Skip to main content

Patients outcomes in lung adenocarcinoma transforming to small-cell lung cancer after tyrosine kinase inhibitor therapy

Abstract

Background

Non-small cell lung cancer (NSCLC) transforming to small cell lung cancer (SCLC) is one of the mechanisms of resistance to tyrosine kinase inhibitors (TKIs). Cases of NSCLC transforming into SCLC have been discovered. However, we lack concentrated data on the characteristics of this population and the transformed SCLC to aid our insight of the biology and clinical value of NSCLC transforming with positive.

Methods

We systematically reviewed the published literatures and summarized the pathological and clinical characteristics, and the prognosis, of published cases.

Results

140 patients with lung adenocarcinoma (LUAD) were included in this study, with a median age of 56.8 years. The median time from the first diagnosis of LUAD transforming to SCLC (ttSCLC) was 20.0 months. The median overall survival (mOS) after the diagnosis of SCLC was 11.0 months (95% CI, 7.41 to 14.59 months). In the univariate analysis, ever smoking (either former or current) was a promising predictor of a shorter ttSCLC (HR, 1.73; 95% CI, 1.14 to 2.62; P = 0.010). TKIs therapy administered as a second line and beyond treatment was related to a significant delay in SCLC onset compared to first-line therapy (HR, 0.62; 95% CI, 0.40 to 0.96; P = 0.031). The median progression-free survival (mPFS) on first-line platinum plus etoposide after the conversion to SCLC was 3.0 months. Female appeared to be related to worse outcomes after transformation of SCLC.

Conclusion

Transformed SCLC exhibited poor response to primary SCLC classic chemotherapy and immunotherapy. It carries a worse prognosis. Exploring novel therapeutic strategies for transformed SCLC is imperative.

Introduction

Lung cancer ranks among the most prevalent malignancies globally and stands as the primary contributor to cancer-related fatalities [1]. It can be categorized into two primary pathological subtypes: non-small cell lung cancer (NSCLC) (85%), with approximately 50% being lung adenocarcinoma (LUAD), and small cell lung cancer (SCLC) (15%) [2]. Generally, SCLC is characterized by high aggressiveness, malignancy, and poor outcomes. The 5-year survival rate is only 5% to 10% [1]. Transformed SCLC is a phenotype of typical SCLC.

Epidermal growth factor receptor (EGFR) mutations are identified in about 50% of advanced NSCLC in Asian patients [3]. Anaplastic lymphoma kinase (ALK)-positive tumors constitute approximately 3–7% of NSCLC [4, 5]. Tumors carrying targetable driver oncogenes such as EGFR, ALK, rearranged during transfection (RET) and c-ros oncogene 1 (ROS1) inevitably face treatment resistance and disease progression. Histological transformation into SCLC is among the mechanisms of resistance to TKIs in NSCLC. Lineage plasticity in cancer can result in the transformation of tumor cells into different histological subtypes, leading to treatment resistance [6]. Studies on the mechanism of EGFR-TKIs resistance have revealed that SCLC transformation occurs in 3–14% of TKIs-resistant tumors [7, 8]. A less common and poorly understood resistance mechanism observed in around 1.2% of TKI-resistant ALK-positive NSCLC is lineage conversion, typically manifesting as a histological transformation from adenocarcinoma to neuroendocrine or squamous histology [9]. Neuroendocrine carcinomas are often sensitive, albeit transiently, to platinum-etoposide but are resistant to immune checkpoint inhibitors(ICIs) [10, 11] and have a poorer prognosis [12].In 2006, the first case of SCLC transformed from NSCLC after treatment with EGFR-TKIs was reported [13]. Consequently, numerous reports on the transformation of NSCLC to SCLC and related research have emerged. However, the literature on the patient characteristics, treatment, and prognosis of SCLC transformed from TKI resistance is still limited, and there is a lack of studies with large sample sizes. Transformed SCLC is more aggressive and has a worse prognosis [14], and there are currently no unified treatment guidelines for transformed SCLC.

Therefore, we systematically reviewed the literatures on SCLC patients who transformed from LUAD carrying targeted mutations after receiving TKIs treatment, and aimed to obtain exploratory information on the clinicopathological characteristics of patients with phenotypic transformation of SCLC, and to provide clinical reference for clinicians with the efficacy and prognosis.

Patients and methods

Search strategy

We carried out an extensive review of the literature using the PubMed/Medline database from 2006 to the present. Our search strategy included keywords like "transition from NSCLC to SCLC," "NSCLC conversion to SCLC," "TKI resistance," and "TKI therapy." The search was specifically limited to the human studies in English. Additionally, we manually reviewed reference lists in relevant publications to identify any additional articles. It is worth noting that we excluded scientific conference abstracts from our systematic literature search, considering that the general data and treatment information of the cases available in scientific conference abstracts are limited. In cases of duplicated publications, we opted for the most recent version. The PRISMA flow diagram for this systematic review was shown in Fig. 1.

Fig. 1
figure 1

PRISMA flow-chart of this study

Inclusion and exclusion criteria

We included case reports that recorded a histopathological shift to SCLC after previous treatment with TKIs for LUAD. The TKIs encompassed in this study included Gefitinib, Icotinib, Erlotinib, Afatinib, Osimertinib, Aumolertinib, Crizotinib, Alectinib, Ceritinib, or Pralsetinib. The diagnosis of SCLC had to be confirmed through high-quality tumor biopsies or well-preserved cytological samples in accordance with the 2015 WHO classification [15].

Statistical analysis

Demographic details, tumor histology, clinical treatments, molecular pathology, and outcomes for all reported cases were meticulously extracted from the included literatures by Shuai Wang and Yongsen Wang. This information was then compiled into a dedicated database and subjected to detailed analysis as a case series.

In our study, we defined ttSCLC as the duration between the initial pathological diagnosis of LUAD and the subsequent biopsy confirming the presence of metachronous SCLC phenotype. The T-ttSCLC interval referred to the time elapsed from the initiation of TKIs treatment to the additional biopsy that revealed the metachronous SCLC phenotype. The t-patients referred to the patients from the initial pathological diagnosis of LUAD to the additional biopsy revealing the metachronous SCLC phenotype. The T-patients referred to the patients from the initial TKIs usage to the additional biopsy revealing the metachronous SCLC phenotype. Survival curves were generated using the Kaplan–Meier method and compared using the log-rank test. Exploratory analyses were conducted using Cox proportional hazards regression models to evaluate the predictive value of patient and tumor characteristics (reported as hazard ratios and 95% confidence intervals) for ttSCLC. Statistical significance was set at P values < 0.05 (two-sided). We performed all statistical analyses using SPSS version 25.0 (SPSS, Armonk, USA) and R package version 4.3.2 (R Foundation for Statistical Computing, Vienna, Austria). For graphical representation, we utilized GraphPad Prism version 8.0.0 (GraphPad Software, San Diego, USA).

Results

Case characteristics of the incorporated studies

Our search approach yielded a total of 706 articles (Fig. 1). Within this pool,76 articles constituted pertinent accounts of cases undergoing histopathological transformation from LUAD to SCLC after TKIs treatment. Out of the 76 literatures, 61 were individual case reports [10, 16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75], while 15 presented findings small patient series [76,77,78,79,80,81,82,83,84,85,86,87,88,89,90]. In aggregate, we identified 140 patients who experienced the transformation to SCLC from a previous diagnosis of adenocarcinoma.

Pre-transformation characteristics

Patients characteristics

The baseline characteristics of the 140 identified cases were encapsulated in Table 1.

Table 1 Clinical characteristics of patients with mutated lung adenocarcinoma transforming to SCLC

Among the available data for 131 patients, 64 were male (48.9%), and 67 were female (51.1%). Available in 122 patients, the median age stood at 56.8 years (with a range of 31–84 years). Among 126 patients where smoking status data were available, 43 patients (34.1%), including former smokers, tested positive, while the majority, 83 patients (65.9%), had never smoked. The majority, 83 patients (65.9%), were never smokers. Nearly all cases (99%) were diagnosed of advanced-stage LUAD (TNM stage III or IV), with positivity noted for EGFR mutation (122/140), ALK rearrangement (16/140), ROS1 fusion (1/140) or CCDC6: RET rearrangement (1/140).EGFR mutation profiles were distributed as follows: 93 patients (66.4%) had exon 19-deletion, 24 patients (17.1%) exhibited exon 21-L858R mutation, one patient (0.7%) showed co-mutations of exon 21-L858R and exon 18-G719X, another patient (0.7%) presented with exon 20-S768I mutation, and one patient (0.7%) had exon 18-G719X mutation. Two patients were reported with an EGFR mutation without specifying the involved exon. The remaining 18 patients (12.9%) presented with ALK rearrangement in 16 patients (11.4%), ROS1 fusion in one patient (0.7%) and CCDC6: RET rearrangement in one patient (0.7%).

Among 140 patients, information on TKIs medication was available for 124 patients. Out of the 106 patients harboring EGFR mutations, 88 (83.0%) received first-line therapy, while 18(17.0%) received second-line therapy. 79 patients (74.5%) received first-generation TKIs (Gefitinib, Icotinib, Erlotinib), nine patients (8.5%) received second-generation TKI (Afatinib), and 18 patients (17.0%) received third-generation TKIs (Osimertinib, Aumolertinib). Among the 16 patients with ALK rearrangement, (25.0%) were treated as first-line therapy, 10 (62.5%) as second-line therapy, one (6.25%) as third-line therapy, and one (6.25%) as fifth-line therapy. Out of these, 11 patients (68.75%) used first-generation TKIs (Crizotinib), while five patients (31.25%) opted for second-generation TKIs (Alectinib, Ceritinib). Additionally, one patient with ROS1 fusion received Crizotinib as second-line treatment, and another patient with CCDC6: RET fusion underwent first-line treatment with Pralsetinib, a tyrosine protein kinase receptor RET inhibitor.

Predictors of SCLC transformation

The projected median duration to ttSCLC was 20.0 months (95% CI, 16.90 to 23.11 months), whereas the anticipated median timeframe to T-ttSCLC stood at 17.0 months (95% CI, 13.44 to 20.57 months) (Fig. 2A). Compared with non-smoking patients, the ttSCLC was significantly shorter in smoking patients (16.0 months vs. 22.7 months, P = 0.009) (Fig. 2B). Assessing the number of lines of TKI application before transformation, there was a significant reduction in ttSCLC when TKI was used in the first-line treatment compared to later-line treatment (18.0 months vs. 28.0 months, P = 0.027) (Fig. 2C). Gender, age, TNM stage, and initial gene mutation type (EGFR vs. ALK) showed no impact on ttSCLC. In the univariate analysis, ever smoking (either former or current) emerged as a predictor of shorter ttSCLC (HR,1.73; 95% CI, 1.14 to 2.62; P = 0.01). Administering TKIs therapy as second-line treatment was related to a significant delay in the onset of SCLC compared to first-line therapy (HR, 0.62;95% CI, 0.40 to 0.96; P = 0.031). Moreover, stage IV was related to an early occurrence of the SCLC phenotype, approaching borderline significance (HR, 1.78; 95% CI, 0.90 to 3.52; P = 0.098). Age, gender and mutation type (EGFR vs. ALK) showed no association with ttSCLC (Fig. 3).

Fig. 2
figure 2

Time to Small Cell Lung Cancer and Overall Survival (OS). A Kaplan–Meier estimates of time from initial diagnosis of LUAD to development of SCLC phenotype (ttSCLC) and time from start of TKI treatment to development of SCLC phenotype (T-ttSCLC). B Kaplan–Meier estimates of ttSCLC according to the smoking status (current or former smokers vs. never smokers). C Kaplan–Meier estimates ttSCLC according to TKI-line treatment (1st line vs. 2nd line and beyond). D Kaplan–Meier estimates of OS after transformed and OS since diagnosed. E Kaplan–Meier estimates of OS after occurrence of SCLC according to gender (Male vs. Female). F Kaplan–Meier estimates of OS from initial diagnosis of LUAD and OS after occurrence of SCLC in ICIs subgroups

Fig. 3
figure 3

Forest plots of the effect of several factors on time from initial pathological diagnosis to development of SCLC phenotype

Post-transformed characteristics

First-line therapy and efficacy

First-line therapeutic data post-transformation were available for a total of 86 cases. (Table 2). Among these, 46 cases (53.5%) received the platinum + etoposide chemotherapy regimen, and the mPFS was 3.0 months (95% CI, 2.05 to 3.95 months)0.19 cases (22.1%) received the platinum + etoposide + TKI regimen, with a mPFS of 3.2 months (95% CI, 2.21 to 4.19 months). 21 cases (24.4%) received other therapies. After transformation to SCLC, the mOS was 5.7 months in the platinum plus etoposide group. The mOS since diagnosis in the platinum + etoposide group and the platinum + etoposide + TKI group were 42.7 months and 59.3 months, respectively, with no statistically significant difference (P = 0.096).

Table 2 First-line treatment and efficacy after SCLC transformation

Thirteen patients received ICIs after transformation to SCLC (Table 3). Among them, four patients (30.8%) used ICIs for first-line treatment, while nine patients (69.2%) used ICIs for late-line treatment (3rd line and beyond). Immunotherapy efficacy information was available for 11 patients, with an objective response rate (ORR) of 33.3% and a mPFS of 3.0 months (95% CI, 1.58 to 4.42 months). Among these, six cases were treated with EP plus anti-PD-1/PD-L1, and the mPFS was 6.0 months (95% CI, 1.38 to 10.62 months). Treatment information for anti-PD-1/PD-L1 was available for 3 patients, with a mPFS of 2.0 months (95% CI, 1.20 to 2.80 months).

Table 3 Clinical characteristics of patients undergoing immunotherapy after transformation to SCLC

Survival after LUAD transformation to SCLC

Survival data post-SCLC diagnosis was accessible for all 140 patients. The calculated mOS post-SCLC diagnosis was determined to be 11.0 months (95% CI, 7.41 to 14.59 months). And, the mOS from the time of LUAD diagnosis was 44.0 months (95% CI, 35.10 to 52.90 months) (Fig. 2D). In comparison to male, female had a shorter OS after converting to SCLC (19.0 months vs. 86.0 months, P = 0.002) (Fig. 2E). However, smoking status, initial mutation type (EGFR vs. ALK), age, TNM stage, and the number of TKI treatment lines had no statistically significant effects on OS after conversion to SCLC. Subgroup analysis using ICIs after conversion to SCLC revealed that the mOS after conversion to SCLC was 18.0 months (95% CI, 10.34 to 25.66 months), and the mOS from the diagnosis to death was 48.0 months (95% CI, 8.40 to 87.54 months)(Fig. 2F).

Discussion

According to reports, histological transformation from NSCLC to SCLC is identified as one of the mechanisms of therapeutic resistance in patients receiving TKIs targeting EGFR, ALK, and ROS1 or immunotherapy [12, 91]. We conducted a systematic search and provided a summary of published studies concerning the transition from LUAD to SCLC, encompassing EGFR, ALK, ROS1, CCDC6: RET, and other potentially targetable driver gene mutations.

As commonly known, resistance to TKIs generally occurs in about one year. Our analysis revealed that the median time estimated for ttSCLC was 20.0 months (95% CI, 16.90 to 23.11 months), and the median time estimated for T-ttSCLC was 17.0 months (95% CI, 13.44 to 20.57 months). When comparing first-line treatment to later-line treatment, the use of TKIs in the first-line significantly shortened ttSCLC (18.0 months vs. 28.0 months, P = 0.027). Univariate analysis revealed a significant delay in the onset of SCLC when TKIs therapy was administered as a second-line treatment compared to first-line treatment (HR, 0.62; 95% CI, 0.40 to 0.96; P = 0.031).

Previous studies have shown that classic SCLC is closely related to exposure to tobacco carcinogens [92]. In our research, we explored factors predicting histological transformation of SCLC and found that smoking was an independent risk factor associated with shorter ttSCLC, suggesting that tobacco exposure may facilitate the histological transformation of NSCLC to SCLC. In the study results of Elisa Roca et al., female was associated with longer ttSCLC [12]. Li et al. found transformed-SCLC showed an immuno-exhausted status which was associated with the duration of EGFR-TKI before transformation [93]. Our analysis suggested that transformation of SCLC may require long-term exposure to TKIs. Moreover, in our study, female sex is associated with shorter post-conversion OS. This may be related to the exhaustion of the tumor immune microenvironment (TIME) caused by long-term exposure to TKIs. Therefore, Gender appears to be one variable that predicts OS after SCLC transformation. In our study, exon 19-deletion was more common in EGFR mutation profiles. The results of Nicolas et al. showed that among 63 patients with LUAD who underwent SCLC transformation and had EGFR mutations, including exon 19 deletion mutations and exon 21 L858R mutations, exon 19 deletion mutations accounted for 69% [11]. This is consistent with our research conclusions. Facchinetti et al. found no phenomenon related to the histological transformation of SCLC by studying the molecular mechanisms of BRAF V600E NSCLC resistance to BRAF/MEK inhibitors [94]. Case of NSCLC in which CCDC6:RET fusion was found in our study have been reported [63], which was inconsistent with the results of Lin et al.'s study on the resistance of RET fusion-positive NSCLC cancers to selective RET tyrosine kinase inhibitors [95]. In the future, we may discover more LUAD carrying targetable genes that transform into SCLC after the application of TKIs.

The treatment outcomes in patients with transformed SCLC have been reported in only a limited number of studies. Previous studies have shown that many patients underwent cytotoxic chemotherapy following the transformation to SCLC, with the most frequently utilized regimen being SCLC-based chemotherapy (comprising etoposide plus platinum or irinotecan and platinum) [11, 96]. The study results of MarCoux et al. showed that after conversion, the response rates of platinum plus etoposide and taxane were 54% and 50% respectively, and the PFS was 3.4 months and 2.7 months respectively, while patients receiving single or combined ICIs treatment failed to get response [11].In another retrospective study, the results indicated that after conversion, the mPFS in the group receiving platinum plus etoposide was 3.5 months [97], similar to the findings reported by MarCoux. In our study, platinum plus etoposide chemotherapy was the most common first-line treatment option after transformation to SCLC. Our study, involving 46 patients who received platinum plus etoposide after histological transformation to SCLC, revealed a median PFS of 3.0 months, like previous findings, but shorter than the median PFS of 5.0 months in classic first-line SCLC treatment [98]. In addition, our analysis results showed that in the platinum + etoposide + TKI regimen, the mPFS was 3.2 months, which was not significantly different from the platinum + etoposide group (3.0 months vs. 3.2 months; P = 0.35). Research conducted by Wang et al. demonstrated that, compared with chemotherapy without EGFR-TKIs, a subgroup analysis of 16 patients treated with chemotherapy combined with EGFR-TKIs versus eight patients without chemotherapy showed that chemotherapy using EGFR-TKIs improved mPFS (5.2 months vs. 3.0 months, P = 0.014) [99]. Nonetheless, there was no statistically significant disparity in OS between patients who underwent chemotherapy with or without EGFR-TKIs (14.8 months vs. 13.0 months; P = 0.474). Similarly, in our study, we found no significant difference in OS between patients treated with first-line platinum plus etoposide chemotherapy with or without TKI after histological transformation to SCLC (42.7 months vs. 59.3 months; P = 0.096).

In another study, the results indicated that the ORR and disease control rate (DCR) of ICIs treatment, including anti-PD-1/PD-L1 monotherapy or pemetrexed platinum combined with ant-PD-1/PD-L1 treatment, were 0% and 17%, respectively, with a mPFS of 2.0 months [96]. In our study, the survival outcomes from 13 patients who received ICIs alone or in combination after transformation to SCLC showed an ORR of 33.3% and an mPFS of 3.0 months (95%CI, 1.58 to 4.42 months). Additionally, six patients with available survival outcomes received EP plus anti-PD-1/PD-L1, showing a mPFS of 6.0 months (95% CI, 1.38 to 10.62 months). For three patients with information on anti-PD-1/PD-L1 treatment, the mPFS was 2.0 months (95%CI, 1.20 to 2.80 months). In our study, one patient received EC plus Anlotinib (multi-target TKI) plus radiotherapy as a first-line therapy after transformation to SCLC, with a PFS of 4.7 months. Previous research results suggested a mPFS of 6.5 months for the group receiving anlotinib after SCLC transformation [97]. An analysis conducted retrospectively by Zhan et al. indicated that PD-L1 inhibitors combined with chemotherapy ± bevacizumab might be a potentially safe option for patients with SCLC transformation [100].

Nicolas et al. showed that median OS since the time of SCLC was 10.9 months (95% CI, 8.0 to 13.7 months) [11]. In our study, the calculated mOS post-SCLC diagnosis was determined to be 11.0 months, which was similar to previous study. And after transformation to SCLC, the mOS was 5.7 months in the platinum-etoposide group, shorter than the median OS of 10 months in classic SCLC [98]. In addition, in our research, subgroup analysis using ICIs after conversion to SCLC revealed that the mOS after conversion to SCLC was 18.0 months (95% CI, 10.34 to 25.66 months), and the mOS from the diagnosis to death was 48.0 months (95% CI, 8.40 to 87.54 months). Considering the small sample size of the subgroup analysis, the results may be affected, and large-scale data are needed to confirm our findings. With the application of immunotherapy in primary SCLC, it is very necessary to study the characteristics of the tumor immune microenvironment of transformed SCLC and clarify whether transformed SCLC may can benefit from immunotherapy.

A major limitation of our study is the retrospective collection of patients data from published articles. And the small sample size in subgroup analyses may have caused some results to be underpowered. Moreover, patients had varying baseline gene testing panel ranges, and the known gene testing range was limited. Each panel had different coverage of genes within the testing range, making it challenging to obtain a standardized genomic mutation map before and after transformation. Further exploration will be needed based on large-scale prospective RCT studies in the future. In summary, our comprehensive analysis indicated that NSCLC lineage transformation was a manifestation of TKIs resistance, and the prognosis of transformed SCLC patients was worse.

Conclusion

Limited data are available to recommend the optimal treatment for patients exhibiting lineage transformation to drug-resistant tumors, constituting a distinct clinical subgroup. Irrespective of the presence of oncogene-driven NSCLC, the significance of tissue biopsy and next-generation sequencing (NGS) cannot be overstated, especially as the disease advances. It is crucial to explore new therapeutic strategies for transformed SCLC with predictive biology. Platinum-etoposide is a commonly used treatment with a high response rate and should be considered as the first-line treatment for transformed SCLC. In addition, platinum-etoposide-based combination therapy may be a potential treatment strategy to replace chemotherapy alone. The exact mechanism by which TKIs treatment leads to histological transformation to SCLC remains to be elucidated. Understanding the specific molecular characteristics and signaling pathways of transformed tumors will help find new treatments strategies.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.

    Article  PubMed  Google Scholar 

  2. Pikor LA, Ramnarine VR, Lam S, Lam WL. Genetic alterations defining NSCLC subtypes and their therapeutic implications. Lung Cancer. 2013;82:179–89.

    Article  PubMed  Google Scholar 

  3. Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer. 2019;121:725–37.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Soda M, Choi YL, Enomoto M, Takada S, Yamashita Y, Ishikawa S, Fujiwara S, Watanabe H, Kurashina K, Hatanaka H, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561–6.

    Article  CAS  PubMed  Google Scholar 

  5. Shaw AT, Yeap BY, Mino-Kenudson M, Digumarthy SR, Costa DB, Heist RS, Solomon B, Stubbs H, Admane S, McDermott U, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. J Clin Oncol. 2009;27:4247–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Quintanal-Villalonga A, Chan JM, Yu HA, Pe’er D, Sawyers CL, Sen T, Rudin CM. Publisher Correction: Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 2020;17:382.

    Article  PubMed  Google Scholar 

  7. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, Bergethon K, Shaw AT, Gettinger S, Cosper AK, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3:75ra26.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, Kris MG, Miller VA, Ladanyi M, Riely GJ. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lin JJ, Langenbucher A, Gupta P, Yoda S, Fetter IJ, Rooney M, Do A, Kem M, Chang KP, Oh AY, et al. Small cell transformation of ROS1 fusion-positive lung cancer resistant to ROS1 inhibition. NPJ Precis Oncol. 2020;4:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tokaca N, Wotherspoon A, Nicholson AG, Fotiadis N, Thompson L, Popat S. Lack of response to nivolumab in a patient with EGFR-mutant non-small cell lung cancer adenocarcinoma sub-type transformed to small cell lung cancer. Lung Cancer. 2017;111:65–8.

    Article  PubMed  Google Scholar 

  11. Marcoux N, Gettinger SN, O’Kane G, Arbour KC, Neal JW, Husain H, Evans TL, Brahmer JR, Muzikansky A, Bonomi PD, 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.

    Article  CAS  PubMed  Google Scholar 

  12. 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.

    Article  CAS  PubMed  Google Scholar 

  13. Zakowski MF, Ladanyi M, Kris MG. Memorial Sloan-Kettering Cancer Center Lung Cancer OncoGenome G: EGFR mutations in small-cell lung cancers in patients who have never smoked. N Engl J Med. 2006;355:213–5.

    Article  CAS  PubMed  Google Scholar 

  14. Quintanal-Villalonga A, Chan JM, Yu HA, Pe’er D, Sawyers CL, Sen T, Rudin CM. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 2020;17:360–71.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, et al. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol. 2015;10:1243–60.

    Article  PubMed  Google Scholar 

  16. Cha YJ, Cho BC, Kim HR, Lee HJ, Shim HS. A Case of ALK-Rearranged Adenocarcinoma with Small Cell Carcinoma-Like Transformation and Resistance to Crizotinib. J Thorac Oncol. 2016;11:e55–8.

    Article  PubMed  Google Scholar 

  17. Xu Y, Huang Z, Gong L, Fan Y. A case of resistance to tyrosine kinase inhibitor therapy: small cell carcinoma transformation concomitant with plasma-genotyped T790M positivity. Anticancer Drugs. 2017;28:1056–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nishikawa S, Tambo Y, Ninomiya H, Oguri T, Kawashima Y, Takano N, Kitazono S, Ohyanagi F, Horiike A, Yanagitani N, et al. A case treated with nivolumab after small cell lung cancer transformation of mutant EGFR non-small cell lung cancer. Ann Oncol. 2016;27:2300–2.

    Article  CAS  PubMed  Google Scholar 

  19. Yamagata A, Yokoyama T, Fukuda Y, Ishida T. Alectinib re-challenge in small cell lung cancer transformation after chemotherapy failure in a patient with ALK-positive lung cancer: A case report. Respir Med Case Rep. 2021;33:101440.

    PubMed  PubMed Central  Google Scholar 

  20. Chowaniecova G, Berzinec P, Kosturiakova G, Plank L, Farkasova A, Sekeresova M, Juskanic D, Ondrus D. An EGFR-mutant lung adenocarcinoma that transformed into small-cell lung cancer. A case report. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2022;166:451–4.

    Article  PubMed  Google Scholar 

  21. Zhai X, Liu J, Liang Z, Li Z, Liu Y, Huang L, Wang W, Luo F. Case Report: Re-Sensitization to Gefitinib in Lung Adenocarcinoma Harboring EGFR Mutation and High PD-L1 Expression After Immunotherapy Resistance, Which Finally Transform Into Small Cell Carcinoma. Front Oncol. 2021;11:661034.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Li J, Wei B, Feng J, Wu X, Chang Y, Wang Y, Yang X, Zhang H, Han S, Zhang C, et al. Case report: TP53 and RB1 loss may facilitate the transformation from lung adenocarcinoma to small cell lung cancer by expressing neuroendocrine markers. Front Endocrinol (Lausanne). 2022;13:1006480.

    Article  PubMed  Google Scholar 

  23. Fang G, Liu W, Shang Y, Huo R, Shi X, Wang Y, Li J. Characterization of non-small cell lung cancer transforming to small cell lung cancer and its response to EGFR-TKI: a case report. Ann Transl Med. 2022;10:115.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Ou SI, Lee TK, Young L, Fernandez Rocha MY, Pavlick D, Schrock AB, Zhu VW, Milliken J, Ali SM, Gitlitz BJ. Dual occurrence of ALK G1202R solvent front mutation and small cell lung cancer transformation as resistance mechanisms to second generation ALK inhibitors without prior exposure to crizotinib. Pitfall of solely relying on liquid re-biopsy?. Lung Cancer. 2017;106:110–4.

    Article  PubMed  Google Scholar 

  25. Takegawa N, Hayashi H, Iizuka N, Takahama T, Ueda H, Tanaka K, Takeda M, Nakagawa K. Transformation of ALK rearrangement-positive adenocarcinoma to small-cell lung cancer in association with acquired resistance to alectinib. Ann Oncol. 2016;27:953–5.

    Article  CAS  PubMed  Google Scholar 

  26. 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.

    Article  PubMed  Google Scholar 

  27. Miyamoto S, Ikushima S, Ono R, Awano N, Kondo K, Furuhata Y, Fukumoto K, Kumasaka T. Transformation to small-cell lung cancer as a mechanism of acquired resistance to crizotinib and alectinib. Jpn J Clin Oncol. 2016;46:170–3.

    PubMed  Google Scholar 

  28. Caumont C, Veillon R, Gros A, Laharanne E, Begueret H, Merlio JP. Neuroendocrine phenotype as an acquired resistance mechanism in ALK-rearranged lung adenocarcinoma. Lung Cancer. 2016;92:15–8.

    Article  PubMed  Google Scholar 

  29. Li YC. Durable response to durvalumab-based immunochemotherapy in small-cell lung carcinoma transformation from EGFR-mutant non-small cell lung cancer: A case report. Thorac Cancer. 2022;13:775–9.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Iijima Y, Hirotsu Y, Mochizuki H, Amemiya K, Oyama T, Uchida Y, Kobayashi Y, Tsutsui T, Kakizaki Y, Miyashita Y, Omata M. Dynamic Changes and Drug-Induced Selection of Resistant Clones in a Patient With EGFR-Mutated Adenocarcinoma That Acquired T790M Mutation and Transformed to Small-Cell Lung Cancer. Clin Lung Cancer. 2018;19:e843–7.

    Article  PubMed  Google Scholar 

  31. Levacq D, D’Haene N, de Wind R, Remmelink M, Berghmans T. Histological transformation of ALK rearranged adenocarcinoma into small cell lung cancer: A new mechanism of resistance to ALK inhibitors. Lung Cancer. 2016;102:38–41.

    Article  PubMed  Google Scholar 

  32. Sonoda T, Nishikawa S, Sakakibara R, Saiki M, Ariyasu R, Koyama J, Kitazono S, Yanagitani N, Horiike A, Ohyanagi F, et al. EGFR T790M mutation after chemotherapy for small cell lung cancer transformation of EGFR-positive non-small cell lung cancer. Respir Med Case Rep. 2018;24:19–21.

    PubMed  PubMed Central  Google Scholar 

  33. Digiacomo N, De Pas T, Rossi G, Bossi P, Stucchi E, Conforti F, Cocorocchio E, Laszlo D, Pala L, Zattarin E, Catania C. Exon-18-EGFR Mutated Transformed Small-Cell Lung Cancer: A Case Report and Literature Review. Curr Oncol. 2023;30:3494–9.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Majeed U, Li S, Seegobin K, Nassar A, Manochakian R, Zhao Y, Lou Y. First Report of Management of Sequential Small Cell Transformation and ALK I1171T Mutation as Resistance Mechanisms in a Patient With ALK-EML4 Fused Non-Small Cell Lung Adenocarcinoma With a Novel Combination of Temozolomide and Lorlatinib: A Case Report. JTO Clin Res Rep. 2023;4: 100536.

    PubMed  PubMed Central  Google Scholar 

  35. Zhu YC, Liao XH, Wang WX, Xu CW, Zhuang W, Zhong LH, Du KQ, Chen YP, Chen G, Fang MY. Patients harboring ALK rearrangement adenocarcinoma after acquired resistance to crizotinib and transformation to small-cell lung cancer: a case report. Onco Targets Ther. 2017;10:3187–92.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Oya Y, Yoshida T, Uemura T, Murakami Y, Inaba Y, Hida T. Serum ProGRP and NSE levels predicting small cell lung cancer transformation in a patient with ALK rearrangement-positive non-small cell lung cancer: A case report. Oncol Lett. 2018;16:4219–22.

    PubMed  PubMed Central  Google Scholar 

  37. Furugen M, Uechi K, Hirai J, Aoyama H, Saio M, Yoshimi N, Kinjo T, Miyagi K, Haranaga S, Higa F, et al. An Autopsy Case of Two Distinct, Acquired Drug Resistance Mechanisms in Epidermal Growth Factor Receptor-mutant Lung Adenocarcinoma: Small Cell Carcinoma Transformation and Epidermal Growth Factor Receptor T790M Mutation. Intern Med. 2015;54:2491–6.

    Article  CAS  PubMed  Google Scholar 

  38. Liu J, Xia L, Peng Y, Huang YS, Yang ZZ. Gastric metastasis and transformation of primary lung adenocarcinoma to small cell cancer after acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors: A case report. Medicine (Baltimore). 2021;100: e27289.

    Article  PubMed  Google Scholar 

  39. Ninomaru T, Hata A, Hara S, Komatsu M. Heterogeneity or transformation? A whack-a-mole case of EGFR-mutant lung adenocarcinoma and small cell carcinoma: A case report. Thorac Cancer. 2022;13:2394–7.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Santoni-Rugiu E, Grauslund M, Melchior LC, Costa JC, Sorensen JB, Urbanska EM. Heterogeneous resistance mechanisms in an EGFR exon 19-mutated non-small cell lung cancer patient treated with erlotinib: Persistent FGFR3-mutation, localized transformation to EGFR-mutated SCLC, and acquired T790M EGFR-mutation. Lung Cancer. 2017;113:14–7.

    Article  PubMed  Google Scholar 

  41. Yao Y, Zhu Z, Wu Y, Chai Y. Histologic transformation from adenocarcinoma to both small cell lung cancer and squamous cell carcinoma after treatment with gefitinib: A case report. Medicine (Baltimore). 2018;97: e0650.

    Article  PubMed  Google Scholar 

  42. Fiore M, Trecca P, Perrone G, Amato M, Righi D, Trodella L, D’Angelillo RM, Ramella S. Histologic transformation to small-cell lung cancer following gefitinib and radiotherapy in a patient with pulmonary adenocarcinoma. Tumori. 2019;105:NP12–6.

    Article  PubMed  Google Scholar 

  43. Kim WJ, Kim S, Choi H, Chang J, Shin HJ, Park CK, Oh IJ, Kim KS, Kim YC, Choi YD. Histological transformation from non-small cell to small cell lung carcinoma after treatment with epidermal growth factor receptor-tyrosine kinase inhibitor. Thorac Cancer. 2015;6:800–4.

    Article  PubMed  Google Scholar 

  44. Haruta Inoue Y, Kataoka Y, Nikaido J, Nakajima N. Histological transformation from non-small cell to small cell lung carcinoma. BMJ Case Rep. 2016:2016.bcr2016216232.

  45. Hui M, Uppin SG, Stalin BJ, Sadashivudu G. Histological transformation of adenocarcinoma to small cell carcinoma lung as a rare mechanism of resistance to epidermal growth factor receptor-tyrosine kinase inhibitors: Report of a case with review of literature. Lung India. 2018;35:160–3.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Ren X, Cai X, Li J, Zhang X, Yu J, Song X, Zhang H, Song X. Histological transformation of lung adenocarcinoma to small cell lung cancer with mutant C797S conferring acquired resistance to osimertinib. J Int Med Res. 2020;48:300060520927918.

    Article  CAS  PubMed  Google Scholar 

  47. Liu H, Chen LH, Zhang ZH, Wang N, Zhuang SH, Chen H, Du J, Pang LJ, Qi Y. Histomorphological transformation from non-small cell lung carcinoma to small cell lung carcinoma after targeted therapy or immunotherapy: A report of two cases. Front Oncol. 2022;12:1022705.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Pignataro D, Bertaglia V, Bironzo P, Olmetto E, Pisano C, Napoli VM, Mantovani C, Righi L, Novello S. Oligoprogressive Disease With SCLC Transformation in EGFR-Mutated NSCLC: How Biology Knowledge Can Change the Game Rules. J Thorac Oncol. 2020;15:e170–2.

    Article  CAS  PubMed  Google Scholar 

  49. Kato Y, Tanaka Y, Hino M, Gemma A. ProGRP as early predictive marker of non-small-cell lung cancer to small-cell lung cancer transformation after EGFR-TKI treatment. Respir Med Case Rep. 2019;27: 100837.

    PubMed  PubMed Central  Google Scholar 

  50. Zhang Y, Li XY, Tang Y, Xu Y, Guo WH, Li YC, Liu XK, Huang CY, Wang YS, Wei YQ. Rapid increase of serum neuron specific enolase level and tachyphylaxis of EGFR-tyrosine kinase inhibitor indicate small cell lung cancer transformation from EGFR positive lung adenocarcinoma? Lung Cancer. 2013;81:302–5.

    Article  PubMed  Google Scholar 

  51. Wang X, Liang J, Li L, Pan Z, Wang L. Reuse of osimertinib after small cell lung cancer transformation in lung adenocarcinoma with de-novo epidermal growth factor receptor T790M mutation: case report. Anticancer Drugs. 2023;34:306–10.

    Article  CAS  PubMed  Google Scholar 

  52. Fallet V, Ruppert AM, Poulot V, Lacave R, Belmont L, Antoine M, Cadranel J, Wislez M, Lavole A. Secondary resistance to erlotinib: acquired T790M mutation and small-cell lung cancer transformation in the same patient. J Thorac Oncol. 2012;7:1061–3.

    Article  CAS  PubMed  Google Scholar 

  53. Wahab A, Kesari K, Chaudhary S, Khan M, Khan H, Smith S, Boumber Y. Sequential occurrence of small cell and non-small lung cancer in a male patient: Is it a transformation? Cancer Biol Ther. 2017;18:940–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hong E, Chen XE, Mao J, Zhou JJ, Chen L, Xu JY, Tao W. Sequential occurrence of T790M mutation and small cell lung cancer transformation in EGFR-positive lung adenocarcinoma: A case report. World J Clin Cases. 2022;10:2836–43.

    Article  PubMed  PubMed Central  Google Scholar 

  55. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Chen X, Li D, Miao K, Shou T, Zhang W. Small cell lung cancer transformation after EGFR-TKIs treatment in lung adenocarcinoma: A case report and literatures review. Medicine (Baltimore). 2023;102: e32697.

    Article  PubMed  Google Scholar 

  57. Yang MH, Yu J, Cai CL, Li W. Small cell lung cancer transformation and tumor heterogeneity after sequential targeted therapy and immunotherapy in EGFR-mutant non-small cell lung cancer: A case report. Front Oncol. 2022;12:1029282.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Leonetti A, Minari R, Mazzaschi G, Gnetti L, La Monica S, Alfieri R, Campanini N, Verze M, Olivani A, Ventura L, Tiseo M. Small Cell Lung Cancer Transformation as a Resistance Mechanism to Osimertinib in Epidermal Growth Factor Receptor-Mutated Lung Adenocarcinoma: Case Report and Literature Review. Front Oncol. 2021;11: 642190.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Liu Y. Small cell lung cancer transformation from EGFR-mutated lung adenocarcinoma: A case report and literatures review. Cancer Biol Ther. 2018;19:445–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Shee-Chai C, Liam CK, Mun KS. Small Cell Transformation and T790M Mutation as Coresistance Mechanisms for First-line Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor (TKI) Therapy Failure. J Thorac Oncol. 2017;12:e171–3.

    Article  PubMed  Google Scholar 

  61. Wu CH, Su PL, Hsu CW, Chu CY, Lin CC. Small cell transformation in crizotinib-resistant ROS1-rearranged non-small cell lung cancer with retention of ROS1 fusion: A case report. Thorac Cancer. 2021;12:3068–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Gazeu A, Aubert M, Pissaloux D, Lantuejoul S, Perol M, Ikhlef N, Bouhamama A, Franceschi T, Swalduz A. Small-Cell Lung Cancer Transformation as a Mechanism of Resistance to Pralsetinib in RET-Rearranged Lung Adenocarcinoma: A Case Report. Clin Lung Cancer. 2023;24:72–5.

    Article  CAS  PubMed  Google Scholar 

  64. Jiang Y, Shou L, Guo Q, Bao Y, Xu X, An S, Lu J. Small-cell lung cancer transformation from EGFR-mutant adenocarcinoma after EGFR-TKIs resistance: A case report. Medicine (Baltimore). 2021;100: e26911.

    Article  PubMed  Google Scholar 

  65. Jiang SY, Zhao J, Wang MZ, Huo Z, Zhang J, Zhong W, Xu Y. Small-Cell Lung Cancer Transformation in Patients With Pulmonary Adenocarcinoma: A Case Report and Review of Literature. Medicine (Baltimore). 2016;95: e2752.

    Article  PubMed  Google Scholar 

  66. Otoshi R, Sekine A, Okudela K, Asaoka M, Sato Y, Ikeda S, Baba T, Komatsu S, Hagiwara E, Ogura T. Small-cell lung carcinoma transformation of lung adenocarcinoma diagnosed by pericardial effusion: A case report. Mol Clin Oncol. 2020;13:129–32.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Nishioka N, Yamada T, Harita S, Hirai S, Katayama Y, Nakano T, Okura N, Tamiya N, Kaneko Y, Uchino J, Takayama K. Successful sequential treatment of refractory tumors caused by small cell carcinoma transformation and EGFR-T790M mutation diagnosed by repeated genetic testing in a patient with lung adenocarcinoma harboring epidermal growth factor receptor mutations: A case report. Respir Med Case Rep. 2018;25:261–3.

    PubMed  PubMed Central  Google Scholar 

  68. Xia G, Huang J, Ni J, Song M, Zhang J, Hofman P, Christopoulos P, Grenda A, Huang M. Transformation of ALK-positive NSCLC to SCLC after alectinib resistance and response to combined atezolizumab: a case report. Transl Lung Cancer Res. 2023;12:637–46.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Yang Z, Lin Y, Wang H. Transformation of non-small cell lung cancer into small cell lung cancer in a patient with advanced lung cancer: a case report. J Int Med Res. 2021;49:3000605211035005.

    Article  PubMed  Google Scholar 

  70. Hao L, Chen H, Wang L, Zhou H, Zhang Z, Han J, Hou J, Zhu Y, Zhang H, Wang Q. Transformation or tumor heterogeneity: Mutations in EGFR, SOX2, TP53, and RB1 persist in the histological rapid conversion from lung adenocarcinoma to small-cell lung cancer. Thorac Cancer. 2023;14:1036–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. 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.

    Article  CAS  PubMed  Google Scholar 

  72. Coleman N, Wotherspoon A, Yousaf N, Popat S. Transformation to neuroendocrine carcinoma as a resistance mechanism to lorlatinib. Lung Cancer. 2019;134:117–20.

    Article  PubMed  Google Scholar 

  73. Hwang KE, Jung JW, Oh SJ, Park MJ, Shon YJ, Choi KH, Jeong ET, Kim HR. Transformation to small cell lung cancer as an acquired resistance mechanism in EGFR-mutant lung adenocarcinoma: a case report of complete response to etoposide and cisplatin. Tumori. 2015;101:e96-98.

    Article  CAS  PubMed  Google Scholar 

  74. Li L, Wang H, Li C, Wang Z, Zhang P, Yan X. Transformation to small-cell carcinoma as an acquired resistance mechanism to AZD9291: A case report. Oncotarget. 2017;8:18609–14.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Watanabe S, Sone T, Matsui T, Yamamura K, Tani M, Okazaki A, Kurokawa K, Tambo Y, Takato H, Ohkura N, 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.

    Article  PubMed  Google Scholar 

  76. Yu L, Bazhenova L, Gold K, Tran L, Hilburn V, Vu P, Patel SP. Clinicopathologic and molecular characteristics of EGFR-mutant lung adenocarcinomas that transform to small cell lung cancer after TKI therapy. Transl Lung Cancer Res. 2022;11:452–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mc Leer A, Foll M, Brevet M, Antoine M, Novello S, Mondet J, Cadranel J, Girard N, Giaj Levra M, Demontrond P, 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.

    Article  CAS  PubMed  Google Scholar 

  78. Ding J, Leng Z, Gu H, Jing X, Song Y. Etoposide/platinum plus anlotinib for patients with transformed small-cell lung cancer from EGFR-mutant lung adenocarcinoma after EGFR-TKI resistance: a retrospective and observational study. Front Oncol. 2023;13:1153131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Woo CG, Son SM, Lee HC, Han HS, Lee KH, Kim D, Kim EG, Lee OJ. Histologic Changes in Non-Small Cell Lung Cancer under Various Treatments: A Comparison of Histology and Mutation Status in Serial Samples. Cancer Res Treat. 2022;54:737–43.

    Article  CAS  PubMed  Google Scholar 

  80. 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.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Chu X, Xu Y, Li Y, Zhou Y, Chu L, Yang X, Ni J, Li Y, Guo T, Zheng Z, et al. Neuroendocrine transformation from EGFR/ALK-wild type or TKI-naive non-small cell lung cancer: An under-recognized phenomenon. Lung Cancer. 2022;169:22–30.

    Article  CAS  PubMed  Google Scholar 

  82. Editorial O. Erratum to NOTCH alteration in EGFR-mutated lung adenocarcinoma leads to histological small-cell carcinoma transformation under EGFR-TKI treatment. Transl Lung Cancer Res. 2022;11:2358–9.

    Article  Google Scholar 

  83. Zhou Y, Bai H, Xia J, Xu WY, Cheng L, Xiong L. Novel ETV1 mutation in small cell lung cancer transformation resistant to EGFR tyrosine kinase inhibitors. Ann Transl Med. 2021;9:1150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Xie T, Li Y, Ying J, Cai W, Li J, Lee KY, Ricciuti B, Pacheco J, Xing P. Whole exome sequencing (WES) analysis of transformed small cell lung cancer (SCLC) from lung adenocarcinoma (LUAD). Transl Lung Cancer Res. 2020;9:2428–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Mambetsariev I, Arvanitis L, Fricke J, Pharaon R, Baroz AR, Afkhami M, Koczywas M, Massarelli E, Salgia R. Small Cell Lung Cancer Transformation following Treatment in EGFR-Mutated Non-Small Cell Lung Cancer. J Clin Med. 2022;11(5):1429.

  86. Norkowski E, Ghigna MR, Lacroix L, Le Chevalier T, Fadel E, Dartevelle P, Dorfmuller P. Thomas de Montpreville V: Small-cell carcinoma in the setting of pulmonary adenocarcinoma: new insights in the era of molecular pathology. J Thorac Oncol. 2013;8:1265–71.

    Article  PubMed  Google Scholar 

  87. Yang H, Liu L, Zhou C, Xiong Y, Hu Y, Yang N, Qu J. The clinicopathologic of pulmonary adenocarcinoma transformation to small cell lung cancer. Medicine (Baltimore). 2019;98: e14893.

    Article  CAS  PubMed  Google Scholar 

  88. Lai L, Meng W, Wei J, Zhang X, Tan Z, Lu Y, Hou E. Transformation of NSCLC to SCLC after 1st- and 3rd-generation EGFR-TKI resistance and response to EP regimen and erlotinib: 2 CARE-compliant case reports. Medicine (Baltimore). 2021;100: e25046.

    Article  CAS  PubMed  Google Scholar 

  89. Ahn S, Hwang SH, Han J, Choi YL, Lee SH, Ahn JS, Park K, Ahn MJ, Park WY. Transformation to Small Cell Lung Cancer of Pulmonary Adenocarcinoma: Clinicopathologic Analysis of Six Cases. J Pathol Transl Med. 2016;50:258–63.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Antonia SJ, Uchida J, Cohen S, Cohen MC. Attachment of tumor cells to endothelial monolayers: detection of surface molecules involved in cell-cell binding. Clin Immunol Immunopathol. 1989;53:281–96.

    Article  CAS  PubMed  Google Scholar 

  91. Xu J, Xu L, Wang B, Kong W, Chen Y, Yu Z. Outcomes in Patients With Lung Adenocarcinoma With Transformation to Small Cell Lung Cancer After EGFR Tyrosine Kinase Inhibitors Resistance: A Systematic Review and Pooled Analysis. Front Oncol. 2021;11:766148.

    Article  CAS  PubMed  Google Scholar 

  92. Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Primers. 2021;7:3.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Li Y, Xie T, Wang S, Yang L, Hao X, Wang Y, Hu X, Wang L, Li J, Ying J, Xing P. Mechanism exploration and model construction for small cell transformation in EGFR-mutant lung adenocarcinomas. Signal Transduct Target Ther. 2024;9:261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Facchinetti F, Lacroix L, Mezquita L, Scoazec JY, Loriot Y, Tselikas L, Gazzah A, Rouleau E, Adam J, Michiels S, et al. Molecular mechanisms of resistance to BRAF and MEK inhibitors in BRAF(V600E) non-small cell lung cancer. Eur J Cancer. 2020;132:211–23.

    Article  CAS  PubMed  Google Scholar 

  95. Lin JJ, Liu SV, McCoach CE, Zhu VW, Tan AC, Yoda S, Peterson J, Do A, Prutisto-Chang K, Dagogo-Jack I, et al. Mechanisms of resistance to selective RET tyrosine kinase inhibitors in RET fusion-positive non-small-cell lung cancer. Ann Oncol. 2020;31:1725–33.

    Article  CAS  PubMed  Google Scholar 

  96. Fujimoto D, Akamatsu H, Morimoto T, Wakuda K, Sato Y, Kawa Y, Yokoyama T, Tamiya M, Hiraoka R, Shingu N, et al. Histologic transformation of epidermal growth factor receptor-mutated lung cancer. Eur J Cancer. 2022;166:41–50.

    Article  CAS  PubMed  Google Scholar 

  97. Wang W, Xu C, Chen H, Jia J, Wang L, Feng H, Wang H, Song Z, Yang N, Zhang Y. 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.

    Article  CAS  PubMed  Google Scholar 

  98. Farago AF, Keane FK. Current standards for clinical management of small cell lung cancer. Transl Lung Cancer Res. 2018;7:69–79.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Wang S, Xie T, Hao X, Wang Y, Hu X, Wang L, Li Y, Li J, Xing P. Comprehensive analysis of treatment modes and clinical outcomes of small cell lung cancer transformed from epidermal growth factor receptor mutant lung adenocarcinoma. Thorac Cancer. 2021;12:2585–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Zhang CY, Sun H, Su JW, Chen YQ, Zhang SL, Zheng MY, Li YF, Huang J, Zhang C, Tai ZX, et al. A potential treatment option for transformed small-cell lung cancer on PD-L1 inhibitor-based combination therapy improved survival. Lung Cancer. 2023;175:68–78.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This research did not receive any specific funding.

Author information

Authors and Affiliations

Authors

Contributions

Shuai Wang: Conceptualization; Data curation; Writing – original draft; Writing – review & editing. Yongsen Wang: Data curation; Investigation; Methodology; Software. Xuan Wu: Conceptualization; Data curation; Formal analysis; Supervision; Writing – review & editing. Li Yang: Conceptualization; Data curation; Formal analysis. Xiaoju Zhang: Conceptualization; Formal analysis; Investigation; Methodology; Project administration; Supervision; Writing – original draft; Writing – review & editing.

Corresponding author

Correspondence to Xiaoju Zhang.

Ethics declarations

Ethics approval and consent to participate

This retrospective study does not involve original research with human or animal subjects. Therefore, no ethical approval was required. All included studies were reviewed for ethical compliance, and approval for each primary study was obtained by the respective ethics committees.

Consent for publication

All authors provided consent to publish this study.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, S., Wang, Y., Wu, X. et al. Patients outcomes in lung adenocarcinoma transforming to small-cell lung cancer after tyrosine kinase inhibitor therapy. World J Surg Onc 23, 34 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03687-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03687-4

Keywords