cancer. Cancer Res 2009;69:845–54.
67. Hassan WA, Takebayashi SI, Abdalla MOA, Fujino K, Kudoh S, Motooka Y, et al. Correlation between histone acetylation and expression of Notch1 in human lung carcinoma and its possible role in combined small-cell lung carcinoma. Lab Invest 2017;97:913–21.
68. Mc Leer A, Foll M, Brevet M, Antoine M, Novello S, Mondet J, et al. Detection of acquired TERT amplification in addition to predisposing p53 and Rb pathways alterations in EGFR-mutant lung adenocarcinomas transformed into small-cell lung cancers. Lung Cancer 2022;167:98–106.
69. Xie T, Li Y, Ying J, Cai W, Li J, Lee KY, et al. Whole exome sequencing (WES) analysis of transformed small cell lung cancer (SCLC) from lung adenocarcinoma (LUAD). Transl Lung Cancer Res 2020;9:2428–39.
70. Roca E, Gurizzan C, Amoroso V, Vermi W, Ferrari V, Berruti A. Outcome of patients with lung adenocarcinoma with transformation to small-cell lung cancer following tyrosine kinase inhibitors treatment:A systematic review and pooled analysis. Cancer Treat Rev 2017;59:117–22.
71. Marcoux N, Gettinger SN, O'Kane G, Arbour KC, Neal JW, Husain H, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas:Clinical outcomes. J Clin Oncol 2019;37:278–85.
72. Ferrer L, GiajLevra M, Brevet M, Antoine M, Mazieres J, Rossi G, et al. A brief report of transformation from NSCLC to SCLC:Molecular and therapeutic characteristics. J Thorac Oncol 2019;14:130–4.
73. Sabari JK, Lok BH, Laird JH, Poirier JT, Rudin CM. Unravelling the biology of SCLC:Implications for therapy. Nat Rev Clin Oncol 2017;14:549–61.
74. Nagy-Mignotte H, Guillem P, Vignoud L, Coudurier M, Vesin A, Bonneterre V, et al. Outcomes in recurrent small-cell lung cancer after one to four chemotherapy lines:A retrospective study of 300 patients. Lung Cancer 2012;78:112–20.
75. Wang W, Xu C, Chen H, Jia J, Wang L, Feng H, et al. Genomic alterations and clinical outcomes in patients with lung adenocarcinoma with transformation to small cell lung cancer after treatment with EGFR tyrosine kinase inhibitors:A multicenter retrospective study. Lung Cancer 2021;155:20–7.
76. Niederst MJ, Hu H, Mulvey HE, Lockerman EL, Garcia AR, Piotrowska Z, et al. The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies. Clin Cancer Res 2015;21:3924–33.
77. Watanabe S, Sone T, Matsui T, Yamamura K, Tani M, Okazaki A, et al. Transformation to small-cell lung cancer following treatment with EGFR tyrosine kinase inhibitors in a patient with lung adenocarcinoma. Lung Cancer 2013;82:370–2.
78. Farago AF, Piotrowska Z, Sequist LV. Unlocking the mystery of small-cell lung cancer transformations in EGFR mutant adenocarcinoma. J Clin Oncol 2017;35:2987–8.
79. Toyokawa G, Taguchi K, Ohba T, Morodomi Y, Takenaka T, Hirai F, et al. First case of combined small-cell lung cancer with adenocarcinoma harboring EML4-ALK fusion and an exon 19, EGFR mutation in each histological component. J Thorac Oncol 2012;7:e39–41.
Keywords:

Lung adenocarcinoma; small-cell lung cancer; transformation

Copyright: © 2023 Journal of Cancer Research and Therapeutics