Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (15)

Search Parameters:
Keywords = etoposide-resistant lung cancer

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 7558 KB  
Article
Reversible Upregulation of the Senescence-Associated Beta-Galactosidase Marker Induced by Cell Detachment in Cancer Cells
by Nina Semenova, Juan Sebastian Yakisich, Robyn Ascue, Anand K. V. Iyer and Neelam Azad
Cells 2025, 14(21), 1667; https://doi.org/10.3390/cells14211667 - 24 Oct 2025
Viewed by 667
Abstract
During metastasis, cancer cells detach from the primary tumor, and the floating cells enter the circulation and reattach in distant organs. Floating cells are highly chemoresistant to anticancer drugs, but the underlying mechanisms are poorly understood. We hypothesized that floating cells transition into [...] Read more.
During metastasis, cancer cells detach from the primary tumor, and the floating cells enter the circulation and reattach in distant organs. Floating cells are highly chemoresistant to anticancer drugs, but the underlying mechanisms are poorly understood. We hypothesized that floating cells transition into a quiescent/senescent (Q/S) state. Using human lung carcinoma H460 and H23, human prostate adenocarcinoma PC3, and human breast adenocarcinoma MDA-MB-231 cells, we found (1) a progressive increase in activity of β-galactosidase (β-Gal), a marker associated with Q/S cells, (2) a transition to a non-proliferative state while growing under anchorage-independent conditions, and (3) upon reattachment, the β-Gal activity returned to the basal level and cells resumed proliferation. Similar experiments were performed in parallel with cells treated with etoposide (Eto), a well-known inductor of senescence. Eto-untreated floating cells resumed proliferation faster and showed a quicker decrease in β-Gal activity compared to Eto-induced senescent cells. We conclude that cell detachment per se triggers a reversible (plastic) increase in β-Gal. Our findings provide a partial explanation for chemoresistance under anchorage-independent conditions and a new target to eliminate highly resistant floating cells. Ultimately, eliminating Q/S floating cells may prevent or reduce metastasis. Full article
(This article belongs to the Section Cellular Aging)
Show Figures

Figure 1

19 pages, 731 KB  
Review
Transformation to Neuroendocrine Phenotype in Non-Small-Cell Lung Carcinoma: A Literature Review
by Irene Hernández de Córdoba, Xabier Mielgo-Rubio, Paloma Cejas, Jorge Palomar Ramos, Beatriz Garrido-Rubiales, Sandra Falagán Martínez, Gustavo Rubio Romero, María Morales Parga, Laura Moll Taltavull, Andrea Fernández González, Enrique Casado Sáenz and María Sereno
Int. J. Mol. Sci. 2025, 26(11), 5096; https://doi.org/10.3390/ijms26115096 - 26 May 2025
Cited by 2 | Viewed by 6145
Abstract
Neuroendocrine transformation in non-small-cell lung cancer (NSCLC) is an uncommon but clinically significant resistance mechanism to targeted therapy, immunotherapy, and chemotherapy. This phenomenon, primarily observed in adenocarcinoma (ADC) with EGFR mutations under tyrosine kinase inhibitor (TKI) treatment, leads to histological transformation into small-cell [...] Read more.
Neuroendocrine transformation in non-small-cell lung cancer (NSCLC) is an uncommon but clinically significant resistance mechanism to targeted therapy, immunotherapy, and chemotherapy. This phenomenon, primarily observed in adenocarcinoma (ADC) with EGFR mutations under tyrosine kinase inhibitor (TKI) treatment, leads to histological transformation into small-cell lung cancer (SCLC), commonly associated with an aggressive clinical course and poor prognosis. Standard platinum–etoposide chemotherapy provides only transient disease control, highlighting the urgent need for improved therapeutic strategies. Early identification of transformation through biopsy, liquid biopsy, or biomarkers like neuron-specific enolase (NSE) and pro-gastrin-releasing peptide (pro-GRP) may allow for early intervention. As targeted therapies continue to develop, understanding the molecular drivers of neuroendocrine transformation is crucial for optimizing treatment. Further research into novel treatment approaches, including combination therapies with TKIs, chemotherapy, immunotherapy, and epigenetic modulators, is required to improve outcomes for these patients. Full article
(This article belongs to the Special Issue Molecular and Translational Research of Non-Small Cell Lung Cancer)
Show Figures

Figure 1

26 pages, 6968 KB  
Article
Roles of Annexin A1 Expression in Small Cell Lung Cancer
by Ágnes Paál, David Dora, Ákos Takács, Christopher Rivard, Shivaun Lueke Pickard, Fred R. Hirsch, Brigitta Roskó, Peter Kiraly, Péter Ferdinandy, Zoltán V. Varga, Zoltan Lohinai and Anikó Görbe
Cancers 2025, 17(9), 1407; https://doi.org/10.3390/cancers17091407 - 23 Apr 2025
Viewed by 1846
Abstract
Background/Objectives: Small cell lung cancer (SCLC) is one of the malignancies with the worst prognosis, and there have been no major breakthroughs in its treatment for a long time. The majority of patients are diagnosed at the extensive stage, where the only option [...] Read more.
Background/Objectives: Small cell lung cancer (SCLC) is one of the malignancies with the worst prognosis, and there have been no major breakthroughs in its treatment for a long time. The majority of patients are diagnosed at the extensive stage, where the only option is chemotherapy, and even the addition of immune checkpoint inhibitors results in only modest benefits. The characterization of the molecular mechanisms behind therapy resistance has relevance in finding novel therapeutic approaches. Previous studies showed the possibility of annexin A1’s (ANXA1) involvement in the immunosuppressive tumor microenvironment in SCLC, and there are studies showing the direct effects of ANXA1 modulation on cancer cell aggressiveness. Methods: We aimed to characterize the roles of ANXA1 expression using publicly available transcriptomic data, the RNA-seq-based predictive algorithms EPIC and ESTIMATE, and immunohistochemistry on patient samples. For the in vitro studies, we silenced ANXA1 expression with short hairpin RNA in three SCLC cell lines, measured the growth rate with the trypan blue exclusion assay, assessed the chemosensitivity to cisplatin and etoposide with the Presto BlueTM viability assay, and performed Western blots to assess changes in the levels of metabolic and mesenchymal markers and transcriptional drivers. Results: ANXA1-high tumors are associated with significantly increased immune infiltrates, stromality, and tumor-associated macrophages (TAMs). The ANXA1 protein is expressed on tumor cells and TAMs at the tissue level. ANXA1 silencing in H841 cells did not affect the growth rate; in SW1271 cells, shANXA1 cells grew significantly slower than shCTRL cells. Meanwhile, in H1048 cells, proliferation was significantly faster. Despite the different growth rates of the tested cell lines, ANXA1 silencing decreased the chemosensitivity to both cisplatin and etoposide in all three cell lines. Gene expression changes in mesenchymal markers, metabolic markers, dominant transcriptional drivers, and immune-relevant molecules were also characterized. Conclusions: This is the first comprehensive characterization of ANXA1 in SCLC to reveal its role in the tumor’s cell biology and the TME, aiming to boost further research in the field. Full article
Show Figures

Figure 1

17 pages, 1705 KB  
Article
Associating Patient Responses with Drug Sensitivity in Non-Small Cell Lung Carcinoma Using an Immunoassay on Patient-Derived Cell Cultures
by Ana Podolski-Renić, Sofija Jovanović Stojanov, Dragana Marić, Jelena Dinić, Miodrag Dragoj, Ana Stepanović, Ema Lupšić, Milica Pajović, Sofija Glumac, Maja Ercegovac and Milica Pešić
Curr. Issues Mol. Biol. 2025, 47(4), 281; https://doi.org/10.3390/cimb47040281 - 17 Apr 2025
Viewed by 1235
Abstract
Background/Objectives: Non-small cell lung carcinoma (NSCLC) is characterized by its diverse molecular profiles and varying responses to treatment, highlighting the importance of precision medicine in optimizing therapeutic outcomes. A promising approach involves using patient-derived cellular models, which provide insights into the unique [...] Read more.
Background/Objectives: Non-small cell lung carcinoma (NSCLC) is characterized by its diverse molecular profiles and varying responses to treatment, highlighting the importance of precision medicine in optimizing therapeutic outcomes. A promising approach involves using patient-derived cellular models, which provide insights into the unique biology of individual tumors and their responsiveness to treatment. Methods: We established short-term primary cell cultures from thirteen patients with NSCLC of different subtypes and stages, including both cancer and stromal cells. To evaluate the ex vivo cytotoxicity and selectivity of eight chemotherapeutics and erlotinib, we employed an immunoassay, and the results were analyzed using an automated imaging system. Scoring of the obtained results was also performed. The ex vivo responses to cisplatin, etoposide, and paclitaxel were correlated with the patients’ responses to therapy. We used Kaplan–Meier analysis to assess progression-free survival (PFS) differences among patient groups. Results: NSCLC cells exhibited significant variability in their responses to drugs, with stromal cells demonstrating greater sensitivity. Tumors at stages I-III responded to multiple treatments, whereas stage IV cells showed considerable resistance. Erlotinib effectively reduced cancer cell growth at lower doses but plateaued at higher concentrations. The immunoassay indicated 67% sensitivity and 100% specificity in predicting patient responses to chemotherapy. Sensitivity to etoposide and paclitaxel correlated with progression-free survival (PFS). Conclusions: A personalized treatment strategy, such as our immunoassay based on the ex vivo responses of cancer patients’ cells, can guide treatment decisions and, in some cases, serve as surrogate biomarkers for tumor types that lack actionable biomarkers. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Graphical abstract

25 pages, 4721 KB  
Article
Cytotoxic Effect of Escitalopram/Etoposide Combination on Etoposide-Resistant Lung Cancer
by Serap Özkaya Gül, Beyzanur Şimşek, Fidan Yıldız and Esra Aydemir
Pharmaceuticals 2025, 18(4), 531; https://doi.org/10.3390/ph18040531 - 5 Apr 2025
Cited by 2 | Viewed by 2109
Abstract
Background: Antidepressants are a class of pharmaceuticals utilized for the management of many psychiatric disorders, including depression. A considerable number of antidepressants, particularly selective serotonin reuptake inhibitors (SSRIs), have been documented to demonstrate significant anticancer properties in various cancer cell lines. Objectives: The [...] Read more.
Background: Antidepressants are a class of pharmaceuticals utilized for the management of many psychiatric disorders, including depression. A considerable number of antidepressants, particularly selective serotonin reuptake inhibitors (SSRIs), have been documented to demonstrate significant anticancer properties in various cancer cell lines. Objectives: The aim of this study was to evaluate the selective cytotoxic and apoptotic effects of escitalopram oxalate (ES) alone and in combination with etoposide (ET) on ET-resistant A549 (A549/90E) lung cancer cells. Methods: The cytotoxic effects of the drugs were determined by CCK-8, trypan blue, and neutral red assays. Apoptosis was observed by Annexin V fluorescein isothiocyanate (FITC)/PI and mitochondrial membrane potential (ΔΨm) assays. Moreover, the effects of the drugs, alone and in combination, on apoptosis-related proteins, caspase-3, PTEN, and resistance-related P-gP were determined by ELISA. The relationship between drugs and lung cancer was determined with protein–protein interaction (PPI) network analysis. Results: Our results revealed that ES significantly exerted cytotoxic effects on both wild-type and A549/90E cells compared with BEAS-2B cells. The IC50 values of 48.67 and 51.6 μg/mL obtained for ET and ES, respectively, at the end of 24 h of incubation for A549 cells were applied reciprocally for each cell by including BEAS-2B together with the 2xIC50 and ½ IC50 values. The results of each combination were statistically evaluated with combination indices (CIs) obtained using the Compusyn synergistic effect analysis program. Combination doses with a synergistic effect in A549 and A549/90E cells and an antagonistic effect in BEAS-2B cells have been determined as ½ IC50 for ET and ½ IC50 for ES. ET ½ IC50, ES ½ IC50, and an ET ½ IC50 + ES ½ IC50 combination caused 18.37%, 55.19%, and 57.55% death in A549 cells, whereas they caused 44.9%, 22.4%, and 51.94% death in A549/90E cells, respectively. In A549 cells, the combination of ES ½ IC50 and ET ½ IC50 caused increased levels of caspase-3 (p < 0.01) and P-gP (p < 0.001), while PTEN levels remained unchanged. The combination resulted in an increase in caspase-3 (p < 0.001) and PTEN (p < 0.001) amounts, alongside a decrease in P-gP (p < 0.01) levels in A549/90E cells. The death mechanism induced by the combination was found to be apoptotic by Annexin V-FITC and ΔΨm assays. Conclusions: Based on our findings, ES was observed to induce cytotoxic and apoptotic activities in A549/90E cells in vitro. ES in combination therapy is considered to be effective to overcome ET resistance by reducing the amount of P-gP in A549/90E cells. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

17 pages, 1314 KB  
Review
Etoposide as a Key Therapeutic Agent in Lung Cancer: Mechanisms, Efficacy, and Emerging Strategies
by Jung Yoon Jang, Donghwan Kim, Eunok Im and Nam Deuk Kim
Int. J. Mol. Sci. 2025, 26(2), 796; https://doi.org/10.3390/ijms26020796 - 18 Jan 2025
Cited by 14 | Viewed by 6557
Abstract
Topoisomerase II inhibitors, particularly etoposide, have long been integral to the treatment of lung cancer, especially small cell lung cancer. This review comprehensively examines the mechanisms of action of etoposide, its clinical efficacy, and its role in current lung cancer treatment regimens. Etoposide [...] Read more.
Topoisomerase II inhibitors, particularly etoposide, have long been integral to the treatment of lung cancer, especially small cell lung cancer. This review comprehensively examines the mechanisms of action of etoposide, its clinical efficacy, and its role in current lung cancer treatment regimens. Etoposide exerts its anticancer effects by inducing DNA strand breaks through the inhibition of topoisomerase II, leading to cancer cell apoptosis. Despite their widespread use, challenges such as drug resistance, toxicity, and limited efficacy in non-small cell lung cancer have spurred ongoing research on combination therapies and novel drug formulations. Emerging therapeutic strategies include the integration of etoposide with immunotherapy, targeted therapies, and novel drug delivery systems aimed at enhancing the therapeutic window and overcoming drug resistance. This article aims to inform the development of more effective treatment strategies by providing a critical overview of the clinical applications of etoposide and exploring future directions for lung cancer therapy. Full article
(This article belongs to the Special Issue Topoisomerase Inhibitors: Future Perspectives and Challenges)
Show Figures

Figure 1

35 pages, 2039 KB  
Review
Recent Advances in the Clinical Translation of Small-Cell Lung Cancer Therapeutics
by Subhadeep Das and Shayak Samaddar
Cancers 2025, 17(2), 255; https://doi.org/10.3390/cancers17020255 - 14 Jan 2025
Cited by 10 | Viewed by 5347
Abstract
Small-cell lung cancer (SCLC) is a recalcitrant form of cancer, representing 15% of lung cancer cases globally. SCLC is classified within the range of neuroendocrine pulmonary neoplasms, exhibiting shared morphologic, ultrastructural, immunohistochemical, and molecular genomic features. It is marked by rapid proliferation, a [...] Read more.
Small-cell lung cancer (SCLC) is a recalcitrant form of cancer, representing 15% of lung cancer cases globally. SCLC is classified within the range of neuroendocrine pulmonary neoplasms, exhibiting shared morphologic, ultrastructural, immunohistochemical, and molecular genomic features. It is marked by rapid proliferation, a propensity for early metastasis, and an overall poor prognosis. The current conventional therapies involve platinum–etoposide-based chemotherapy in combination with immunotherapy. Nonetheless, the rapid emergence of therapeutic resistance continues to pose substantial difficulties. The genomic profiling of SCLC uncovers significant chromosomal rearrangements along with a considerable mutation burden, typically involving the functional inactivation of the tumor suppressor genes TP53 and RB1. Identifying biomarkers and evaluating new treatments is crucial for enhancing outcomes in patients with SCLC. Targeted therapies such as topoisomerase inhibitors, DLL3 inhibitors, HDAC inhibitors, PARP inhibitors, Chk1 inhibitors, etc., have introduced new therapeutic options for future applications. In this current review, we will attempt to outline the key molecular pathways that play a role in the development and progression of SCLC, together with a comprehensive overview of the most recent advancements in the development of novel targeted treatment strategies, as well as some ongoing clinical trials against SCLC, with the goal of improving patient outcomes. Full article
(This article belongs to the Special Issue The Genetic Analysis and Clinical Therapy in Lung Cancer)
Show Figures

Figure 1

17 pages, 3223 KB  
Article
Exploring the Role and Pathophysiological Significance of Aldehyde Dehydrogenase 1B1 (ALDH1B1) in Human Lung Adenocarcinoma
by Ilias Tsochantaridis, Dimitris Brisimis, Margaritis Tsifintaris, Anastasia Anastasiadou, Efthymios Lazos, Antreas Ermogenous, Sylia Christou, Nefeli Antonopoulou, Mihalis I. Panayiotidis, Michail I. Koukourakis, Alexandra Giatromanolaki and Aglaia Pappa
Int. J. Mol. Sci. 2024, 25(19), 10301; https://doi.org/10.3390/ijms251910301 - 25 Sep 2024
Cited by 5 | Viewed by 4277
Abstract
Aldehyde dehydrogenases (ALDHs) constitute a diverse superfamily of NAD(P)+-dependent enzymes pivotal in oxidizing endogenous and exogenous aldehydes to carboxylic acids. Beyond metabolic roles, ALDHs participate in essential biological processes, including differentiation, embryogenesis and the DNA damage response, while also serving as [...] Read more.
Aldehyde dehydrogenases (ALDHs) constitute a diverse superfamily of NAD(P)+-dependent enzymes pivotal in oxidizing endogenous and exogenous aldehydes to carboxylic acids. Beyond metabolic roles, ALDHs participate in essential biological processes, including differentiation, embryogenesis and the DNA damage response, while also serving as markers for cancer stem cells (CSCs). Aldehyde dehydrogenase 1B1 (ALDH1B1) is a mitochondrial enzyme involved in the detoxification of lipid peroxidation by-products and metabolism of various aldehyde substrates. This study examines the potential role of ALDH1B1 in human lung adenocarcinoma and its association with the CSC phenotype. To this end, we utilized the lung adenocarcinoma cell line A549, engineered to stably express the human ALDH1B1 protein tagged with green fluorescent protein (GFP). Overexpression of ALDH1B1 led to notable changes in cell morphology, proliferation rate and clonogenic efficiency. Furthermore, ALDH1B1-overexpressing A549 cells exhibited enhanced resistance to the chemotherapeutic agents etoposide and cisplatin. Additionally, ALDH1B1 overexpression correlated with increased migratory potential and epithelial–mesenchymal transition (EMT), mediated by the upregulation of transcription factors such as SNAI2, ZEB2 and TWIST1, alongside the downregulation of E-cadherin. Moreover, Spearman’s rank correlation coefficient analysis using data from 507 publicly available lung adenocarcinoma clinical samples revealed a significant correlation between ALDH1B1 and various molecules implicated in CSC-related signaling pathways, including Wnt, Notch, hypoxia, Hedgehog, retinoic acid, Hippo, NF-κΒ, TGF-β, PI3K/PTEN-AKT and glycolysis/gluconeogenesis. These findings provide insights into the role of ALDH1B1 in lung tumor progression and its relation to the lung CSC phenotype, thereby offering potential therapeutic targets in the clinical management of lung adenocarcinoma. Full article
Show Figures

Figure 1

8 pages, 2089 KB  
Case Report
Durable Objective Response to Lurbinectedin in Small Cell Bladder Cancer with TP53 Mutation: A Molecular-Directed Strategy
by Mohammad Jad Moussa, Jaanki Khandelwal, Nathaniel R. Wilson, Sagar A. Naik, Vivek Subbiah, Matthew T. Campbell, Pavlos Msaouel, Parminder Singh and Omar Alhalabi
Curr. Oncol. 2024, 31(6), 3342-3349; https://doi.org/10.3390/curroncol31060254 - 13 Jun 2024
Cited by 5 | Viewed by 4709
Abstract
Small cell bladder cancer (SCBC) is a rare and aggressive disease, often treated with platinum/etoposide-based chemotherapy. Key molecular drivers include the inactivation of onco-suppressor genes (TP53, RB1) and amplifications in proto-oncogenes (MYC). We report a patient with SCBC [...] Read more.
Small cell bladder cancer (SCBC) is a rare and aggressive disease, often treated with platinum/etoposide-based chemotherapy. Key molecular drivers include the inactivation of onco-suppressor genes (TP53, RB1) and amplifications in proto-oncogenes (MYC). We report a patient with SCBC who achieved an objective and prolonged response to lurbinectedin, which has been approved for metastatic small cell lung cancer, after developing disease progression on cisplatin/etoposide and nivolumab/ipilimumab. A genomic analysis of a metastatic biopsy prior to lurbinectedin initiation revealed a TP53 mutation and amplification of the cell cycle regulators E2F3 and MYCL. A repeat biopsy following the development of lurbinectedin resistance showed a new actionable ERBB2 alteration without significant change in the tumor mutation burden (six mutations/Mb). The present report suggests that lurbinectedin may be active and should be further explored in SCBC harboring TP53 mutations and amplifications in E2F3 and MYC family complexes. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Figure 1

20 pages, 5831 KB  
Article
Effects of KEAP1 Silencing on NRF2 and NOTCH Pathways in SCLC Cell Lines
by Federico Pio Fabrizio, Angelo Sparaneo, Giusy Gorgoglione, Pierpaolo Battista, Flavia Centra, Francesco Delli Muti, Domenico Trombetta, Antonella Centonza, Paolo Graziano, Antonio Rossi, Vito Michele Fazio and Lucia Anna Muscarella
Cancers 2024, 16(10), 1885; https://doi.org/10.3390/cancers16101885 - 15 May 2024
Cited by 4 | Viewed by 2433
Abstract
The KEAP1/NRF2 pathway is a master regulator of several redox-sensitive genes implicated in the resistance of tumor cells against therapeutic drugs. The dysfunction of the KEAP1/NRF2 system has been correlated with neoplastic patients’ outcomes and responses to conventional therapies. In lung tumors, the [...] Read more.
The KEAP1/NRF2 pathway is a master regulator of several redox-sensitive genes implicated in the resistance of tumor cells against therapeutic drugs. The dysfunction of the KEAP1/NRF2 system has been correlated with neoplastic patients’ outcomes and responses to conventional therapies. In lung tumors, the growth and the progression of cancer cells may also involve the intersection between the molecular NRF2/KEAP1 axis and other pathways, including NOTCH, with implications for antioxidant protection, survival of cancer cells, and drug resistance to therapies. At present, the data concerning the mechanism of aberrant NRF2/NOTCH crosstalk as well as its genetic and epigenetic basis in SCLC are incomplete. To better clarify this point and elucidate the contribution of NRF2/NOTCH crosstalk deregulation in tumorigenesis of SCLC, we investigated genetic and epigenetic dysfunctions of the KEAP1 gene in a subset of SCLC cell lines. Moreover, we assessed its impact on SCLC cells’ response to conventional chemotherapies (etoposide, cisplatin, and their combination) and NOTCH inhibitor treatments using DAPT, a γ-secretase inhibitor (GSI). We demonstrated that the KEAP1/NRF2 axis is epigenetically controlled in SCLC cell lines and that silencing of KEAP1 by siRNA induced the upregulation of NRF2 with a consequent increase in SCLC cells’ chemoresistance under cisplatin and etoposide treatment. Moreover, KEAP1 modulation also interfered with NOTCH1, HES1, and DLL3 transcription. Our preliminary data provide new insights about the downstream effects of KEAP1 dysfunction on NRF2 and NOTCH deregulation in this type of tumor and corroborate the hypothesis of a cooperation of these two pathways in the tumorigenesis of SCLC. Full article
(This article belongs to the Special Issue Unique Perspectives in Cancer Signaling)
Show Figures

Graphical abstract

7 pages, 573 KB  
Communication
Castasterone, a Plant Steroid Hormone, Affects Human Small-Cell Lung Cancer Cells and Reverses Multi-Drug Resistance
by David Sadava and Shiuan Chen
Pharmaceuticals 2023, 16(2), 170; https://doi.org/10.3390/ph16020170 - 23 Jan 2023
Cited by 6 | Viewed by 2505
Abstract
Small-cell lung cancer (SCLC) has a dismal prognosis, in part because of the development of multi-drug resistance. Castasterone (CAS) is the metabolic precursor of the plant steroid hormone epibrassinolide (EB). In some plants, EB accounts for the total hormone activity, whereas in other [...] Read more.
Small-cell lung cancer (SCLC) has a dismal prognosis, in part because of the development of multi-drug resistance. Castasterone (CAS) is the metabolic precursor of the plant steroid hormone epibrassinolide (EB). In some plants, EB accounts for the total hormone activity, whereas in other plants, CAS is the active form. The effects of CAS, a BR present in most plants, on animal cells in general and cancer cells in particular have not been described. Here, we report the effects of CAS on drug-sensitive (H69) and drug-resistant (VPA17) SCLC cells. CAS was equally cytotoxic to both cell lines (IC50 = 1 μM), indicating a lack of cross-resistance. Pre-incubation of VPA17 cells with CAS for 96 h reversed drug resistance to etoposide and doxorubicin. Synergism between CAS and EB, as well as with chemotherapy drugs, was investigated by exposure of VPA17 cells to 1:1 ratios of CAS and the other drugs at the respective IC50 values, with dilutions at 0.25 to 2.0 × IC50 and determination of the combination index (CI). CAS and EB were additive, indicating that the two drugs act on the same pathway, whereas CAS–etoposide (CI = 0.77) and CAS–doxorubicin were synergistic, indicating that CAS and the two chemotherapeutic drugs act on different pathways. Apoptosis in SCLC cells was measured by immuno-detection of single-strand DNA breaks. Following 96 h incubation of SCLC H69 cells in CAS, the level of DNA breaks was similar to measurements made after incubation in EB and etoposide, indicating that CAS is pro-apoptotic. Incubation of SCLC cells in CAS led to a time-dependent reduction (by 80%) in the transcriptional activator β-catenin. These data indicate that CAS may act via Wnt signaling. Taken together, our study reveals that CAS is pharmacologically active in both drug-sensitive and drug-resistant SCLC cells. Full article
(This article belongs to the Special Issue Natural Products for the Treatment of Lung Cancer)
Show Figures

Figure 1

20 pages, 7508 KB  
Article
Lung Adenocarcinoma Cell Sensitivity to Chemotherapies: A Spotlight on Lipid Droplets and SREBF1 Gene
by Anna Ricarda Gründing, Marc A. Schneider, Sarah Richtmann, Mark Kriegsmann, Hauke Winter, Beatriz Martinez-Delgado, Sarai Varona, Bin Liu, David S. DeLuca, Julia Held, Sabine Wrenger, Thomas Muley, Michael Meister, Tobias Welte and Sabina Janciauskiene
Cancers 2022, 14(18), 4454; https://doi.org/10.3390/cancers14184454 - 14 Sep 2022
Cited by 3 | Viewed by 3571
Abstract
To explore the relationship between cancer cell SREBF1 expression, lipid droplets (LDs) formation, and the sensitivity to chemotherapies, we cultured lung adenocarcinoma cells H1299 (with LD) and H1563 (without LD) in a serum-free basal medium (BM) or neutrophil degranulation products containing medium (NDM), [...] Read more.
To explore the relationship between cancer cell SREBF1 expression, lipid droplets (LDs) formation, and the sensitivity to chemotherapies, we cultured lung adenocarcinoma cells H1299 (with LD) and H1563 (without LD) in a serum-free basal medium (BM) or neutrophil degranulation products containing medium (NDM), and tested cell responses to cisplatin and etoposide. By using the DESeq2 Bioconductor package, we detected 674 differentially expressed genes (DEGs) associated with NDM/BM differences between two cell lines, many of these genes were associated with the regulation of sterol and cholesterol biosynthesis processes. Specifically, SREBF1 markedly declined in both cell lines cultured in NDM or when treated with chemotherapeutics. Despite the latter, H1563 exhibited LD formation and resistance to etoposide, but not to cisplatin. Although H1299 cells preserved LDs, these cells were similarly sensitive to both drugs. In a cohort of 292 patients with non-small-cell lung cancer, a lower SREBF1 expression in tumors than in adjacent nontumor tissue correlated with overall better survival, specifically in patients with adenocarcinoma at stage I. Our findings imply that a direct correlation between SREBF1 and LD accumulation can be lost due to the changes in cancer cell environment and/or chemotherapy. The role of LDs in lung cancer development and response to therapies remains to be examined in more detail. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Lung Cancer and Mesothelioma)
Show Figures

Graphical abstract

17 pages, 792 KB  
Review
Small-Cell Lung Cancer Long-Term Survivor Patients: How to Find a Needle in a Haystack?
by Andrea Plaja, Teresa Moran, Enric Carcereny, Maria Saigi, Ainhoa Hernández, Marc Cucurull and Marta Domènech
Int. J. Mol. Sci. 2021, 22(24), 13508; https://doi.org/10.3390/ijms222413508 - 16 Dec 2021
Cited by 16 | Viewed by 4724
Abstract
Small-cell lung cancer (SCLC) is an aggressive malignancy characterized by a rapid progression and a high resistance to treatments. Unlike other solid tumors, there has been a scarce improvement in emerging treatments and survival during the last years. A better understanding of SCLC [...] Read more.
Small-cell lung cancer (SCLC) is an aggressive malignancy characterized by a rapid progression and a high resistance to treatments. Unlike other solid tumors, there has been a scarce improvement in emerging treatments and survival during the last years. A better understanding of SCLC biology has allowed for the establishment of a molecular classification based on four transcription factors, and certain therapeutic vulnerabilities have been proposed. The universal inactivation of TP53 and RB1, along with the absence of mutations in known targetable oncogenes, has hampered the development of targeted therapies. On the other hand, the immunosuppressive microenvironment makes the success of immune checkpoint inhibitors (ICIs), which have achieved a modest improvement in overall survival in patients with extensive disease, difficult. Currently, atezolizumab or durvalumab, in combination with platinum–etoposide chemotherapy, is the standard of care in first-line setting. However, the magnitude of the benefit is scarce and no predictive biomarkers of response have yet been established. In this review, we describe SCLC biology and molecular classification, examine the SCLC tumor microenvironment and the challenges of predictive biomarkers of response to new treatments, and, finally, assess clinical and molecular characteristics of long-term survivor patients in order to identify possible prognostic factors and treatment vulnerabilities. Full article
Show Figures

Figure 1

14 pages, 269 KB  
Review
Evidence to Date: Evaluating Pembrolizumab in the Treatment of Extensive-Stage Small-Cell Lung Cancer
by Ivy Riano, Shruti R. Patel, Stephen V. Liu and Narjust Duma
Clin. Pract. 2021, 11(3), 441-454; https://doi.org/10.3390/clinpract11030059 - 6 Jul 2021
Cited by 3 | Viewed by 4963
Abstract
Small-cell lung cancer (SCLC) is an aggressive subtype of lung cancer characterized by a rapid initial response and early development of resistance to systemic therapy and radiation. The management of SCLC significantly changed for the first time in decades with the introduction of [...] Read more.
Small-cell lung cancer (SCLC) is an aggressive subtype of lung cancer characterized by a rapid initial response and early development of resistance to systemic therapy and radiation. The management of SCLC significantly changed for the first time in decades with the introduction of immune checkpoint inhibitors. Pembrolizumab, a humanized IgG4 isotype antibody, targets the programmed cell death protein 1 (PD-1) pathway to restore anti-tumor immunity. Prospective trials of pembrolizumab in patients with previously treated SCLC showed significant durability of responses. These results led to the U.S. Food and Drug Administration (FDA) granting pembrolizumab accelerated approval as second- or third-line monotherapy for patients with extensive-stage (ES) SCLC. In a recent clinical trial that included patients with previously untreated ES-SCLC, pembrolizumab in combination with platinum/etoposide met its progression-free survival endpoint, but overall survival (OS) did not cross the threshold for superiority. With the therapeutic landscape for SCLC rapidly evolving, we review prior experience and future directions of pembrolizumab in ES-SCLC. Full article
17 pages, 10326 KB  
Article
Ex Vivo Expansion and Drug Sensitivity Profiling of Circulating Tumor Cells from Patients with Small Cell Lung Cancer
by Hsin-Lun Lee, Jeng-Fong Chiou, Peng-Yuan Wang, Long-Sheng Lu, Chia-Ning Shen, Han-Lin Hsu, Thierry Burnouf, Lai-Lei Ting, Pai-Chien Chou, Chi-Li Chung, Kai-Ling Lee, Her-Shyong Shiah, Yen-Lin Liu and Yin-Ju Chen
Cancers 2020, 12(11), 3394; https://doi.org/10.3390/cancers12113394 - 16 Nov 2020
Cited by 38 | Viewed by 5740
Abstract
Small cell lung cancer (SCLC) represents one of the most aggressive malignancies among cancer types. Not only tumor sample availability is limited, but also the ability for tumor cells to rapidly acquire drug resistance are the rate-limiting bottlenecks for overall survival in current [...] Read more.
Small cell lung cancer (SCLC) represents one of the most aggressive malignancies among cancer types. Not only tumor sample availability is limited, but also the ability for tumor cells to rapidly acquire drug resistance are the rate-limiting bottlenecks for overall survival in current clinical settings. A liquid biopsy capable of capturing and enriching circulating tumor cells (CTCs), together with the possibility of drug screening, is a promising solution. Here, we illustrate the development of a highly efficient ex vivo CTC expansion system based on binary colloidal crystals substrate. Clinical samples were enrolled from 22 patients with SCLC in the study. The CTCs were enriched and expanded from the collected peripheral blood samples. Expanded cells were analyzed for protein expression and observed for drug sensitivity with the use of immunofluorescence and ATP titer evaluation, respectively. Successful CTC spheroid proliferation was established after 4 weeks within 82% of all the collected peripheral blood samples from enrolled patients. Upon immunofluorescence analysis, the enriched cells showed positive markers for EpCAM, TTF-1, synaptophysin and negative for CD45. Additionally, the expanded CTCs demonstrated marked heterogeneity in the expression of E-cadherin and N-cadherin. In a preliminary case series, the drug sensitivity of patient-derived CTC to cisplatin and etoposide was studied to see the correlation with the corresponding therapeutic outcome. In conclusion, our study demonstrates that it is possible to efficiently expand CTCs from SCLC within a clinically relevant time frame; the biomarker information generated from enriched CTCs can assist the selection of effective drugs and improve disease outcome. Full article
(This article belongs to the Special Issue Liquid Biopsy in Cancer)
Show Figures

Figure 1

Back to TopTop