Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.193
Filter
Add more filters

Publication year range
1.
Cell ; 187(1): 184-203.e28, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181741

ABSTRACT

We performed comprehensive proteogenomic characterization of small cell lung cancer (SCLC) using paired tumors and adjacent lung tissues from 112 treatment-naive patients who underwent surgical resection. Integrated multi-omics analysis illustrated cancer biology downstream of genetic aberrations and highlighted oncogenic roles of FAT1 mutation, RB1 deletion, and chromosome 5q loss. Two prognostic biomarkers, HMGB3 and CASP10, were identified. Overexpression of HMGB3 promoted SCLC cell migration via transcriptional regulation of cell junction-related genes. Immune landscape characterization revealed an association between ZFHX3 mutation and high immune infiltration and underscored a potential immunosuppressive role of elevated DNA damage response activity via inhibition of the cGAS-STING pathway. Multi-omics clustering identified four subtypes with subtype-specific therapeutic vulnerabilities. Cell line and patient-derived xenograft-based drug tests validated the specific therapeutic responses predicted by multi-omics subtyping. This study provides a valuable resource as well as insights to better understand SCLC biology and improve clinical practice.


Subject(s)
Lung Neoplasms , Proteogenomics , Small Cell Lung Carcinoma , Humans , Cell Line , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/chemistry , Small Cell Lung Carcinoma/genetics , Heterografts , Biomarkers, Tumor/analysis
2.
Cell ; 166(2): 275-276, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27419866

ABSTRACT

What changes need to occur in a primary tumor to make it metastatic? Denny et al. address this question for small cell lung cancer (SCLC), finding that changes in genomic accessibility mediated by a single transcription factor, NFIB, comprise at least one mechanism influencing metastasis.


Subject(s)
Cell Line, Tumor , Chromatin , Humans , Lung Neoplasms , Neoplasm Metastasis , Small Cell Lung Carcinoma/genetics , Transcription Factors/genetics
3.
Cell ; 166(2): 328-342, 2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27374332

ABSTRACT

Metastases are the main cause of cancer deaths, but the mechanisms underlying metastatic progression remain poorly understood. We isolated pure populations of cancer cells from primary tumors and metastases from a genetically engineered mouse model of human small cell lung cancer (SCLC) to investigate the mechanisms that drive the metastatic spread of this lethal cancer. Genome-wide characterization of chromatin accessibility revealed the opening of large numbers of distal regulatory elements across the genome during metastatic progression. These changes correlate with copy number amplification of the Nfib locus, and differentially accessible sites were highly enriched for Nfib transcription factor binding sites. Nfib is necessary and sufficient to increase chromatin accessibility at a large subset of the intergenic regions. Nfib promotes pro-metastatic neuronal gene expression programs and drives the metastatic ability of SCLC cells. The identification of widespread chromatin changes during SCLC progression reveals an unexpected global reprogramming during metastatic progression.


Subject(s)
Lung Neoplasms/pathology , NFI Transcription Factors/metabolism , Neoplasm Metastasis/pathology , Small Cell Lung Carcinoma/pathology , Amino Acid Motifs , Animals , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , NFI Transcription Factors/genetics , Promoter Regions, Genetic , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Up-Regulation
4.
Nature ; 627(8005): 880-889, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480884

ABSTRACT

The evolutionary processes that underlie the marked sensitivity of small cell lung cancer (SCLC) to chemotherapy and rapid relapse are unknown1-3. Here we determined tumour phylogenies at diagnosis and throughout chemotherapy and immunotherapy by multiregion sequencing of 160 tumours from 65 patients. Treatment-naive SCLC exhibited clonal homogeneity at distinct tumour sites, whereas first-line platinum-based chemotherapy led to a burst in genomic intratumour heterogeneity and spatial clonal diversity. We observed branched evolution and a shift to ancestral clones underlying tumour relapse. Effective radio- or immunotherapy induced a re-expansion of founder clones with acquired genomic damage from first-line chemotherapy. Whereas TP53 and RB1 alterations were exclusively part of the common ancestor, MYC family amplifications were frequently not constituents of the founder clone. At relapse, emerging subclonal mutations affected key genes associated with SCLC biology, and tumours harbouring clonal CREBBP/EP300 alterations underwent genome duplications. Gene-damaging TP53 alterations and co-alterations of TP53 missense mutations with TP73, CREBBP/EP300 or FMN2 were significantly associated with shorter disease relapse following chemotherapy. In summary, we uncover key processes of the genomic evolution of SCLC under therapy, identify the common ancestor as the source of clonal diversity at relapse and show central genomic patterns associated with sensitivity and resistance to chemotherapy.


Subject(s)
Evolution, Molecular , Immunotherapy , Lung Neoplasms , Platinum , Small Cell Lung Carcinoma , Animals , Female , Humans , Male , Mice , Middle Aged , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Genes, myc/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mutation , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Platinum/pharmacology , Platinum/therapeutic use , Recurrence , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/therapy
5.
Genes Dev ; 36(5-6): 241-258, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35318269

ABSTRACT

Small cell lung cancer (SCLC) is a rapidly growing, highly metastatic, and relatively immune-cold lung cancer subtype. Historically viewed in the laboratory and clinic as a single disease, new discoveries suggest that SCLC comprises multiple molecular subsets. Expression of MYC family members and lineage-related transcription factors ASCL1, NEUROD1, and POU2F3 (and, in some studies, YAP1) define unique molecular states that have been associated with distinct responses to a variety of therapies. However, SCLC tumors exhibit a high degree of intratumoral heterogeneity, with recent studies suggesting the existence of tumor cell plasticity and phenotypic switching between subtype states. While SCLC plasticity is correlated with, and likely drives, therapeutic resistance, the mechanisms underlying this plasticity are still largely unknown. Subtype states are also associated with immune-related gene expression, which likely impacts response to immune checkpoint blockade and may reveal novel targets for alternative immunotherapeutic approaches. In this review, we synthesize recent discoveries on the mechanisms of SCLC plasticity and how these processes may impinge on antitumor immunity.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/therapy , Small Cell Lung Carcinoma/genetics , Transcription Factors/metabolism
6.
Cell ; 156(6): 1298-1311, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24630729

ABSTRACT

Small cell lung carcinoma (SCLC) is a highly lethal, smoking-associated cancer with few known targetable genetic alterations. Using genome sequencing, we characterized the somatic evolution of a genetically engineered mouse model (GEMM) of SCLC initiated by loss of Trp53 and Rb1. We identified alterations in DNA copy number and complex genomic rearrangements and demonstrated a low somatic point mutation frequency in the absence of tobacco mutagens. Alterations targeting the tumor suppressor Pten occurred in the majority of murine SCLC studied, and engineered Pten deletion accelerated murine SCLC and abrogated loss of Chr19 in Trp53; Rb1; Pten compound mutant tumors. Finally, we found evidence for polyclonal and sequential metastatic spread of murine SCLC by comparative sequencing of families of related primary tumors and metastases. We propose a temporal model of SCLC tumorigenesis with implications for human SCLC therapeutics and the nature of cancer-genome evolution in GEMMs.


Subject(s)
Carcinogenesis , Disease Models, Animal , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Animals , Humans , Liver Neoplasms/secondary , Lymphatic Metastasis , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Small Cell Lung Carcinoma/secondary
7.
Genes Dev ; 35(11-12): 847-869, 2021 06.
Article in English | MEDLINE | ID: mdl-34016693

ABSTRACT

ASCL1 is a neuroendocrine lineage-specific oncogenic driver of small cell lung cancer (SCLC), highly expressed in a significant fraction of tumors. However, ∼25% of human SCLC are ASCL1-low and associated with low neuroendocrine fate and high MYC expression. Using genetically engineered mouse models (GEMMs), we show that alterations in Rb1/Trp53/Myc in the mouse lung induce an ASCL1+ state of SCLC in multiple cells of origin. Genetic depletion of ASCL1 in MYC-driven SCLC dramatically inhibits tumor initiation and progression to the NEUROD1+ subtype of SCLC. Surprisingly, ASCL1 loss promotes a SOX9+ mesenchymal/neural crest stem-like state and the emergence of osteosarcoma and chondroid tumors, whose propensity is impacted by cell of origin. ASCL1 is critical for expression of key lineage-related transcription factors NKX2-1, FOXA2, and INSM1 and represses genes involved in the Hippo/Wnt/Notch developmental pathways in vivo. Importantly, ASCL1 represses a SOX9/RUNX1/RUNX2 program in vivo and SOX9 expression in human SCLC cells, suggesting a conserved function for ASCL1. Together, in a MYC-driven SCLC model, ASCL1 promotes neuroendocrine fate and represses the emergence of a SOX9+ nonendodermal stem-like fate that resembles neural crest.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , SOX9 Transcription Factor/genetics , Small Cell Lung Carcinoma/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neural Crest/cytology , Small Cell Lung Carcinoma/physiopathology , Stem Cells/cytology
8.
Genes Dev ; 34(17-18): 1210-1226, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32820040

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive neuroendocrine cancer characterized by initial chemosensitivity followed by emergence of chemoresistant disease. To study roles for MYCN amplification in SCLC progression and chemoresistance, we developed a genetically engineered mouse model of MYCN-overexpressing SCLC. In treatment-naïve mice, MYCN overexpression promoted cell cycle progression, suppressed infiltration of cytotoxic T cells, and accelerated SCLC. MYCN overexpression also suppressed response to cisplatin-etoposide chemotherapy, with similar findings made upon MYCL overexpression. We extended these data to genetically perturb chemosensitive patient-derived xenograft (PDX) models of SCLC. In chemosensitive PDX models, overexpression of either MYCN or MYCL also conferred a switch to chemoresistance. To identify therapeutic strategies for MYCN-overexpressing SCLC, we performed a genome-scale CRISPR-Cas9 sgRNA screen. We identified the deubiquitinase USP7 as a MYCN-associated synthetic vulnerability. Pharmacological inhibition of USP7 resensitized chemoresistant MYCN-overexpressing PDX models to chemotherapy in vivo. Our findings show that MYCN overexpression drives SCLC chemoresistance and provide a therapeutic strategy to restore chemosensitivity.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , N-Myc Proto-Oncogene Protein/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Heterografts , Humans , Lung Neoplasms/enzymology , Mice , N-Myc Proto-Oncogene Protein/genetics , Small Cell Lung Carcinoma/enzymology , Small Cell Lung Carcinoma/genetics
9.
Proc Natl Acad Sci U S A ; 121(23): e2317790121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38814866

ABSTRACT

The transformation of lung adenocarcinoma to small cell lung cancer (SCLC) is a recognized resistance mechanism and a hindrance to therapies using epidermal growth factor receptor tyrosine kinase inhibitors (TKIs). The paucity of pretranslational/posttranslational clinical samples limits the deeper understanding of resistance mechanisms and the exploration of effective therapeutic strategies. Here, we developed preclinical neuroendocrine (NE) transformation models. Next, we identified a transcriptional reprogramming mechanism that drives resistance to erlotinib in NE transformation cell lines and cell-derived xenograft mice. We observed the enhanced expression of genes involved in the EHMT2 and WNT/ß-catenin pathways. In addition, we demonstrated that EHMT2 increases methylation of the SFRP1 promoter region to reduce SFRP1 expression, followed by activation of the WNT/ß-catenin pathway and TKI-mediated NE transformation. Notably, the similar expression alterations of EHMT2 and SFRP1 were observed in transformed SCLC samples obtained from clinical patients. Importantly, suppression of EHMT2 with selective inhibitors restored the sensitivity of NE transformation cell lines to erlotinib and delayed resistance in cell-derived xenograft mice. We identify a transcriptional reprogramming process in NE transformation and provide a potential therapeutic target for overcoming resistance to erlotinib.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Transformation, Neoplastic , Erlotinib Hydrochloride , Lung Neoplasms , Humans , Animals , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Mice , Erlotinib Hydrochloride/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Drug Resistance, Neoplasm/genetics , Wnt Signaling Pathway/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transcription, Genetic , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase
10.
Genes Dev ; 32(13-14): 915-928, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29945888

ABSTRACT

Small cell lung cancer (SCLC) is widely considered to be a tumor of pulmonary neuroendocrine cells; however, a variant form of this disease has been described that lacks neuroendocrine features. Here, we applied domain-focused CRISPR screening to human cancer cell lines to identify the transcription factor (TF) POU2F3 (POU class 2 homeobox 3; also known as SKN-1a/OCT-11) as a powerful dependency in a subset of SCLC lines. An analysis of human SCLC specimens revealed that POU2F3 is expressed exclusively in variant SCLC tumors that lack expression of neuroendocrine markers and instead express markers of a chemosensory lineage known as tuft cells. Using chromatin- and RNA-profiling experiments, we provide evidence that POU2F3 is a master regulator of tuft cell identity in a variant form of SCLC. Moreover, we show that most SCLC tumors can be classified into one of three lineages based on the expression of POU2F3, ASCL1, or NEUROD1. Our CRISPR screens exposed other unique dependencies in POU2F3-expressing SCLC lines, including the lineage TFs SOX9 and ASCL2 and the receptor tyrosine kinase IGF1R (insulin-like growth factor 1 receptor). These data reveal POU2F3 as a cell identity determinant and a dependency in a tuft cell-like variant of SCLC, which may reflect a previously unrecognized cell of origin or a trans-differentiation event in this disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Humans , Lung/pathology , Mice , Receptor, IGF Type 1/metabolism
11.
Semin Cancer Biol ; 90: 45-56, 2023 05.
Article in English | MEDLINE | ID: mdl-36787870

ABSTRACT

The addition of immune checkpoint blockade (ICB) therapy to standard chemotherapy has been shown to improve survival in patients with metastatic small cell lung cancer. However, the benefit is modest and there remains an unmet need for novel therapeutic approaches to enhance the effectiveness of immunotherapy in this disease, both in the early and late stages. Ionizing radiation, which is a standard treatment for small cell lung cancer, is known to trigger immunogenic cell death in tumor cells, making it an attractive partner for ICB therapies in multiple solid tumor types. However, the optimal radiation dosage and fractionation scheme, target sites for radiation, and sequencing of radiation in relation to ICB treatment are still unclear. In this review we discuss the molecular biology underlying radiation-induced tumor immunity as well as pre-clinical and clinical studies combining radiation with ICB treatments, with a focus on translational and clinical trials in small cell lung cancer.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/radiotherapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Combined Modality Therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy
12.
Semin Cancer Biol ; 94: 1-10, 2023 09.
Article in English | MEDLINE | ID: mdl-37244438

ABSTRACT

Lung cancer is the leading cause of cancer related death, and is divided into two major histological subtypes, non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). Histological transformation from NSCLC to SCLC has been reported as a mechanism of treatment resistance in patients who received tyrosine kinase inhibitors (TKIs) targeting EGFR, ALK and ROS1 or immunotherapies. The transformed histology could be due to therapy-induced lineage plasticity or clonal selection of pre-existing SCLC cells. Evidence supporting either mechanism exist in the literature. Here, we discuss potential mechanisms of transformation and review the current knowledge about cell of origin of NSCLC and SCLC. In addition, we summarize genomic alterations that are frequently observed in both "de novo" and transformed SCLC, such as TP53, RB1 and PIK3CA. We also discuss treatment options for transformed SCLC, including chemotherapy, radiotherapy, TKIs, immunotherapy and anti-angiogenic agents.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/therapeutic use , ErbB Receptors/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/therapy , Mutation
13.
Mol Cancer ; 23(1): 41, 2024 02 24.
Article in English | MEDLINE | ID: mdl-38395864

ABSTRACT

Current treatment guidelines refer to small cell lung cancer (SCLC), one of the deadliest human malignancies, as a homogeneous disease. Accordingly, SCLC therapy comprises chemoradiation with or without immunotherapy. Meanwhile, recent studies have made significant advances in subclassifying SCLC based on the elevated expression of the transcription factors ASCL1, NEUROD1, and POU2F3, as well as on certain inflammatory characteristics. The role of the transcription regulator YAP1 in defining a unique SCLC subset remains to be established. Although preclinical analyses have described numerous subtype-specific characteristics and vulnerabilities, the so far non-existing clinical subtype distinction may be a contributor to negative clinical trial outcomes. This comprehensive review aims to provide a framework for the development of novel personalized therapeutic approaches by compiling the most recent discoveries achieved by preclinical SCLC research. We highlight the challenges faced due to limited access to patient material as well as the advances accomplished by implementing state-of-the-art models and methodologies.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Immunotherapy , Transcription Factors
14.
Mol Cancer ; 23(1): 115, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38811992

ABSTRACT

BACKGROUND: We explored potential predictive biomarkers of immunotherapy response in patients with extensive-stage small-cell lung cancer (ES-SCLC) treated with durvalumab (D) + tremelimumab (T) + etoposide-platinum (EP), D + EP, or EP in the randomized phase 3 CASPIAN trial. METHODS: 805 treatment-naïve patients with ES-SCLC were randomized (1:1:1) to receive D + T + EP, D + EP, or EP. The primary endpoint was overall survival (OS). Patients were required to provide an archived tumor tissue block (or ≥ 15 newly cut unstained slides) at screening, if these samples existed. After assessment for programmed cell death ligand-1 expression and tissue tumor mutational burden, residual tissue was used for additional molecular profiling including by RNA sequencing and immunohistochemistry. RESULTS: In 182 patients with transcriptional molecular subtyping, OS with D ± T + EP was numerically highest in the SCLC-inflamed subtype (n = 10, median 24.0 months). Patients derived benefit from immunotherapy across subtypes; thus, additional biomarkers were investigated. OS benefit with D ± T + EP versus EP was greater with high versus low CD8A expression/CD8 cell density by immunohistochemistry, but with no additional benefit with D + T + EP versus D + EP. OS benefit with D + T + EP versus D + EP was associated with high expression of CD4 (median 25.9 vs. 11.4 months) and antigen-presenting and processing machinery (25.9 vs. 14.6 months) and MHC I and II (23.6 vs. 17.3 months) gene signatures, and with higher MHC I expression by immunohistochemistry. CONCLUSIONS: These findings demonstrate the tumor microenvironment is important in mediating better outcomes with D ± T + EP in ES-SCLC, with canonical immune markers associated with hypothesized immunotherapy mechanisms of action defining patient subsets that respond to D ± T. TRIAL REGISTRATION: ClinicalTrials.gov, NCT03043872.


Subject(s)
Biomarkers, Tumor , Immunotherapy , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/immunology , Small Cell Lung Carcinoma/therapy , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/mortality , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/metabolism , Female , Male , Immunotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Aged , Antibodies, Monoclonal/therapeutic use , Treatment Outcome , Neoplasm Staging , Antibodies, Monoclonal, Humanized/therapeutic use , Prognosis , Adult
15.
Curr Opin Oncol ; 36(1): 51-56, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37865844

ABSTRACT

PURPOSE OF REVIEW: Small cell lung cancer (SCLC) remains one of the most aggressive thoracic malignancies with an especially dismal prognosis. While the detection of various targetable driver mutations and immune checkpoints have revolutionized the treatment of non-small cell lung cancer (NSCLC), there has been only modest therapeutic innovation over the past decades in SCLC. In this review, we aim to provide a brief summary on the clinical relevance of recent research findings, which could soon pave the way towards a more personalized and targeted management of SCLC patients. RECENT FINDINGS: Substantial research on the biological and molecular heterogeneity of SCLC has been conducted in the last years. Recent results from comprehensive profiling studies have shown that unique major SCLC subtypes can be distinguished based on the relative expression of key transcription regulators (ASCL1, NEUROD1, POU2F3) or distinct inflammatory features. Understanding the differing molecular characteristics of these distinct subtypes has resulted in the identification of specific therapeutic vulnerabilities. SUMMARY: The recently introduced molecular SCLC subtype classification represents a substantial progress towards a personalized and more efficacious approach in SCLC. The consequences of this paradigm shift provide hope for improved patient care and clinical outcomes in this exceptionally lethal thoracic malignancy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy
16.
J Transl Med ; 22(1): 189, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383412

ABSTRACT

BACKGROUND: Combined small-cell lung carcinoma (cSCLC) represents a rare subtype of SCLC, the mechanisms governing the evolution of cancer genomes and their impact on the tumor immune microenvironment (TIME) within distinct components of cSCLC remain elusive. METHODS: Here, we conducted whole-exome and RNA sequencing on 32 samples from 16 cSCLC cases. RESULTS: We found striking similarities between two components of cSCLC-LCC/LCNEC (SCLC combined with large-cell carcinoma/neuroendocrine) in terms of tumor mutation burden (TMB), tumor neoantigen burden (TNB), clonality structure, chromosomal instability (CIN), and low levels of immune cell infiltration. In contrast, the two components of cSCLC-ADC/SCC (SCLC combined with adenocarcinoma/squamous-cell carcinoma) exhibited a high level of tumor heterogeneity. Our investigation revealed that cSCLC originated from a monoclonal source, with two potential transformation modes: from SCLC to SCC (mode 1) and from ADC to SCLC (mode 2). Therefore, cSCLC might represent an intermediate state, potentially evolving into another histological tumor morphology through interactions between tumor and TIME surrounding it. Intriguingly, RB1 inactivation emerged as a factor influencing TIME heterogeneity in cSCLC, possibly through neoantigen depletion. CONCLUSIONS: Together, these findings delved into the clonal origin and TIME heterogeneity of different components in cSCLC, shedding new light on the evolutionary processes underlying this enigmatic subtype.


Subject(s)
Adenocarcinoma , Carcinoma, Large Cell , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Microdissection , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Adenocarcinoma/genetics , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/pathology , Genomics , Tumor Microenvironment/genetics
17.
Toxicol Appl Pharmacol ; 485: 116915, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38537875

ABSTRACT

N-myc downstream-regulated gene 2 (NDRG2) has been recognised as a negative regulator of the progression of numerous tumours, yet its specific role in small-cell lung carcinoma (SCLC) is not fully understood. The purpose of the current study was to investigate the biological role and mechanism of NDRG2 in SCLC. Initial investigation using the Gene Expression Omnibus (GEO) dataset revealed marked downregulation of NDRG2 transcripts in SCLC. The decreased abundance of NDRG2 in SCLC was verified by examining clinical specimens. Increasing NDRG2 expression in SCLC cell lines caused significant changes in cell proliferation, cell cycle progression, colony formation, and chemosensitivity. NDRG2 overexpression decreased the levels of phosphorylated PTEN, AKT and mTOR. In PTEN-depleted SCLC cells, the upregulation of NDRG2 did not result in any noticeable impact on AKT or mTOR activation. Additionally, the reactivation of AKT reversed the antitumour effects of NDRG2 in SCLC cells. Notably, increasing NDRG2 expression retarded the growth of SCLC cell-derived xenografts in vivo. In conclusion, NDRG2 serves as an inhibitor of SCLC, and its cancer-inhibiting effects are achieved through the suppression of AKT/mTOR via the activation of PTEN. This work suggests that NDRG2 is a potential druggable target for SCLC treatment.


Subject(s)
Cell Proliferation , Lung Neoplasms , Mice, Nude , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt , Signal Transduction , Small Cell Lung Carcinoma , TOR Serine-Threonine Kinases , Tumor Suppressor Proteins , Humans , TOR Serine-Threonine Kinases/metabolism , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Mice , Disease Progression , Gene Expression Regulation, Neoplastic , Female , Male , Mice, Inbred BALB C , Xenograft Model Antitumor Assays
18.
Arch Biochem Biophys ; 751: 109824, 2024 01.
Article in English | MEDLINE | ID: mdl-37984759

ABSTRACT

Small-cell lung cancer (SCLC), which accounts for about 15 % of all lung cancers, progresses more rapidly than other histologic types and is rarely detected at an operable early stage. Therefore, chemotherapy, radiation therapy, or their combination are the primary treatments for this type of lung cancer. However, the tendency to acquire resistance to anticancer drugs is a severe problem. Recently, we found that an intercellular adhesion molecule, claudin (CLDN) 1, known to be involved in the migration and invasion of lung cancer cells, is involved in the acquisition of anticancer drug resistance. In the present study, we investigated the effect of CLDN1 on the anticancer-drug sensitivity of SCLC SBC-3 cells. Since epithelial-mesenchymal transition (EMT), which is involved in cancer cell migration and invasion, is well known for its involvement in anticancer-drug sensitivity via inhibition of apoptosis, we also examined EMT involvement in decreased anticancer-drug sensitivity by CLDN1. Sensitivity to doxorubicin (DOX) in SBC-3 cells was significantly decreased by CLDN1 overexpression. CLDN1 overexpression resulted in increased TGF-ß1 levels, enhanced EMT induction, and increased migratory potency of SBC-3 cells. The decreased sensitivity of SBC-3 cells to anticancer drugs upon TGF-ß1 treatment suggested that activation of the TGF-ß1/EMT signaling pathway by CLDN1 causes the decreased sensitivity to anticancer drugs and increased migratory potency. Furthermore, treatments with antiallergic agents tranilast and zoledronic acid, known EMT inhibitors, significantly mitigated the decreased sensitivity of CLDN1-overexpressing SBC-3 cells to DOX. These results suggest that EMT inhibitors might effectively overcome reduced sensitivity to anticancer drugs in CLDN1-overexpressing SCLC cells.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Claudin-1/genetics , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Epithelial-Mesenchymal Transition
19.
FASEB J ; 37(10): e23170, 2023 10.
Article in English | MEDLINE | ID: mdl-37676718

ABSTRACT

Small cell lung cancer (SCLC) is one of the most malignant tumors that has an extremely poor prognosis. RNA-binding protein (RBP) and long noncoding RNA (lncRNA) have been shown to be key regulators during tumorigenesis as well as lung tumor progression. However, the role of RBP ELAVL4 and lncRNA LYPLAL1-DT in SCLC remains unclear. In this study, we verified that lncRNA LYPLAL1-DT acts as an SCLC oncogenic lncRNA and was confirmed in vitro and in vivo. Mechanistically, LYPLAL1-DT negatively regulates the expression of miR-204-5p, leading to the upregulation of PFN2, thus, promoting SCLC cell proliferation, migration, and invasion. ELAVL4 has been shown to enhance the stability of LYPLAL1-DT and PFN2 mRNA. Our study reveals a regulatory pathway, where ELAVL4 stabilizes PFN2 and LYPLAL1-DT with the latter further increasing PFN2 expression by blocking the action of miR-204-5p. Upregulated PFN2 ultimately promotes tumorigenesis and invasion in SCLC. These findings provide novel prognostic indicators as well as promising new therapeutic targets for SCLC.


Subject(s)
Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Small Cell Lung Carcinoma , Humans , RNA, Long Noncoding/genetics , Profilins/genetics , Small Cell Lung Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Carcinogenesis/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , ELAV-Like Protein 4
20.
Cell Commun Signal ; 22(1): 45, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233864

ABSTRACT

OBJECTIVES: Histological transformation to small cell lung cancer (SCLC) has been identified as a mechanism of TKIs resistance in EGFR-mutant non-small cell lung cancer (NSCLC). We aim to explore the prevalence of transformation in EGFR-wildtype NSCLC and the mechanism of SCLC transformation, which are rarely understood. METHODS: We reviewed 1474 NSCLC patients to investigate the NSCLC-to-SCLC transformed cases and the basic clinical characteristics, driver gene status and disease course of them. To explore the potential functional genes in SCLC transformation, we obtained pre- and post-transformation specimens and subjected them to a multigene NGS panel involving 416 cancer-related genes. To validate the putative gene function, we established knocked-out models by CRISPR-Cas 9 in HCC827 and A549-TP53-/- cells and investigated the effects on tumor growth, drug sensitivity and neuroendocrine phenotype in vitro and in vivo. We also detected the expression level of protein and mRNA to explore the molecular mechanism involved. RESULTS: We firstly reported an incidence rate of 9.73% (11/113) of SCLC transformation in EGFR-wildtype NSCLC and demonstrated that SCLC transformation is irrespective of EGFR mutation status (P = 0.16). We sequenced 8 paired tumors and identified a series of mutant genes specially in transformed SCLC such as SMAD4, RICTOR and RET. We firstly demonstrated that SMAD4 deficiency can accelerate SCLC transition by inducing neuroendocrine phenotype regardless of RB1 status in TP53-deficient NSCLC cells. Further mechanical experiments identified the SMAD4 can regulate ASCL1 transcription competitively with Myc in NSCLC cells and Myc inhibitor acts as a potential subsequent treatment agent. CONCLUSIONS: Transformation to SCLC is irrespective of EFGR status and can be accelerated by SMAD4 in non-small cell lung cancer. Myc inhibitor acts as a potential therapeutic drug for SMAD4-mediated resistant lung cancer. Video Abstract.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Lung Neoplasms/pathology , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Retinoblastoma Binding Proteins/genetics , Smad4 Protein/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL