Skip to main content
Chinese Medical Journal logoLink to Chinese Medical Journal
. 2023 Sep 1;137(2):130–139. doi: 10.1097/CM9.0000000000002693

Molecular classification of small cell lung cancer subtypes: Characteristics, prognostic factors, and clinical translation

Hanfei Guo 1, Wenqian Li 1, Ye Guo 1, Naifei Chen 1, Jiuwei Cui 1,
Editor: Jing Ni1
PMCID: PMC10798698  PMID: 37660289

Abstract

Small cell lung cancer (SCLC) is a highly malignant tumor with a very poor prognosis; therefore, more effective treatments are urgently needed for patients afflicted with the disease. In recent years, emerging molecular classifications based on key transcription factors of SCLC have provided more information on the tumor pathophysiology, metastasis, immune microenvironment, and acquired therapeutic resistance and reflected the intertumoral heterogeneity of the various SCLC phenotypes. Additionally, advances in genomics and single-cell sequencing analysis have further revealed the high intratumoral heterogeneity and plasticity of the disease. Herein, we review and summarize these recent lines of evidence and discuss the possible pathogenesis of SCLC.

Keywords: Small cell lung carcinoma, Molecular classification, Tumor heterogeneity, Drug resistance, Transcription factors, Genomics, Phenotype, Prognosis

Introduction

Lung cancer is one of the most common malignant tumors that affect humans worldwide, with small cell lung cancer (SCLC) accounting for approximately 15% of all lung cancers.[1] Occurring frequently in smokers,[2] SCLC is characterized by its high malignancy, invasiveness, easy recurrence, and metastasis, and has a 5-year survival rate of less than 7%.[3] Platinum-based chemotherapy and radiation therapy remain the first-line treatments for patients with SCLC,[4] and there have been no significant improvements in survival and therapeutic approaches for more than 30 years.[5] Although SCLC is initially highly sensitive to chemotherapy, with a first-response rate of up to 75%–80%, the development of resistance is essentially a universal occurrence.[6]

In recent years, a breakthrough in the treatment of SCLC has come in the form of immunotherapy, especially with immune checkpoint inhibitors. Nivolumab and pembrolizumab, which are anti-programed cell death protein 1 (PD1) monoclonal antibodies, were granted accelerated approval by the US Food and Drug Administration for the third-line treatment of patients with metastatic SCLC.[79] Both atezolizumab and durvalumab are recommended for use in combination with chemotherapy for the first-line treatment of patients with extensive-stage SCLC in National Comprehensive Cancer Network (NCCN) Guidelines for Small Cell Lung Cancer Version 1.2021.[10] However, because patients with SCLC generally have a poor overall response to immune checkpoint inhibitors and the drugs therefore have limited survival benefit, clinical research on new treatments for SCLC is still urgently needed.

As a highly heterogeneous disease, the subtypes of SCLC are defined by their differential expression of transcriptional regulators, such as achaete-scute homolog 1 (ASCL1), neurogenic differentiation factor 1 (NEUROD1), yes-associated protein 1 (YAP1), and POU class 2 homeobox 3 (POU2F3). At the same time, exploration of the intratumoral heterogeneity of SCLC is also expected to reveal its mechanisms of drug resistance and evolution, which may help in the search for new therapeutic targets. In this paper, we review recent advances in the study of the classification and translational research of SCLC as well as the elucidation of its intra- and intertumoral heterogeneity. A better understanding of these biological features may yield new therapeutic targets and combination drug strategies for the treatment of SCLC in the future.

Molecular Classification of SCLC and the Intertumoral Heterogeneity of Its Phenotypes

The first classification model of SCLC, which was proposed in 1986, divides the disease into the following two subtypes based on cytological characteristics: "classical SCLC," which grows in the form of floating spherical aggregates in cell culture, with or without central necrosis; and "variant SCLC", which grows as loosely adherent aggregates or tightly adherent monolayers.[11] In 2015, the World Health Organization attributed SCLC to one of four tumors with a neuroendocrine (NE) morphology, the key features of which were the following: a dense cell arrangement (palisade, chrysanthemum conglomerate, or trabecular), a unique cell morphology (scant cytoplasm, dense core granules, and lack of significant nucleoli), high grade (high proliferation rate and very high Ki-67 proliferation index [50%–100%]), and positive NE markers (including synaptophysin, chromogranin A [CgA], neural cell adhesion molecule 1 [NCAM1], and insulinoma-associated protein 1 [INSM1]).[12] However, a few SCLC specimens were negative for all standard NE markers and therefore could not be correctly classified.

Gene expression studies have revealed that some SCLC cells express high levels of CgA, gastrin-releasing peptide (GRP), ASCL1, and protein delta homolog 1 (DLK1) (thought to represent tumors with low NOTCH signaling pathway activity) and have thus been named "NE SCLCs". By contrast, other SCLC cells lack the expression of these NE markers and are therefore called "non-NE SCLCs".[13]

The roles of transcription factors in shaping tumor behavior and regulating the various different NE patterns of SCLCs have recently been elucidated. ASCL1 and NEUROD1 are considered to be key transcription factors affecting the development and maturation of pulmonary NE cells.[14,15] The ASCL1 gene, which is expressed in approximately 75% of SCLCs, controls several crucial cellular mechanisms, including cell growth and survival.[16] SCLC cell lines that express high levels of ASCL1 grow as loosely adherent clusters in a "variant" morphology.[17] NEUROD1 is expressed in approximately 24% of human tumor specimens of SCLC.[18] The morphology of NEUROD1-expressing SCLC cells ranges from "classical" to "variant" forms, growing loosely in adherent monolayers.[15] Borromeo et al[15] found that ASCL1 and NEUROD1 distinguished heterogeneity in SCLC with distinct genomic landscapes and gene expression profiles, and the tumors could be divided into ASCL1high, NEUROD1high, and "double-negative SCLC" subtypes (with low levels of both ASCL1 and NEUROD1 expression). The use of clustered regularly interspaced short palindromic repeats (CRISPR) gene editing technology for the screening of important proteins that affect tumor cell growth revealed that the transcription factor POU2F3 was overexpressed in only a few tumor specimens of SCLC that expressed NE markers at a low level. POU2F3 was also demonstrated to be a major regulator of tuft cells, a variant of non-NE SCLC cells.[19]

The MD Anderson Cancer Research Center in the United States recognizes four biologically distinct clusters among 81 resected SCLC tumor samples and 62 SCLC cell lines. Three of the clusters are defined almost solely by their differential expression of ASCL1 (SCLC-A, 36%), NEUROD1 (SCLC-N, 31%), and POU2F3 (SCLC-P, 16%), and the remaining cluster is defined by the absence of all three (SCLC-inflamed/mesenchymal [SCLC-I/IM], 17%).[20] By studying the expression of ASCL1, NEUROD1, YAP1, and POU2F3 in 81 SCLC tumor samples and 54 SCLC cell lines, the following four subtypes of non-NE SCLC: SCLC-A type (70%), SCLC-N type (11%), SCLC-P type (10%), and SCLC-Y type (2%) were defined.[21] However, in another study of samples from 174 patients with SCLC, the comprehensive immunohistochemical and histopathological analyses revealed that YAP1 was expressed at low levels and primarily in combined SCLC variants, and its expression was not exclusive of other subtypes.[22]

It was shown in a recent study that the level of hes family bHLH transcription factor (HES1) expression could be used to identify a population of long-term tumor-propagating cells (TPCs) of SCLC,[23] which also showed higher levels of NE differentiation than non-TPCs.[24] Wooten et al[25] classified SCLCs into four subtypes based on differential HES1 expression levels and NE, non-NE, and NEv1 marker expression. Three of them corresponded to the known subtypes SCLC-A, SCLC-Y, and SCLC-N, respectively, whereas the fourth was a previously undescribed ASCL1 + NE variant (designated NEv2 or SCLC-A2).[25]

Simpson et al[26] described a biobank of 38 circulating-tumor-cell-derived explant (CDX) models, the transcriptomic analysis of which confirmed three of four previously described subtypes based on ASCL1, NEUROD1, and POU2F3 expression and identified a previously unreported subtype based on another NE transcription factor, atonal BHLH transcription factor 1 (ATOH1). A high prevalence of ATOH1 in these CDX models reflects their potential as a circulating tumor cell source for in vivo clonal selection and expansion. However, according to other studies, ATOH1 is present in only approximately 1% (1/81) of surgically resected SCLC samples.[13,18] At present, there are different opinions on the classification of SCLC, and thus more data are needed to confirm these findings. Figure 1 summarizes the different classifications of SCLC mentioned in the studies to date.

Figure 1.

Figure 1

Molecular subtypes of small cell lung cancer. ASCL: Achaete-scute homolog; ATOH1: Atonal BHLH transcription factor 1; NE: Neuroendocrine; NEUROD1: Neurogenic differentiation factor 1; POU2F3: POU class 2 homeobox 3; SCLC: Small cell lung cancer.

Key Genetic and Molecular Pathways of SCLC

Whole-genome analysis has revealed two key tumor suppressor genes (TP53 and retinoblastoma 1 [RB1]) that are commonly mutated and inactivated in human SCLC cells (>90% and ~65%, respectively).[13,2729] In a study of the molecular characterization of NE carcinoma in a Chinese population, mutations of both TP53 and RB1 occurred in 83% (29/35) of patients with SCLC.[30]

The Hedgehog (HH) signaling pathway regulates the fate and self-renewal of cancer cells.[31] The activation of this signaling pathway has also been detected in SCLC cells.[32] Constitutive activation of the HH signaling molecule Smoothened (Smo) promoted the clonogenicity of human SCLC cells in vitro and the initiation and progression of the disease in mice in vivo.[33] These results suggest that the HH pathway may be a therapeutic target in SCLC.

The transcription factor INSM1 is also a sensitive and specific marker of NE neoplasms and functions as an NE differentiation factor.[34] Because 93% of SCLCs have been found to be positive for INSM1,[35] this transcription factor has emerged as a sensitive and specific biomarker for the disease.[36]

Through the use of in vitro and mouse models, it has been validated that loss of function of the tumor suppressor phosphatase and the tensin homolog deleted on chromosome 10 (PTEN),[37] NOTCH receptors,[38] and the chromatin regulator CREB-binding protein (CREBBP)[39] can also lead to SCLC. The frequent amplification of myelocytomatosis oncogene cellular homolog (MYC) family proto-oncogenes (MYC, MYCL, and MYCN) has also been observed in SCLC.[13,27,40,41] Members of the MYC gene family are associated with high-frequency mutations in epigenetic regulators (e.g., histone lysine acetyltransferases eP300 and CREBBP, and histone methyltransferase KMt2D) and inactivating mutations of NOTCH family genes.[42] The ASCL1high subtype of SCLC is highly associated with the expression of MYCL,[17] whereas the NEUROD1high and POU2F3high subtypes are related to the upregulation of MYC.[19,43,44] Aurora kinase (AURK) is a main regulatory serine/threonine kinase of mitosis and provides a growth advantage in SCLC when overexpression is accompanied by MYC family amplification.[41,45,46]

Epithelial–Mesenchymal Transition and Drug Resistance in SCLC

SCLC cells, which often have a round or wattle-shaped, immature, and small appearance, have weak adhesive properties and reduced E-cadherin expression levels. Moreover, they exhibit morphological characteristics typical of epithelial–mesenchymal transition (EMT), which has been implicated in the tumor cell resistance to treatments.[47,48]

The NOTCH signaling and ASCL1 expression status of SCLC cells may determine their resistance to treatments by modifying the EMT phenotype. The ASCL1 gene drives the expression of many oncogenes, such as SRY-box 2 (SOX2), MYCL, and B-cell lymphoma 2 (BCL-2), and is associated with EMT in SCLC.[15,16] The NOTCH signaling pathway plays an inhibitory role in SCLC tumorigenesis,[49] with the majority of cancerous cells exhibiting low NOTCH pathway activity with a concomitant high expression level of Delta-like 1 homolog (DLK1), a non-canonical NOTCH ligand.[50] NOTCH signaling can enhance cell adhesion and inhibit the EMT process in SCLC cells by inducing the production of E-cadherin. It can also repress ASCL1 expression at both the transcriptional and post-transcriptional levels.[51] Approximately 25% of SCLC tumors have an inactivation mutation of the NOTCH signaling pathway,[13] which has also been associated with EMT. Chemotherapy and radiation therapy can inactivate NOTCH signaling, thereby inhibiting NE differentiation of the SCLC cells and promoting the EMT process.[42]

Neuropeptides Associated with SCLC

SCLC cells have the capacity to produce neuropeptides for promoting their survival and proliferation as well as for communicating with the microenvironment in autocrine, paracrine, and endocrine manners. Both ASCL1-dominant and NEUROD1-dominant SCLC subtypes are associated with an NE markerhigh profile, whereas POU2F3 and other ASCL1/NEUROD1 double-negative subtypes are NE markerlow.[22]

The NE function of SCLC is mainly related to tumor growth and metastasis. It is possible that the migration potential of SCLC cells is inherently related to the striking migratory phenotype of NE cells during lung development.[52] c-Kit and its natural ligand, stem cell growth factor (SCF), are co-expressed in approximately 40–70% of tumor specimens and cell lines of SCLC,[53] where the SCF/c-Kit pathway is functional in an autocrine or a paracrine fashion.[54] SCLC cells also express a high level of thyroid transcription factor-1 (TTF-1), which regulates the expression of the BCL-2 gene family and can partly coordinate with ASCL1 to promote growth of the tumor cells and contribute to their NE and antiapoptotic gene expression.[55] The chemoattractant stromal cell-derived factor-1 (SDF-1) and its receptor C-X-C motif chemokine receptor 4 (CXCR4) are key modulators of cancer development and crosstalk between tumors and their microenvironment.[56] CXCR4 is highly expressed in SCLC cells,[57] where it cooperates with c-Kit to enhance cell motility and metastasis as well as induce the phosphorylation of viability-signaling molecules, such as Akt and p70 S6 kinase.[58] It has been shown that the transcription factor polyomavirus enhancer activator 3 (Pea3) plays a critical role in facilitating the metastasis of SCLC.[59] In an intravenous transplantation model, paracrine signaling between NE and non-NE subclones of SCLC via fibroblast growth factor 2 (FGF2) and mitogen-activated protein kinase (MAPK) caused enhanced expression of Pea3, which then resulted in metastatic dissemination of the NE tumor subclones.[59]

Bombesin (BOM), neuromedin B (NMB), and GRP, which are all neuropeptides produced by SCLC cells, can inhibit the maturation of dendritic cells in a dose-dependent manner. BOM and GRP also inhibit the production of interleukin-12 (IL-12) by dendritic cells and thus reduce their ability to activate T cells.[60] This suggests that the paracrine function may be a mechanism of tumor immune escape and new target for the treatment of SCLC could be discovered upon paracrine function related pathways.

Neural autoantibodies, such as antineuronal nuclear antibody-type 1 (ANNA-1) and N-type voltage-gated calcium channel (VGCC-N) autoantibodies, are commonly found in the serum of patients with SCLC with neurological manifestations,[61,62] reflecting the presence of mutated cancer autoantigens.[63] These patients tend to have a better prognosis than those without neurological complications.[62,6466] The comorbidity of endocrine paraneoplastic syndromes in patients with SCLC suggests the existence of communication between SCLC cells and other cells in the body, which is a phenomenon that has yet to be fully studied.[67]

Tumor Evolution and Intratumoral Heterogeneity of SCLC

The fact that SCLC tumors can be assigned to different molecular subtypes based on their differential expression of transcription factors suggests that they have significant intertumoral heterogeneity. However, some studies have indicated that different molecular subtypes of SCLC may exist in a single tumor, which implies the intratumoral heterogeneity of the disease.[26,68] For example, frequent dual-high expression (19.8%) of ASCL1 and NEUROD1 was found in a small set of formalin-fixed paraffin-embedded samples (N = 81) of human SCLC tissue.[18] In another study with a larger sample set (N = 174), significant co-expression of ASCL1 and NEUROD1 was found in 37% of the specimens, with 22% exhibiting dual-high expression (H-score >50 for both markers).[22] The apparent difference in transcription factor expression levels among different studies may be due to the different types of tissues being analyzed (cell cultures, resection specimens, biopsies, genetically engineered mouse models [GEMMs]), which in turn reflects the difference and variability in tumor compositions. Samples derived from animal and pathological models of SCLC are generally more homogeneous than their native counterparts. For example, CDX models derived from SCLC patient samples and RNA sequencing data were used to explore the co-expression of subtype-defining transcription factors, whereupon fewer than 1% of cells in any of the models were found to express POU2F3; and of these rare POU2F3-positive cells, all exhibited ASCL1 co-expression.[20] Furthermore, this study also showed an ASCL1/NEUROD1 co-expression rate of approximately 10% in one of the models.[20] Overall, although most SCLC tumors express only one of these transcription factors, the expression is not mutually exclusive. It is worth emphasizing that SCLC is not just a heterogeneous tumor, but this heterogeneity should underlie different tumors of different origins and/or pathogenesis.

Some reports have also confirmed the impact that intratumoral heterogeneity may have on plasticity transformation, or subtype switching, as mechanisms of the therapeutic resistance of SCLC.[23] NE SCLCs can transform into non-NE tumors (e.g., by MYC amplification), and these two components are likely to coexist within the same tumor, thus supporting the different susceptibility of these cell compartments to therapy.[68] NE cells are fast growing and prone to metastasis, whereas the non-NE component(s) account for chemoresistance, with the traits often connecting over time, confirming the profoundly heterogeneous nature of these tumors.[41,69]

The development of GEMMs through targeted alteration of the mouse tumor suppressors Rb and transformation-related protein 53 (Trp53), which are also present in almost all human SCLC cells, has provided an invaluable preclinical platform for identifying and characterizing the mechanisms of SCLC tumorigenesis and progression.[13,2730] These GEMMs have a high tumor incidence and are highly similar to human SCLC tumors.[70] One such model has demonstrated the predominance of ASCL1 in early lesions with Trp53/Rb1/Rbl2 deficiency, suggesting a possible hierarchy where SCLC-A is a precursor of SCLC-N.[15,41] In vitro studies with SCLC cell lines have shown that surface marker profiles of the disease can shift upon drug treatment, indicating a phenomenon of tumor evolution under subtype-specific therapeutic selection.[71]

However, the difficulty in obtaining sufficient biopsies that are suitable for pathological diagnosis and subsequent study is a common problem in the field of SCLC research. Tools for the preclinical study of malignant diseases, such as human cancer cell lines and derived xenografts, GEMMs, patient-derived xenografts, or cell-derived xenografts, are expected to provide us with more evidence on the evolutionary history of SCLCs in the future.

Treatment Strategies and Prognostic Biomarkers for Different SCLC Subtypes

Despite the significant intratumoral heterogeneity of SCLC, all patients with this disease are currently treated with first-line platinum-based chemotherapy, radiation therapy, and immunotherapy.[72] Advances in molecular typing and mechanistic research at the genetic level may provide new therapeutic strategies for patients with SCLC.

SCLC-A

SCLC-A, an NE epithelial tumor with an ASCL1high/c-MYClow profile, is targetable by BCL-2 inhibitors (e.g., venetoclax, nativoclax),[55,73] which can block the growth of the tumor and induce its regression in mice bearing the high BCL-2-expressing subtype. Preclinical studies have shown that BCL-2 expression levels may be an effective predictive biomarker of a tumor's sensitivity to treatment.[74,75] However, BCL-2 inhibitor monotherapy has not shown a significant clinical impact.[74] Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein that is co-expressed with BCL-2 in multiple tumor types, including SCLC. Preclinical studies have shown that ROR1 inhibition (KAN0441571C) occurs synergistically with BCL-2 inhibition in SCLC models, thus presenting a novel therapeutic target for the disease.[76] The bromodomain and extra-terminal (BET) proteins have been shown to regulate the expression of key genes in oncogenesis, such as MYC, cyclin D2 (CCND2), and BCL2-like protein 1 (BCL2L1). Co-treatment of SCLC tumors with a BET inhibitor and the BCL-2 inhibitor venetoclax resulted in strong synergetic antitumor effects both in vitro and in vivo.[77] Additionally, anthracyclines (e.g.,doxorubicin) and cyclin-dependent kinase 9 (CDK9) inhibitors (e.g., dinaciclib) were also shown to act synergistically with a BCL-2 inhibitor (e.g., venetoclax) in downregulating myeloid leukemia-1 (MCL-1); therefore, the combinations of venetoclax with doxorubicin or dinaciclib provide yet another effective therapeutic strategy against SCLC growth and proliferation.[78]

Inactivating mutations in members of the NOTCH family and the abnormally high expression of delta-like 3 protein (DLL3), an atypical NOTCH ligand, are also often observed in SCLC-A tumors.[79] Expression of the NOTCH1 receptor in SCLC serves as a clinically important prognostic factor.[80] Rovalpituzumab tesirine (Rova-T), an antibody–drug conjugate that targets DLL3, had shown encouraging single-agent antitumor activity in phase I/II studies on patients with recurrent SCLC and high DLL3 expression, despite the occurrence of serious adverse events.[81,82] However, a subsequent phase III clinical study showed that compared to that with topotecan, Rova-T resulted in a poorer overall survival rate and higher rates of serosal effusions, photosensitivity reactions, and peripheral edema, which led to the discontinuation of patient enrollment.[83] Lysine-specific histone demethylase 1A (LSD1, also known as KDM1A) binds to the NOTCH1 locus, resulting in the suppression of NOTCH1 expression and its downstream signaling events. In SCLC models, LSD1 inhibitors were shown to activate the NOTCH pathway, thereby suppressing ASCL1 expression and the repression of tumorigenesis.[38] However, in a phase I study (NCT02034123), GSK2879552 (a potent selective LSD1 inhibitor) provided poor disease control and a high adverse event rate in patients with SCLC, and the study was subsequently terminated as the risk–benefit profile did not favor its continuation.[84]

In another study, the quantitative secretome analysis of 13 cell lines representing different NE lung cancer subtypes showed that ASCL1 regulates insulin-like growth factor binding protein 5 (IGFBP5) transcription directly by binding to its E-box element.[85] The study also identified IGFBP5 as a secreted marker for ASCL1high SCLC in vitro and in mouse models, rendering it a potential drug target for SCLC.[85] SCLC-A also expresses a high level of Schlafen 11 (SLFN11) in a bimodal manner which has prognostic predictive value for a variety of cancer treatments (etoposide, topotecan, carboplatin, temozolomide/veliparib, and poly-(ADP)-ribose polymerase [PARP] inhibitors).[48,86,87]

SCLC-N

SCLC-N is an NE MYChigh tumor with susceptibility to AURK inhibitors (AURKi).[73,88] The A-type AURK stabilizes the MYC family of oncoproteins, which have long been considered an undruggable target. It has been confirmed that AURKi can potently inhibit the proliferation of high-MYC-expressing SCLC cell lines.[88] In a phase I/II study, single-agent treatment with alisertib, an investigational AURKi, showed an objective response rate of 21% (10/48) in patients with SCLC.[89] In another phase I study, the combination of alisertib with nab-paclitaxel proved to be synergistically active against rapidly proliferative high-grade NE tumors, especially SCLC, and displayed a manageable side-effect profile.[90] In a phase II study, promising preliminary efficacy was seen with combination alisertib/paclitaxel therapy in relapsed or refractory SCLC, with the drug combination resulting in a median progression-free survival period of 3.32 months vs. 2.17 months with paclitaxel alone (hazard ratio [HR] = 0.71, P = 0.038).[91] Moreover, the c-MYC expression level and mutations in cell cycle regulators may be potential predictive biomarkers of alisertib efficacy.[91] These data support the important need for further clinical assessments of alisertib in patients with SCLC. Another potential strategy against SCLC-N involves Seneca Valley Virus (SVV-001), an NE cancer-selective oncolytic picornavirus that has shown selective efficacy in primary heterotransplant mouse models of NEUROD1high SCLC.[92]

SCLC-P

SCLC-P, a non-NE epithelial tumor, accounts for approximately 7% of all SCLC cases.[22] This tumor subtype is vulnerable to poly (ADP-ribose) polymerase (PARP) inhibitors and anti-metabolites, including anti-folates and nucleoside analogs.[20] The use of small-molecule inhibitors may also be a future direction in the research and development of drugs that specifically target unique dependencies in POU2F3-expressing SCLC cell lines, such as the transcription factors SRY-box transcription factor (SOX) 9 and ASCL2 and the receptor tyrosine kinase insulin-like growth factor 1 receptor (IGF1R).[19] Additionally, the SCLC-P subtype seemed to be a poor prognostic marker for immunotherapy as it does especially poorly relative to the other three subtypes in the subtype-by-subtype analysis comparing survival between atezolizumab vs. placebo in the cohort of IMpower133.[20]

SCLC-Y

SCLC-Y, a non-NE inflamed phenotype that has a better-differentiated tumor histology and an inflamed tumor microenvironment, accounts for 5%–10% of SCLC cases.[93] SCLC-Y cell lines are distinct among the SCLC subtypes, bear transcriptome similarity to that of NSCLC, and are associated with shorter patient survival and increased chemoresistance.[46,94] This subtype is also associated with high expression levels of YAP1, interferon-gamma response genes, T-cell receptor genes, human leukocyte antigen (HLA) genes, and antigen-presenting machinery genes; high T-cell-inflamed gene expression profile scores; and low levels of cancer testis antigens, RB1 mutations, and ASCL1 expression.[93,95] Other characteristics of SCLC-Y cells include their high expression of the ephrin type-A receptor (EPHA2), mesenchymal marker vimentin (VIM), cytoskeleton component and regulator calponin 2 (CNN2), and NOTCH pathway genes.[95] In addition, it has been shown that YAP1 can upregulate programmed death-ligand 1 (PD-L1) expression levels and induce an immunosuppressive tumor microenvironment,[96,97] which suggest that the SCLC-Y subtype may be more likely to benefit from immunotherapy, especially immune checkpoint inhibitors. According to the results of gene expression and in silico studies, SCLC-Y tumors are also likely vulnerable to inhibitors targeting mammalian target of rapamycin (mTOR), polo-like kinase 1 (PLK1), YAP1, and NOTCH1.[51,93,97,98]

SCLC-I

SCLC-I, a non-NE mesenchymal inflamed phenotype of SCLC, is characterized by its high expression of inflammatory markers, immune checkpoint proteins, and stimulator of interferon genes (STING)-related genes, with greater benefit for its suppression being derived from immune checkpoint inhibitors.[30] In the IMpower133 study, patients with SCLC-I tumors, who constituted 18.5% of all participants, showed a significantly higher overall survival rate than those with other SCLC subtypes in the atezolizumab vs. placebo in addition to chemotherapy trial (HR = 0.57; 95% CI: 0.32–0.99).[20,99] However, there is insufficient evidence for the SCLC-I to be a prognostic maker of immunotherapy as the addition of atezolizumab produces more modest gains in median OS in SCLC-A and -N than in SCLC-I.[20] As the most of the clinical trials of immunotherapy were not designed for different subtypes of SCLC, future clinical studies are still needed to confirm whether the molecular classification of SCLC can be used as a prognostic marker for immunotherapy. SCLC-I cells also show a high expression of Bruton's tyrosine kinase (BTK), rendering them potentially sensitive to BTK inhibitors, such as ibrutinb.[20] Table 1 summarizes the gene expression profile and corresponding treatment strategies of different subtypes of SCLC.

Table 1.

Summary of gene expression profile and corresponding treatment strategies of subtypes of SCLC.

Major transcriptional factors Associated subtype and tumor behavioral characteristics of SCLC Related genes Potential therapeutic approaches
ASCL1 SCLC-A; NE tumor, "classic" form, EMT Upregulate SOX2, MYCL,BCL-2, DLK1, INSM1, E-cadherin, DLL3, TTF1
Downregulate NOTCH signaling, MYC
BCL-2 inhibitors (venetoclax, nativoclax); anthracyclines (e.g., doxorubicin); CDK9 inhibitors (e.g., dinaciclib); ROR1 inhibitor (KAN0441571C); BET inhibitor; DLL3 inhibitors (rovalpituzumab tesirine [Rova-T]); LSD1 inhibitors (NCT02034123)
NEUROD1 SCLC-N; NE tumor, "variant" form, growing loosely in adherent monolayers Upregulate MYC, AURK, HES6,INSM1; Downregulate E-cadherin AURK inhibitors, Seneca Valley Virus (SVV-001)
POU2F3 SCLC-P; Non-NE tumor Upregulate MYC, AURK, E-cadherin, SOX9, ASCL2, IGF1R PARP inhibitors, anti-metabolites (including anti-folates and nucleoside analogs), IGF1R inhibitors
YAP1 SCLC-Y; non-NE tumor, associated with increased chemoresistance, high expression of immune suppressive microenvironment-related markers Upregulate YAP1, MYC, interferon-gamma response genes, T-cell receptor genes, HLA genes, AURK, EPHA2, VIM, CNN2, NOTCH signaling, PD-L1, mTOR, PLK1, RB1
Downregulate MYC
Immune checkpoint inhibitors, mTOR inhibitors, PLK1 inhibitors, YAP1 inhibitors
ASCL1/NEUROD1/POU2F3 SCLC-I/IM; Inflamed/mesenchymal,
EMT
Upregulate BTK Immune checkpoint inhibitors, BTKi inhibitors (ibrutinib)

ADP: Adenosine diphosphate; ASCL: Achaete-scute homolog; AURK: Aurora kinase; BCL-2: B cell lymphoma protein-2; BET: Bromodomain and extra-terminal; BTK: Bruton's tyrosine kinase; CDK9: Cyclin-dependent kinase 9; CNN2: Calponin 2; DLK1: Delta-like homolog 1; DLL3: Delta-like 3 protein; EMT: Epithelial–mesenchymal transition; EPHA2: Ephrin type-A receptor 2; HES6: Hes family bHLH transcription factor 6; HLA: Human leukocyte antigen; IGF1R: Insulin-like growth factor 1 receptor; INSM1: Insulinoma-associated protein 1; LSD1: Lysine-specific histone demethylase 1A; mTOR: Mammalian target of rapamycin; MYC: Myelocytomatosis oncogene cellular homolog; NE: Neuroendocrine; NEUROD1: Neurogenic differentiation factor 1; PARP: Poly-(ADP)-ribose polymerase; PD-L1: Programmed death-ligand 1; PLK1: Polo-like kinase 1; POU2F3: POU class 2 homeobox 3; RB1: Retinoblastoma 1; ROR1: Receptor tyrosine kinase-like orphan receptor 1; SCLC: Small cell lung cancer; SOX: SRY-box transcription factor; TTF1: Thyroid transcription factor-1; VIM: Vimentin; YAP1: Yes-associated protein 1.

Summary and Future Prospects

Throughout the progresses made in the field of SCLC research, immunotherapy has not only gradually changed the treatment pattern for this disease but also opened up the possibilities of finding the appropriate patient population, ideal treatment timing, and optimal treatment strategies. With the ongoing study of the disease pathogenesis and the application of new treatment methods, such as immunotherapy, the classification of SCLC is also being continuously updated and integrated with clinical practice. However, the study of molecular subtypes of SCLC at the genetic level belongs to different dimensions with different treatment types (such as surgery, chemotherapy, immunotherapy, etc.), and most clinical trials of immunotherapy have not considered the molecular subtypes of SCLC at the time of design. Whether molecular subtypes of SCLC can be used as a basis for immunotherapy remains to be revealed by more clinical study data. Such advances in knowledge acquisition will lead to better analyses of the biological behavior and clinical characteristics of the various SCLC subtypes, which will more accurately guide their treatment and the evaluation of their prognosis, thereby improving the survival of patients with SCLC in the future.

SCLC is a highly heterogeneous disease. The goal of classifying the various SCLC subtypes is to elucidate the nature of the malignancy. However, any classification model is staged and cannot picture all the characteristics of SCLC. Despite the fact that the molecular-typing classification of SCLC based on ASCL1, NEUROD1, YAP1, and POU2F3 expression levels has reached uniform consensus, some subtypes still cannot be classified. Such classification also poses new problems, and the associations of these subtypes with the tumor stage, metastatic potential, and immune microenvironment remain unknown. The elucidation of these relationships may have prognostic and therapeutic implications and therefore warrant further clinical investigation.

Funding

This work was supported by grants from the Jilin Scientific and Technological Development Program (CN) (No. 20190303146SF) and General Program of National Natural Science Foundation of China (No. 81874052).

Conflicts of interest

None.

Footnotes

How to cite this article: Guo HF, Li WQ, Guo Y, Chen NF, Cui JW. Progression in molecular classification of small cell lung cancer subtypes and research on their characteristics, prognostic factors, and clinical translation. Chin Med J 2024;137:130–139. doi: 10.1097/CM9.0000000000002693

References

  • 1.Cao W, Chen HD, Yu YW, Li N, Chen WQ. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020. Chin Med J 2021;134: 783–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Alexandrov LB Ju YS Haase K Van Loo P Martincorena I Nik-Zainal S, et al. Mutational signatures associated with tobacco smoking in human cancer. Science 2016;354: 618–622. doi: 10.1126/science.aag0299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Govindan R Page N Morgensztern D Read W Tierney R Vlahiotis A, et al. Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: Analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol 2006;24: 4539–4544. doi: 10.1200/jco.2005.04.4859. [DOI] [PubMed] [Google Scholar]
  • 4.Rudin CM Ismaila N Hann CL Malhotra N Movsas B Norris K, et al. Treatment of small-cell lung cancer: American Society of Clinical Oncology Endorsement of the American College of Chest Physicians Guideline. J Clin Oncol 2015;33: 4106–4111. doi: 10.1200/JCO.2015.63.7918. [DOI] [PubMed] [Google Scholar]
  • 5.Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: What we know, what we need to know and the path forward. Nat Rev Cancer 2017;17: 725–737. doi: 10.1038/nrc.2017.87. [DOI] [PubMed] [Google Scholar]
  • 6.Karim SM, Zekri J. Chemotherapy for small cell lung cancer: A comprehensive review. Oncol Rev 2012;6: e4. doi: 10.4081/oncol.2012.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ready N Farago AF de Braud F Atmaca A Hellmann MD Schneider JG, et al. Third-line nivolumab monotherapy in recurrent SCLC: CheckMate 032. J Thorac Oncol 2019;14: 237–244. doi: 10.1016/j.jtho.2018.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ready NE Ott PA Hellmann MD Zugazagoitia J Hann CL de Braud F, et al. Nivolumab monotherapy and nivolumab plus ipilimumab in recurrent small cell lung cancer: Results from the CheckMate 032 randomized cohort. J Thorac Oncol 2020;15: 426–435. doi: 10.1016/j.jtho.2019.10.004. [DOI] [PubMed] [Google Scholar]
  • 9.Chung HC Piha-Paul SA Lopez-Martin J Schellens JHM Kao S Miller WH Jr., et al. Pembrolizumab after two or more lines of previous therapy in patients with recurrent or metastatic SCLC: Results from the KEYNOTE-028 and KEYNOTE-158 studies. J Thorac Oncol 2020;15: 618–627. doi: 10.1016/j.jtho.2019.12.109. [DOI] [PubMed] [Google Scholar]
  • 10.Mathieu L Shah S Pai-Scherf L Larkins E Vallejo J Li X, et al. FDA approval summary: Atezolizumab and durvalumab in combination with platinum-based chemotherapy in extensive stage small cell lung cancer. Oncologist 2021;26: 433–438. doi: 10.1002/onco.13752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Gazdar AF, Carney DN, Nau MM, Minna JD. Characterization of variant subclasses of cell lines derived from small cell lung cancer having distinctive biochemical, morphological, and growth properties. Cancer Res 1985;45: 2924–2930. [PubMed] [Google Scholar]
  • 12.Travis WD Brambilla E Nicholson AG Yatabe Y Austin JHM Beasley MB, 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: 1243–1260. doi: 10.1097/JTO.0000000000000630. [DOI] [PubMed] [Google Scholar]
  • 13.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. doi: 10.1038/nature14664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.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: 852–855. doi: 10.1038/386852a0. [DOI] [PubMed] [Google Scholar]
  • 15.Borromeo MD Savage TK Kollipara RK He M Augustyn A Osborne JK, et al. ASCL1 and NEUROD1 reveal heterogeneity in pulmonary neuroendocrine tumors and regulate distinct genetic programs. Cell Rep 2016;16: 1259–1272. doi: 10.1016/j.celrep.2016.06.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Fujino K Motooka Y Hassan WA Ali Abdalla MO Sato Y Kudoh S, et al. Insulinoma-associated protein 1 is a crucial regulator of neuroendocrine differentiation in lung cancer. Am J Pathol 2015;185: 3164–3177. doi: 10.1016/j.ajpath.2015.08.018. [DOI] [PubMed] [Google Scholar]
  • 17.Kato F Fiorentino FP Alibés A Perucho M Sánchez-Céspedes M Kohno T, et al. MYCL is a target of a BET bromodomain inhibitor, JQ1, on growth suppression efficacy in small cell lung cancer cells. Oncotarget 2016;7: 77378–77388. doi: 10.18632/oncotarget.12671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhang W Girard L Zhang YA Haruki T Papari-Zareei M Stastny V, et al. Small cell lung cancer tumors and preclinical models display heterogeneity of neuroendocrine phenotypes. Transl Lung Cancer Res 2018;7: 32–49. doi: 10.21037/tlcr.2018.02.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Huang YH Klingbeil O He XY Wu XS Arun G Lu B, et al. POU2F3 is a master regulator of a tuft cell-like variant of small cell lung cancer. Genes Dev 2018;32: 915–928. doi: 10.1101/gad.314815.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gay CM Stewart CA Park EM Diao L Groves SM Heeke S, et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell 2021;39: 346–360.e7. doi: 10.1016/j.ccell.2020.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rudin CM Poirier JT Byers LA Dive C Dowlati A George J, et al. Molecular subtypes of small cell lung cancer: A synthesis of human and mouse model data. Nat Rev Cancer 2019;19: 289–297. doi: 10.1038/s41568-019-0133-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Baine MK Hsieh MS Lai WV Egger JV Jungbluth AA Daneshbod Y, et al. SCLC subtypes defined by ASCL1, NEUROD1, POU2F3, and YAP1: A comprehensive immunohistochemical and histopathologic characterization. J Thorac Oncol 2020;15: 1823–1835. doi: 10.1016/j.jtho.2020.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.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: 360–364. doi: 10.1038/nature22323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Jahchan NS Lim JS Bola B Morris K Seitz G Tran KQ, et al. Identification and targeting of long-term tumor-propagating cells in small cell lung cancer. Cell Rep 2016;16: 644–656. doi: 10.1016/j.celrep.2016.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wooten DJ Groves SM Tyson DR Liu Q Lim JS Albert R, et al. Systems-level network modeling of small cell lung cancer subtypes identifies master regulators and destabilizers. PLoS Comput Biol 2019;15: e1007343. doi: 10.1371/journal.pcbi.1007343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Simpson KL Stoney R Frese KK Simms N Rowe W Pearce SP, et al. A biobank of small cell lung cancer CDX models elucidates inter- and intratumoral phenotypic heterogeneity. Nat Cancer 2020;1: 437–451. doi: 10.1038/s43018-020-0046-2. [DOI] [PubMed] [Google Scholar]
  • 27.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: 1104–1110. doi: 10.1038/ng.2396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wistuba II, Gazdar AF, Minna JD. Molecular genetics of small cell lung carcinoma. Semin Oncol 2001;28(2 Suppl 4): 3–13. doi: 10.1053/sonc.2001.25738. [PubMed] [Google Scholar]
  • 29.Cheng DT Mitchell TN Zehir A Shah RH Benayed R Syed A, et al. Memorial sloan kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): A hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 2015;17: 251–264. doi: 10.1016/j.jmoldx.2014.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wu L Cao L Chen L Zhu B Hu X Gen L, et al. Characterization of genomic alterations in Chinese LCNEC and SCLC via comprehensive genomic profiling. J Clin Oncol 2019;37: 1575. doi: 10.1200/JCO.2019.37.15_suppl.1575. [Google Scholar]
  • 31.Szczepny A Rogers S Jayasekara WSN Park K McCloy RA Cochrane CR, et al. The role of canonical and non-canonical Hedgehog signaling in tumor progression in a mouse model of small cell lung cancer. Oncogene 2017;36: 5544–5550. doi: 10.1038/onc.2017.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Vestergaard J Pedersen MW Pedersen N Ensinger C Tümer Z Tommerup N, et al. Hedgehog signaling in small-cell lung cancer: Frequent in vivo but a rare event in vitro. Lung Cancer 2006;52: 281–290. doi: 10.1016/j.lungcan.2005.12.014. [DOI] [PubMed] [Google Scholar]
  • 33.Park KS Martelotto LG Peifer M Sos ML Karnezis AN Mahjoub MR, et al. A crucial requirement for Hedgehog signaling in small cell lung cancer. Nat Med 2011;17: 1504–1508. doi: 10.1038/nm.2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen C, Notkins AL, Lan MS. Insulinoma-associated-1: From neuroendocrine tumor marker to cancer therapeutics. Mol Cancer Res 2019;17: 1597–1604. doi: 10.1158/1541-7786.mcr-19-0286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Doxtader EE, Mukhopadhyay S. Insulinoma-associated protein 1 is a sensitive and specific marker of neuroendocrine lung neoplasms in cytology specimens. Cancer Cytopathol 2018;126: 243–252. doi: 10.1002/cncy.21972. [DOI] [PubMed] [Google Scholar]
  • 36.Mahalakshmi B, Baskaran R, Shanmugavadivu M, Nguyen NT, Velmurugan BK. Insulinoma-associated protein 1 (INSM1): A potential biomarker and therapeutic target for neuroendocrine tumors. Cell Oncol (Dordr) 2020;43: 367–376. doi: 10.1007/s13402-020-00505-9. [DOI] [PubMed] [Google Scholar]
  • 37.Cui M Augert A Rongione M Conkrite K Parazzoli S Nikitin AY, et al. PTEN is a potent suppressor of small cell lung cancer. Mol Cancer Res 2014;12: 654–659. doi: 10.1158/1541-7786.MCR-13-0554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.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. doi: 10.1126/scisignal.aau2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Jia D Augert A Kim DW Eastwood E Wu N Ibrahim AH, et al. Crebbp loss drives small cell lung cancer and increases sensitivity to HDAC inhibition. Cancer Discov 2018;8: 1422–1437. doi: 10.1158/2159-8290.CD-18-0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kim YH Girard L Giacomini CP Wang P Hernandez-boussard T Tibshirani R, et al. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene 2006;25: 130–138. doi: 10.1038/sj.onc.1208997. [DOI] [PubMed] [Google Scholar]
  • 41.Mollaoglu G Guthrie MR Böhm S Brägelmann J Can I Ballieu PM, et al. MYC drives progression of small cell lung cancer to a variant neuroendocrine subtype with vulnerability to Aurora kinase inhibition. Cancer Cell 2017;31: 270–285. doi: 10.1016/j.ccell.2016.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ito T, Kudoh S, Ichimura T, Fujino K, Hassan WAMA, Udaka N. Small cell lung cancer, an epithelial to mesenchymal transition (EMT)-like cancer: Significance of inactive Notch signaling and expression of achaete-scute complex homologue 1. Human Cell 2017;30: 1–10. doi: 10.1007/s13577-016-0149-3. [DOI] [PubMed] [Google Scholar]
  • 43.Carney DN Gazdar AF Bepler G Guccion JG Marangos PJ Moody TW, et al. Establishment and identification of small cell lung cancer cell lines having classic and variant features. Cancer Res 1985;45: 2913–2923. [PubMed] [Google Scholar]
  • 44.Pozo K, Minna JD, Johnson JE. Identifying a missing lineage driver in a subset of lung neuroendocrine tumors. Genes Dev 2018;32: 865–867. doi: 10.1101/gad.316943.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sos ML Dietlein F Peifer M Schöttle J Balke-Want H Müller C, et al. A framework for identification of actionable cancer genome dependencies in small cell lung cancer. Proc Natl Acad Sci U S A 2012;109: 17034–17039. doi: 10.1073/pnas.1207310109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lantuejoul S, Fernandez-Cuesta L, Damiola F, Girard N, McLeer A. New molecular classification of large cell neuroendocrine carcinoma and small cell lung carcinoma with potential therapeutic impacts. Transl Lung Cancer Res 2020;9: 2233–2244. doi: 10.21037/tlcr-20-269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Allison Stewart C Tong P Cardnell RJ Sen T Li L Gay CM, et al. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget 2017;8: 28575–28587. doi: 10.18632/oncotarget.15338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gardner EE Lok BH Schneeberger VE Desmeules P Miles LA Arnold PK, et al. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell 2017;31: 286–299. doi: 10.1016/j.ccell.2017.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kunnimalaiyaan M, Chen H. Tumor suppressor role of Notch-1 signaling in neuroendocrine tumors. Oncologist 2007;12: 535–542. doi: 10.1634/theoncologist.12-5-535. [DOI] [PubMed] [Google Scholar]
  • 50.Augustyn A Borromeo M Wang T Fujimoto J Shao C Dospoy PD, et al. ASCL1 is a lineage oncogene providing therapeutic targets for high-grade neuroendocrine lung cancers. Proc Natl Acad Sci U S A 2014;111: 14788–14793. doi: 10.1073/pnas.1410419111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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: 261–273. doi: 10.1007/s13402-019-00441-3. [DOI] [PubMed] [Google Scholar]
  • 52.Kuo CS, Krasnow MA. Formation of a neurosensory organ by epithelial cell slithering. Cell 2015;163: 394–405. doi: 10.1016/j.cell.2015.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yamanishi Y, Maeda H, Hiyama K, Ishioka S, Yamakido M. Specific growth inhibition of small-cell lung cancer cells by adenovirus vector expressing antisense c-kit transcripts. Jpn J Cancer Res 1996;87: 534–542. doi: 10.1111/j.1349-7006.1996.tb00256.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Krystal GW, Hines SJ, Organ CP. Autocrine growth of small cell lung cancer mediated by coexpression of c-kit and stem cell factor. Cancer Res 1996;56: 370–376. [PubMed] [Google Scholar]
  • 55.Hokari S Tamura Y Kaneda A Katsura A Morikawa M Murai F, et al. Comparative analysis of TTF-1 binding DNA regions in small-cell lung cancer and non-small-cell lung cancer. Mol Oncol 2020;14: 277–293. doi: 10.1002/1878-0261.12608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zhou Y, Cao HB, Li WJ, Zhao L. The CXCL12 (SDF-1)/CXCR4 chemokine axis: Oncogenic properties, molecular targeting, and synthetic and natural product CXCR4 inhibitors for cancer therapy (in Chinese). Chin J Nat Med 2018;16: 801–810. doi: 10.1016/s1875-5364(18)30122-5. [DOI] [PubMed] [Google Scholar]
  • 57.Kijima T Maulik G Ma PC Tibaldi EV Turner RE Rollins B, et al. Regulation of cellular proliferation, cytoskeletal function, and signal transduction through CXCR4 and c-Kit in small cell lung cancer cells. Cancer Res 2002;62: 6304–6311. [PubMed] [Google Scholar]
  • 58.Müller A Homey B Soto H Ge N Catron D Buchanan ME, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 2001;410: 50–56. doi: 10.1038/35065016. [DOI] [PubMed] [Google Scholar]
  • 59.Kwon MC, Proost N, Song JY, Sutherland KD, Zevenhoven J, Berns A. Paracrine signaling between tumor subclones of mouse SCLC: A critical role of ETS transcription factor Pea3 in facilitating metastasis. Genes Dev 2015;29: 1587–1592. doi: 10.1101/gad.262998.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Makarenkova VP Shurin GV Tourkova IL Balkir L Pirtskhalaishvili G Perez L, et al. Lung cancer-derived bombesin-like peptides down-regulate the generation and function of human dendritic cells. J Neuroimmunol 2003;145: 55–67. doi: 10.1016/j.jneuroim.2003.09.009. [DOI] [PubMed] [Google Scholar]
  • 61.Gozzard P Woodhall M Chapman C Nibber A Waters P Vincent A, et al. Paraneoplastic neurologic disorders in small cell lung carcinoma: A prospective study. Neurology 2015;85: 235–239. doi: 10.1212/wnl.0000000000001721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zekeridou A, Majed M, Heliopoulos I, Lennon VA. Paraneoplastic autoimmunity and small-cell lung cancer: Neurological and serological accompaniments. Thorac Cancer 2019;10: 1001–1004. doi: 10.1111/1759-7714.13009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zekeridou A, Griesmann GE, Lennon VA. Mutated cancer autoantigen implicated cause of paraneoplastic myasthenia gravis. Muscle Nerve 2018;58: 600–604. doi: 10.1002/mus.26166. [DOI] [PubMed] [Google Scholar]
  • 64.Iams WT Shiuan E Meador CB Roth M Bordeaux J Vaupel C, et al. Improved prognosis and increased tumor-infiltrating lymphocytes in patients who have SCLC with neurologic paraneoplastic syndromes. J Thorac Oncol 2019;14: 1970–1981. doi: 10.1016/j.jtho.2019.05.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Maddison P, Gozzard P, Grainge MJ, Lang B. Long-term survival in paraneoplastic Lambert-Eaton myasthenic syndrome. Neurology 2017;88: 1334–1339. doi: 10.1212/wnl.0000000000003794. [DOI] [PubMed] [Google Scholar]
  • 66.Gozzard P, Chapman C, Vincent A, Lang B, Maddison P. Novel humoral prognostic markers in small-cell lung carcinoma: A prospective study. PLoS One 2015;10: e0143558. doi: 10.1371/journal.pone.0143558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Vanhees SL, Paridaens R, Vansteenkiste JF. Syndrome of inappropriate antidiuretic hormone associated with chemotherapy-induced tumour lysis in small-cell lung cancer: Case report and literature review. Ann Oncol 2000;11: 1061–1065. doi: 10.1023/a:1008369932384. [DOI] [PubMed] [Google Scholar]
  • 68.Ireland AS Micinski AM Kastner DW Guo B Wait SJ Spainhower KB, et al. MYC drives temporal evolution of small cell lung cancer subtypes by reprogramming neuroendocrine fate. Cancer Cell 2020;38: 60–78.e12. doi: 10.1016/j.ccell.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Stewart CA Gay CM Xi Y Sivajothi S Sivakamasundari V Fujimoto J, et al. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat Cancer 2020;1: 423–436. doi: 10.1038/s43018-019-0020-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dooley AL Winslow MM Chiang DY Banerji S Stransky N Dayton TL, et al. Nuclear factor I/B is an oncogene in small cell lung cancer. Genes Dev 2011;25: 1470–1475. doi: 10.1101/gad.2046711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Udyavar AR Wooten DJ Hoeksema M Bansal M Califano A Estrada L, et al. Novel hybrid phenotype revealed in small cell lung cancer by a transcription factor network model that can explain tumor heterogeneity. Cancer Res 2017;77: 1063–1074. doi: 10.1158/0008-5472.can-16-1467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ferone G, Lee MC, Sage J, Berns A. Cells of origin of lung cancers: Lessons from mouse studies. Genes Dev 2020;34: 1017–1032. doi: 10.1101/gad.338228.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Gay CM Tong P Cardnell RJ Sen T Su X Ma J, et al. Differential sensitivity analysis for resistant malignancies (DISARM) identifies common candidate therapies across platinum-resistant cancers. Clin Cancer Res 2019;25: 346–357. doi: 10.1158/1078-0432.ccr-18-1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rudin CM Hann CL Garon EB Ribeiro de Oliveira M Bonomi PD Camidge DR, et al. Phase II study of single-agent navitoclax (ABT-263) and biomarker correlates in patients with relapsed small cell lung cancer. Clin Cancer Res 2012;18: 3163–3169. doi: 10.1158/1078-0432.ccr-11-3090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lochmann TL Floros KV Naseri M Powell KM Cook W March RJ, et al. Venetoclax is effective in small-cell lung cancers with high BCL-2 expression. Clin Cancer Res 2018;24: 360–369. doi: 10.1158/1078-0432.ccr-17-1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Wang WZ Shilo K Amann JM Shulman A Hojjat-Farsangi M Mellstedt H, et al. Predicting ROR1/BCL2 combination targeted therapy of small cell carcinoma of the lung. Cell Death Dis 2021;12: 577. doi: 10.1038/s41419-021-03855-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lam LT Lin X Faivre EJ Yang Z Huang X Wilcox DM, et al. Vulnerability of small-cell lung cancer to apoptosis induced by the combination of BET bromodomain proteins and BCL2 inhibitors. Mol Cancer Ther 2017;16: 1511–1520. doi: 10.1158/1535-7163.mct-16-0459. [DOI] [PubMed] [Google Scholar]
  • 78.Inoue-Yamauchi A Jeng PS Kim K Chen HC Han S Ganesan YT, et al. Targeting the differential addiction to anti-apoptotic BCL-2 family for cancer therapy. Nat Commun 2017;8: 16078. doi: 10.1038/ncomms16078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Saunders LR Bankovich AJ Anderson WC Aujay MA Bheddah S Black K, et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci Transl Med 2015;7: 302ra136. doi: 10.1126/scitranslmed.aac9459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kikuchi H Sakakibara-Konishi J Furuta M Yokouchi H Nishihara H Yamazaki S, et al. Expression of Notch1 and Numb in small cell lung cancer. Oncotarget 2017;8: 10348–10358. doi: 10.18632/oncotarget.14411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rudin CM Pietanza MC Bauer TM Ready N Morgensztern D Glisson BS, et al. Rovalpituzumab tesirine, a DLL3-targeted antibody-drug conjugate, in recurrent small-cell lung cancer: A first-in-human, first-in-class, open-label, phase 1 study. Lancet Oncol 2017;18: 42–51. doi: 10.1016/s1470-2045(16)30565-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Carbone DP Morgensztern D Moulec SL Santana-Davila R Ready N Hann CL, et al. Efficacy and safety of rovalpituzumab tesirine in patients with DLL3-expressing, ≥3rd line small cell lung cancer: Results from the phase 2 TRINITY study. J Clin Oncol 2018;36(15_suppl): 8507. doi: 10.1200/JCO.2018.36.15_suppl.8507. [Google Scholar]
  • 83.Blackhall F Jao K Greillier L Cho BC Penkov K Reguart N, et al. Efficacy and safety of rovalpituzumab tesirine compared with topotecan as second-line therapy in DLL3-high SCLC: Results from the phase 3 TAHOE Study. J Thorac Oncol 2021;16: 1547–1558. doi: 10.1016/j.jtho.2021.02.009. [DOI] [PubMed] [Google Scholar]
  • 84.Bauer TM Besse B Martinez-Marti A Trigo JM Moreno V Garrido P, et al. Phase I, open-label, dose-escalation study of the safety, pharmacokinetics, pharmacodynamics, and efficacy of GSK2879552 in relapsed/refractory SCLC. J Thorac Oncol 2019;14: 1828–1838. doi: 10.1016/j.jtho.2019.06.021. [DOI] [PubMed] [Google Scholar]
  • 85.Wang XD Hu R Ding Q Savage TK Huffman KE Williams N, et al. Subtype-specific secretomic characterization of pulmonary neuroendocrine tumor cells. Nat Commun 2019;10: 3201. doi: 10.1038/s41467-019-11153-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Farago AF Yeap BY Stanzione M Hung YP Heist RS Marcoux JP, et al. Combination olaparib and temozolomide in relapsed small-cell lung cancer. Cancer Discov 2019;9: 1372–1387. doi: 10.1158/2159-8290.cd-19-0582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pietanza MC Waqar SN Krug LM Dowlati A Hann CL Chiappori A, et al. Randomized, double-blind, phase II study of temozolomide in combination with either veliparib or placebo in patients with relapsed-sensitive or refractory small-cell lung cancer. J Clin Oncol 2018;36: 2386–2394. doi: 10.1200/jco.2018.77.7672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chi YH Yeh TK Ke YY Lin WH Tsai CH Wang WP, et al. Discovery and synthesis of a pyrimidine-based Aurora kinase inhibitor to reduce levels of MYC oncoproteins. J Med Chem 2021;64: 7312–7330. doi: 10.1021/acs.jmedchem.0c01806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Melichar B Adenis A Lockhart AC Bennouna J Dees EC Kayaleh O, et al. Safety and activity of alisertib, an investigational Aurora kinase a inhibitor, in patients with breast cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma: A five-arm phase 2 study. Lancet Oncol 2015;16: 395–405. doi: 10.1016/s1470-2045(15)70051-3. [DOI] [PubMed] [Google Scholar]
  • 90.Lim KH Opyrchal M Acharya A Boice N Wu N Gao F, et al. Phase 1 study combining alisertib with nab-paclitaxel in patients with advanced solid malignancies. Eur J Cancer 2021;154: 102–110. doi: 10.1016/j.ejca.2021.06.012. [DOI] [PubMed] [Google Scholar]
  • 91.Owonikoko TK Niu H Nackaerts K Csoszi T Ostoros G Mark Z, et al. Randomized phase II study of paclitaxel plus alisertib versus paclitaxel plus placebo as second-line therapy for SCLC: Primary and correlative biomarker analyses. J Thorac Oncol 2020;15: 274–287. doi: 10.1016/j.jtho.2019.10.013. [DOI] [PubMed] [Google Scholar]
  • 92.Poirier JT, Dobromilskaya I, Moriarty WF, Peacock CD, Hann CL, Rudin CM. Selective tropism of Seneca Valley virus for variant subtype small cell lung cancer. J Natl Cancer Inst 2013;105: 1059–1065. doi: 10.1093/jnci/djt130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Owonikoko TK Dwivedi B Chen Z Zhang C Barwick B Ernani V, et al. YAP1 expression in SCLC defines a distinct subtype with T-cell-inflamed phenotype. J Thorac Oncol 2021;16: 464–476. doi: 10.1016/j.jtho.2020.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.McColl K, Wildey G, Sakre N, Lipka MB, Behtaj M, Kresak A. Reciprocal expression of INSM1 and YAP1 defines subgroups in small cell lung cancer. Oncotarget 2017;8: 73745–73756. doi: 10.18632/oncotarget.20572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tlemsani C Pongor L Elloumi F Girard L Huffman KE Roper N, et al. SCLC-CellMiner: A resource for small cell lung cancer cell line genomics and pharmacology based on genomic signatures. Cell Rep 2020;33: 108296. doi: 10.1016/j.celrep.2020.108296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Lee BS Park DI Lee DH Lee JE Yeo MK Park YH, et al. Hippo effector YAP directly regulates the expression of PD-L1 transcripts in EGFR-TKI-resistant lung adenocarcinoma. Biochem Biophys Res Commun 2017;491: 493–499. doi: 10.1016/j.bbrc.2017.07.007. [DOI] [PubMed] [Google Scholar]
  • 97.Shibata M, Ham K, Hoque MO. A time for YAP1: Tumorigenesis, immunosuppression and targeted therapy. Int J Cancer 2018;143: 2133–2144. doi: 10.1002/ijc.31561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Horie M, Saito A, Ohshima M, Suzuki HI, Nagase T. YAP and TAZ modulate cell phenotype in a subset of small cell lung cancer. Cancer Sci 2016;107: 1755–1766. doi: 10.1111/cas.13078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Horn L Mansfield AS Szczesna A Havel L Krzakowski M Hochmair MJ, et al. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med 2018;379: 2220–2229. doi: 10.1056/NEJMoa1809064. [DOI] [PubMed] [Google Scholar]

Articles from Chinese Medical Journal are provided here courtesy of Wolters Kluwer Health

RESOURCES