1
|
Zapata Dongo RJ, Fontana D, Mologni L, Faya Castillo JE, Infante Varillas SF. Inhibition of the anti-apoptotic protein BCL2 in EML4-ALK cell models as a second proposed therapeutic target for non-small cell lung cancer. PLoS One 2025; 20:e0308747. [PMID: 39836700 PMCID: PMC11750102 DOI: 10.1371/journal.pone.0308747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/29/2024] [Indexed: 01/23/2025] Open
Abstract
The anaplastic lymphoma kinase (ALK) oncoprotein plays a crucial role in non-small cell lung cancer (NSCLC) by activating signaling pathways involved in cell proliferation and survival through constitutive phosphorylation. While first-line crizotinib can regulate phosphorylation, mutations in the ALK gene can lead to resistance against ALK inhibitors (ALKi) such as ceritinib and alectinib. On the other hand, overexpression of BCL2, a protein involved in cell death regulation, has been observed in NSCLC and is considered a potential therapeutic target. In this study, we propose to inhibit BCL2 as a secondary therapeutic target in EML4-ALK cell models to overcome resistance caused by ALK mutations. Four Ba/F3 EML4-ALK cell models (WT, C1156Y, L1196M, and G1202R) generated by site-directed mutagenesis exhibited varying levels of BCL2 expression. Both the WT and G1202R models showed overexpression of BCL2, while C1156Y and L1196M models approached baseline levels. We treated these cells with ABT-199, a selective BCL2 inhibitor, and found that models with high BCL2 expression exhibited resistance, while those with lower expression showed sensitivity to BCL2 inhibition. In addition, our analysis using bioinformatics indicated that ABT-199 not only targets BCL2 but also binds to the active site of all ALK mutants, it was contrasted by in vitro ALK kinase activity inhibition by ABT-199 (5.5 μM). This interaction was further supported by a significant decrease of ALK phosphorylation in single and combination treatment with 300nM ABT-199. Finally, when ABT-199 was combined with ALKi, we observed a wide range of synergistic effects in the WT and G1202R cell models, while the C1156Y and L1196M models showed limited synergy. In conclusion, our findings indicate that BCL2 targeting with ABT-199, in combination with ALKi, can significantly reduce tumor cell survival in Ba/F3 EML4-ALK cell models.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Cell Line, Tumor
- Sulfonamides/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Anaplastic Lymphoma Kinase/genetics
- Anaplastic Lymphoma Kinase/metabolism
- Anaplastic Lymphoma Kinase/antagonists & inhibitors
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Mutation
Collapse
Affiliation(s)
- Richard Junior Zapata Dongo
- Department of Basic Sciences, Bioethics and Human Life, Faculty of Human Medicine, University of Piura, Miraflores, Lima, Perú
| | - Diletta Fontana
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Juan Enrique Faya Castillo
- Department of Basic Sciences, Bioethics and Human Life, Faculty of Human Medicine, University of Piura, Miraflores, Lima, Perú
| | | |
Collapse
|
2
|
Pozonec V, Pozonec MD, Aigner C, Widder J, Boettiger K, Megyesfalvi Z, Dome B. Prophylactic cranial irradiation for small cell lung cancer in the era of immunotherapy and molecular subtypes. Curr Opin Oncol 2025; 37:27-34. [PMID: 39625049 PMCID: PMC11623382 DOI: 10.1097/cco.0000000000001111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
PURPOSE OF REVIEW Small cell lung cancer (SCLC) is an aggressive disease with a poor prognosis, whereas its metastatic capacity carries a predilection for the brain. Although prophylactic cranial irradiation (PCI) has been used to address this problem, upcoming alternatives might necessitate reflection of its application in SCLC treatment. RECENT FINDINGS The addition of immunotherapy to treatment guidelines has provided a new strategy for the management of brain metastases. Complementation of immunotherapy with active MRI surveillance could potentially replace PCI and avoid irradiation-related cognitive side effects. SCLC's molecular profile is heterogeneous, with differential response to treatment modalities between subgroups. Investigation of these variances might be essential to improve therapeutic outcomes in SCLC patients. SUMMARY The role of PCI in SCLC treatment must be examined in light of immunotherapy. We summarize recent results, bearing SCLC subtypes and therapeutic vulnerabilities in mind, to derive tailored treatment strategies for SCLC patients in future settings.
Collapse
Affiliation(s)
- Veronika Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- Multidisciplinary Centre of Head and Neck Tumors, National Institute of Oncology
| | - Maria Dorothea Pozonec
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | | | - Joachim Widder
- Department of Radiation Oncology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | | | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery
| | - Balazs Dome
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery
- Department of Translational Medicine, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Li H, Zhao P, Tian L, Lu Y, Wang X, Shao W, Cheng Y. Advances in biomarkers for immunotherapy in small-cell lung cancer. Front Immunol 2024; 15:1490590. [PMID: 39723215 PMCID: PMC11668642 DOI: 10.3389/fimmu.2024.1490590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
Small-cell lung cancer (SCLC) is a refractory cancer with rapid growth and high aggressiveness. Extensive-stage SCLC is initially sensitive to chemotherapy; however, drug resistance and recurrence occur rapidly, resulting in a poor survival outcome due to lack of subsequently efficient therapy. The emergence of immune checkpoint inhibitors (ICIs) generated a new landscape of SCLC treatment and significantly prolonged the survival of patients. However, the unselected immunotherapy restrains both beneficiary population and responsive period in SCLC compared to the other tumors. The complex tumor origin, high heterogeneity, and immunosuppressive microenvironment may disturb the value of conventional biomarkers in SCLC including programmed cell death 1 ligand 1 and tumor mutation burden. Transcriptional regulator-based subtypes of SCLC are current research hotspot, revealing that Y (I) subtype can benefit from ICIs. Additionally, molecules related to immune microenvironment, immunogenicity, epigenetics, and SCLC itself also indicated the therapeutic benefits of ICIs, becoming potential predictive biomarkers. In this review, we discussed the advances of biomarkers for prediction and prognosis of immunotherapy, promising directions in the future, and provide reference and options for precision immunotherapy and survival improvement in patients with SCLC.
Collapse
Affiliation(s)
- Hui Li
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Peiyan Zhao
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Lin Tian
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Yuanhua Lu
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Xinyue Wang
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Wenjun Shao
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Postdoctoral Research Workstation, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Medical Oncology Translational Research Lab, Jilin Cancer Hospital, Changchun, China
- Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
4
|
Tang L, Ruan Y, Wang B, Zhang M, Xue J, Wang T. Erianin inhibits the progression of DDP-resistant lung adenocarcinoma by regulating the Wnt/β-catenin pathway and activating the caspase-3 for apoptosis in vitro and in vivo. Hereditas 2024; 161:48. [PMID: 39605083 PMCID: PMC11600767 DOI: 10.1186/s41065-024-00351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Platinum-based chemotherapy is one of the main treatments for lung adenocarcinoma (LUAD). However, the toxic side effects and drug resistance of chemotherapeutic drugs on normal cells are still a thorny problem in clinical treatment. Dendrobium is one of the three largest genera of Orchidaceous family, which has ornamental and medicinal value. Dendrobium is mainly distributed in the tropics and subtropics of South Asia, Oceania and other regions, with 1547 species of Dendrobium currently known. In China, "Shi hu" and "Tie pi shi hu" are well-known traditional medicines and have been included in the Chinese Pharmacopoeia (Editorial Board of Chinese Pharmacopoeia, 2020). Erianin is a natural product isolated from Dendrobium and is considered as a potential anticancer molecule due to its remarkable anti-tumor effects through various mechanisms, among which induced cancer cell apoptosis, inhibited invasion and migration. This study preliminarily explored the mechanism of Erianin inhibiting the progression of cisplatin (DDP) resistant LUAD in vivo and in vitro. METHODS The effect of Erianin on the proliferation of DDP-resistant LUAD cells was detected by CCK-8 assay, wound healing assay and cloning assay. Transwell assay was used to evaluate the effect of Erianin on cell invasion and migration. The changes of cell cycle and apoptosis were detected by flow cytometry and TUNEL assay. Finally, the effects of Erianin on cell function and signaling pathway-related protein expression in vivo and in vitro were examined based on the enrichment analysis. RESULTS Erianin could inhibit the proliferation, invasion and migration, induce apoptosis, altered cell cycle of DDP-resistant LUAD cells, and reverse the resistance to DDP. Western blotting results showed that Erianin exerted its anti-tumor effects by regulating the Wnt/β-catenin cascade in DDP-resistant LUAD cells. CONCLUSION Erianin may exerted its anti-tumor effect in DDP-resistant LUAD cells by regulating the Wnt3/β-Catenin/Survivin/Bcl-2/Caspase-3/Cyclin D1 axis.
Collapse
Affiliation(s)
- Lingxue Tang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiling Ruan
- Department of General practice, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Beibei Wang
- Department of General practice, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mingjun Zhang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jie Xue
- Department of General Practice, Suixi County Hospital, Huaibei, Anhui, China.
| | - Tong Wang
- Department of General practice, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
5
|
Jiang Y, Xie J, Cheng Q, Cai Z, Xu K, Lu W, Wang F, Wu X, Song Y, Lv T, Zhan P. Comprehensive genomic and spatial immune infiltration analysis of survival outliers in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy. Int Immunopharmacol 2024; 141:112901. [PMID: 39151386 DOI: 10.1016/j.intimp.2024.112901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/20/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND A minority of patients with extensive-stage small cell lung cancer (ES-SCLC) exhibit prolonged survival following first-line chemoimmunotherapy, which warrants the use of reliable biomarkers. Here, we investigated the disparities in genomics and immune cell spatial distribution between long- and short-term survival of patients with ES-SCLC. METHODS We retrospectively recruited 11 long-term (>2 years) and 13 short-term (<9 months) ES-SCLC survivors receiving first-line chemoimmunotherapy. The samples were processed using targeted next-generation sequencing (tNGS), programmed death ligand-1 staining, multiplex immunohistochemical staining for immune cells (mIHC), tumor mutation burden (TMB), and chromosomal instability score measurements. The expression of putative genes in SCLC at the bulk and single-cell RNA-sequencing levels, as well as the role of putative genes in pan-cancer immunotherapy cohorts, were analyzed. RESULTS At the genomic level, a greater proportion of the smoking signature and higher TMB (>3.1) were associated with favorable survival. At the single-gene and pathway levels, tNGS revealed that MCL1 and STMN1 amplification and alterations in the apoptosis pathway were more common in short-term survivors, whereas alterations in the DLL3, KMT2B, HGF, EPHA3, ADGRB3, lysine deprivation, and HGF-cMET pathways were observed more frequently in long-term survivors. mIHC analysis of immune cells with different spatial distributions revealed that long-term survivors presented increased numbers of M1-like macrophages in all locations and decreased numbers of CD8+ T cells in the tumor stroma. Bulk transcriptomic analysis demonstrated that high levels of STMN1 and DLL3 represented an immune-suppressive tumor immune microenvironment (TIME), whereas HGF indicated an immune-responsive TIME. The expression levels of our putative genes were comparative in both TP53/RB1 mutant-type and TP53/RB1 wild-type. At the single-cell level, STMN1, MCL1, and DLL3 were highly expressed among all molecular subtypes (SCLC-A, SCLC-N, and SCLC-P), with STMN1 being enriched in cell division and G2M checkpoint pathways. CONCLUSIONS For ES-SCLC patients receiving first-line chemoimmunotherapy, alterations in DLL3, KMT2B, HGF, EPHA3, and ADGRB3 and a greater proportion of M1-like macrophages infiltration in all locations were predictors of favorable survival, while MCL1 and STMN1 amplification, as well as a greater proportion of CD8+ T cells infiltrating the tumor stroma, predicted worse survival.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing 210000, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zijing Cai
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China
| | - Ke Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoying Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yong Song
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| |
Collapse
|
6
|
Li S, Lv J, Li Z, Zhang Q, Lu J, Huo X, Guo M, Liu X, Li C, Wang J, Shi H, Deng L, Chen Z, Du X. Overcoming multi-drug resistance in SCLC: a synergistic approach with venetoclax and hydroxychloroquine targeting the lncRNA LYPLAL1-DT/BCL2/BECN1 pathway. Mol Cancer 2024; 23:243. [PMID: 39478582 PMCID: PMC11526623 DOI: 10.1186/s12943-024-02145-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) stands as one of the most lethal malignancies, characterized by a grim diagnosis and prognosis. The emergence of multi-drug resistance poses a significant hurdle to effective therapy. Although previous studies have implicated the long noncoding RNA LYPLAL1-DT in the tumorigenesis of SCLC, the precise role of the highly expressed LYPLAL1-DT in SCLC chemoresistance and the underlying mechanism remain inadequately understood. METHODS cDDP-, VP-16- and PTX-resistant SCLC cells lines were established. The viabilities of SCLC cells were assessed by CCK-8 assay in vitro and xenograft tumor formation assay in vivo. Apoptosis was evaluated by FACS, Western blot and JC-1 fluorescence staining, while autophagy was explored via autophagic flux detection under confocal microscopy and autophagic vacuole investigation under transmission electron microscopy (TEM). The functional role and mechanism of LYPLAL1-DT were further investigated by gain- and loss-of-function assays in vitro. Furthermore, the therapeutic efficacy of the combination of venetoclax and HCQ with cDDP, VP-16 or PTX was evaluated by cell line, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mice model. RESULTS Our findings revealed that LYPLAL1-DT is upregulated in chemoresistant SCLC cell lines. Gain- and loss-of-function assays demonstrated that LYPLAL1-DT impairs sensitivity to cDDP, VP-16, or PTX both in vitro and in vivo. Overexpression of LYPLAL1-DT significantly enhanced autophagy and inhibited apoptosis in SCLC cells. Further analyses, including RIP and RNA pull-down assays, revealed that LYPLAL1-DT promotes the expression of BCL2 by sponging miR-204-5p and is implicated in the assembly of the autophagy-specific complex (BECN1/PtdIns3K complex). Combining venetoclax and HCQ with cDDP, VP-16, or PTX effectively mitigated chemoresistance in SCLC cells and suppressed tumor growth in CDX and PDX models without inducing obvious toxic effects. CONCLUSIONS Our findings demonstrate that upregulation of LYPLAL1-DT sequesters apoptosis through the LYPLAL1-DT/miR-204-5p/BCL2 axis and promotes autophagy by facilitating the assembly of the BECN1/PtdIns3K complex, thereby mediating multi-drug resistance of SCLC. The triple combination of venetoclax, HCQ, in conjunction with cDDP, VP-16 or PTX overcomes refractory SCLC, shedding light on a potential therapeutic target for combating SCLC chemoresistance.
Collapse
Affiliation(s)
- Shuxin Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jianyi Lv
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Zhihui Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Qiuyu Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jing Lu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xueyun Huo
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Meng Guo
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xin Liu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Changlong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Hanping Shi
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Li Deng
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Zhenwen Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xiaoyan Du
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.
| |
Collapse
|
7
|
Yu T, Lok BH. Strategies to Target Chemoradiotherapy Resistance in Small Cell Lung Cancer. Cancers (Basel) 2024; 16:3438. [PMID: 39456533 PMCID: PMC11506711 DOI: 10.3390/cancers16203438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Small cell lung cancer (SCLC) is a lethal form of lung cancer with few treatment options and a high rate of relapse. While SCLC is initially sensitive to first-line DNA-damaging chemo- and radiotherapy, relapse disease is almost universally therapy-resistant. As a result, there has been interest in understanding the mechanisms of therapeutic resistance in this disease. Conclusions: Progress has been made in elucidating these mechanisms, particularly as they relate to the DNA damage response and SCLC differentiation and transformation, leading to many clinical trials investigating new therapies and combinations. Yet there remain many gaps in our understanding, such as the effect of epigenetics or the tumor microenvironment on treatment response, and no single mechanism has been found to be ubiquitous, suggesting a significant heterogeneity in the mechanisms of acquired resistance. Nevertheless, the advancement of techniques in the laboratory and the clinic will improve our ability to study this disease, especially in patient populations, and identify methods to surmount therapeutic resistance.
Collapse
Affiliation(s)
- Tony Yu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Benjamin H. Lok
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, 6 Queen’s Park Crescent, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
8
|
Jiang Y, Chen Y, Cheng Q, Lu W, Li Y, Zuo X, Wu Q, Wang X, Zhang F, Wang D, Wang Q, Lv T, Song Y, Zhan P. A random survival forest-based pathomics signature classifies immunotherapy prognosis and profiles TIME and genomics in ES-SCLC patients. Cancer Immunol Immunother 2024; 73:241. [PMID: 39358575 PMCID: PMC11448477 DOI: 10.1007/s00262-024-03829-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a highly aggressive neuroendocrine tumor with high mortality, and only a limited subset of extensive-stage SCLC (ES-SCLC) patients demonstrate prolonged survival under chemoimmunotherapy, which warrants the exploration of reliable biomarkers. Herein, we built a machine learning-based model using pathomics features extracted from hematoxylin and eosin (H&E)-stained images to classify prognosis and explore its potential association with genomics and TIME. METHODS We retrospectively recruited ES-SCLC patients receiving first-line chemoimmunotherapy at Nanjing Jinling Hospital between April 2020 and August 2023. Digital H&E-stained whole-slide images were acquired, and targeted next-generation sequencing, programmed death ligand-1 staining, and multiplex immunohistochemical staining for immune cells were performed on a subset of patients. A random survival forest (RSF) model encompassing clinical and pathomics features was established to predict overall survival. The function of putative genes was assessed via single-cell RNA sequencing. RESULTS AND CONCLUSION During the median follow-up period of 12.12 months, 118 ES-SCLC patients receiving first-line immunotherapy were recruited. The RSF model utilizing three pathomics features and liver metastases, bone metastases, smoking status, and lactate dehydrogenase, could predict the survival of first-line chemoimmunotherapy in patients with ES-SCLC with favorable discrimination and calibration. Underlyingly, the higher RSF-Score potentially indicated more infiltration of CD8+ T cells in the stroma as well as a greater probability of MCL-1 amplification and EP300 mutation. At the single-cell level, MCL-1 was associated with TNFA-NFKB signaling and apoptosis-related processes. Hopefully, this noninvasive model could act as a biomarker for immunotherapy, potentially facilitating precision medicine in the management of ES-SCLC.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing, 210000, China
| | - Yueying Chen
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Yu Li
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China
| | - Xueying Zuo
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Qiuxia Wu
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210002, China
| | - Xiaoxia Wang
- Department of Pathology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, School of Medicine, Jinling Hospital, Southeast University, 305 Zhongshan East Road, Nanjing, 210002, China
| | - Dong Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China
- Department of Respiratory and Critical Care Medicine, School of Medicine, Jinling Hospital, Southeast University, 305 Zhongshan East Road, Nanjing, 210002, China
| | - Qin Wang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing, 210000, China.
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, School of Medicine, Jinling Hospital, Southeast University, 305 Zhongshan East Road, Nanjing, 210002, China.
| | - Yong Song
- School of Medicine, Southeast University, Nanjing, 210000, China.
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, School of Medicine, Jinling Hospital, Southeast University, 305 Zhongshan East Road, Nanjing, 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing, 210000, China.
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing, 210002, China.
- Department of Respiratory and Critical Care Medicine, School of Medicine, Jinling Hospital, Southeast University, 305 Zhongshan East Road, Nanjing, 210002, China.
| |
Collapse
|
9
|
Sen T, Takahashi N, Chakraborty S, Takebe N, Nassar AH, Karim NA, Puri S, Naqash AR. Emerging advances in defining the molecular and therapeutic landscape of small-cell lung cancer. Nat Rev Clin Oncol 2024; 21:610-627. [PMID: 38965396 DOI: 10.1038/s41571-024-00914-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/06/2024] [Indexed: 07/06/2024]
Abstract
Small-cell lung cancer (SCLC) has traditionally been considered a recalcitrant cancer with a dismal prognosis, with only modest advances in therapeutic strategies over the past several decades. Comprehensive genomic assessments of SCLC have revealed that most of these tumours harbour deletions of the tumour-suppressor genes TP53 and RB1 but, in contrast to non-small-cell lung cancer, have failed to identify targetable alterations. The expression status of four transcription factors with key roles in SCLC pathogenesis defines distinct molecular subtypes of the disease, potentially enabling specific therapeutic approaches. Overexpression and amplification of MYC paralogues also affect the biology and therapeutic vulnerabilities of SCLC. Several other attractive targets have emerged in the past few years, including inhibitors of DNA-damage-response pathways, epigenetic modifiers, antibody-drug conjugates and chimeric antigen receptor T cells. However, the rapid development of therapeutic resistance and lack of biomarkers for effective selection of patients with SCLC are ongoing challenges. Emerging single-cell RNA sequencing data are providing insights into the plasticity and intratumoural and intertumoural heterogeneity of SCLC that might be associated with therapeutic resistance. In this Review, we provide a comprehensive overview of the latest advances in genomic and transcriptomic characterization of SCLC with a particular focus on opportunities for translation into new therapeutic approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Nobuyuki Takahashi
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Naoko Takebe
- Developmental Therapeutics Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Amin H Nassar
- Division of Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Nagla A Karim
- Inova Schar Cancer Institute Virginia, Fairfax, VA, USA
| | - Sonam Puri
- Division of Medical Oncology, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Abdul Rafeh Naqash
- Medical Oncology/ TSET Phase 1 program, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
10
|
Kai J, Kang K, Jiang Z, Xiong F, Wang S. Capivasertib reverses chemotherapy-induced esophageal cancer resistance via inhibiting Akt-associated Mcl-1 upregulation. Heliyon 2024; 10:e33567. [PMID: 39050467 PMCID: PMC11266993 DOI: 10.1016/j.heliyon.2024.e33567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
The development of resistance to chemotherapy in esophageal cancer represents a significant challenge in cancer treatment. Therefore, our study aimed to identify effective therapeutic strategies by examining the molecules involved in this chemoresistance. We consistently observed an increase in the expression of Mcl-1 in cells exposed to both short and long-term treatment with cisplatin, a drug commonly used in esophageal cancer therapy. Functional analysis showed that Mcl-1 regulates esophageal cancer cell response to cisplatin treatment. Notably, this upregulation of Mcl-1 was not dependent on eukaryotic initiation factor 4E (eIF4E). Instead, it was associated with increased stability due to the activation of Akt. Capivasertib, a potent pan-Akt kinase drug, significantly decreased Mcl-1 level via inhibiting Akt signaling pathway in chemo-resistant cells. In addition, capivasertib not only decreased the viability of chemo-resistant esophageal cancer cells but also synergistically enhanced the effects of cisplatin. In multiple mouse models, representing both chemo-resistant and chemo-sensitive esophageal cancer, capivasertib administered at non-toxic doses demonstrated remarkable efficacy. It significantly extended the overall survival of the mice. Our research underscores the pivotal role of Akt-associated Mcl-1 upregulation in the development of chemo-resistance in esophageal cancer cells. Furthermore, it highlights the potential of capivasertib to reverse this resistance mechanism.
Collapse
Affiliation(s)
| | | | - Zhixiao Jiang
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| | - Fei Xiong
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| | - Sheng Wang
- Department of Thoracic Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Load, Hongshan District, Wuhan, 430079, China
| |
Collapse
|
11
|
Khan S, Cao L, Wiegand J, Zhang P, Zajac-Kaye M, Kaye FJ, Zheng G, Zhou D. PROTAC-Mediated Dual Degradation of BCL-xL and BCL-2 Is a Highly Effective Therapeutic Strategy in Small-Cell Lung Cancer. Cells 2024; 13:528. [PMID: 38534371 PMCID: PMC10968744 DOI: 10.3390/cells13060528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
BCL-xL and BCL-2 are validated therapeutic targets in small-cell lung cancer (SCLC). Targeting these proteins with navitoclax (formerly ABT263, a dual BCL-xL/2 inhibitor) induces dose-limiting thrombocytopenia through on-target BCL-xL inhibition in platelets. Therefore, platelet toxicity poses a barrier in advancing the clinical translation of navitoclax. We have developed a strategy to selectively target BCL-xL in tumors, while sparing platelets, by utilizing proteolysis-targeting chimeras (PROTACs) that hijack the cellular ubiquitin proteasome system for target ubiquitination and subsequent degradation. In our previous study, the first-in-class BCL-xL PROTAC, called DT2216, was shown to have synergistic antitumor activities when combined with venetoclax (formerly ABT199, BCL-2-selective inhibitor) in a BCL-xL/2 co-dependent SCLC cell line, NCI-H146 (hereafter referred to as H146), in vitro and in a xenograft model. Guided by these findings, we evaluated our newly developed BCL-xL/2 dual degrader, called 753b, in three BCL-xL/2 co-dependent SCLC cell lines and the H146 xenograft models. 753b was found to degrade both BCL-xL and BCL-2 in these cell lines. Importantly, it was considerably more potent than DT2216, navitoclax, or DT2216 + venetoclax in reducing the viability of BCL-xL/2 co-dependent SCLC cell lines in cell culture. In vivo, 5 mg/kg weekly dosing of 753b was found to lead to significant tumor growth delay, similar to the DT2216 + venetoclax combination in H146 xenografts, by degrading both BCL-xL and BCL-2. Additionally, 753b administration at 5 mg/kg every four days induced tumor regressions. At this dosage, 753b was well tolerated in mice, without observable induction of severe thrombocytopenia as seen with navitoclax, and no evidence of significant changes in mouse body weights. These results suggest that the BCL-xL/2 dual degrader could be an effective and safe therapeutic for a subset of SCLC patients, warranting clinical trials in future.
Collapse
Affiliation(s)
- Sajid Khan
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lin Cao
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Janet Wiegand
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Maria Zajac-Kaye
- Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Frederic J. Kaye
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
12
|
Khan S, Cao L, Wiegand J, Zhang P, Zajac-Kaye M, Kaye FJ, Zheng G, Zhou D. PROTAC-mediated dual degradation of BCL-xL and BCL-2 is a highly effective therapeutic strategy in small-cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582353. [PMID: 38464204 PMCID: PMC10925307 DOI: 10.1101/2024.02.27.582353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
BCL-xL and BCL-2 are validated therapeutic targets in small-cell lung cancer (SCLC). Targeting these proteins with navitoclax (formerly ABT263, a dual BCL-xL/2 inhibitor) induces dose-limiting thrombocytopenia through on-target BCL-xL inhibition in platelets. Therefore, platelet toxicity poses a barrier in advancing the clinical translation of navitoclax. We have developed a strategy to selectively target BCL-xL in tumors, while sparing platelets, by utilizing proteolysis-targeting chimeras (PROTACs) that hijack the cellular ubiquitin proteasome system for target ubiquitination and subsequent degradation. In our previous study, the first-in-class BCL-xL PROTAC, called DT2216, was shown to have synergistic antitumor activities when combined with venetoclax (formerly ABT199, BCL-2-selective inhibitor) in a BCL-xL/2 co-dependent SCLC cell line, NCI-H146 (hereafter referred to as H146), in vitro and in a xenograft model. Guided by these findings, we evaluated our newly developed BCL-xL/2 dual degrader, called 753b, in three BCL-xL/2 co-dependent SCLC cell lines and the H146 xenograft models. 753b was found to degrade both BCL-xL and BCL-2 in these cell lines. Importantly, it was considerably more potent than DT2216, navitoclax, or DT2216+venetoclax to reduce the viability of BCL-xL/2 co-dependent SCLC cell lines in cell culture. In vivo, 5 mg/kg weekly dosing of 753b leads to significant tumor growth delay similar to the DT2216+venetoclax combination in H146 xenografts by degrading both BCL-xL and BCL-2. Additionally, 753b administration at 5 mg/kg every four days induced tumor regressions. 753b at this dosage was well tolerated in mice without induction of severe thrombocytopenia as seen with navitoclax nor induced significant changes in mouse body weights. These results suggest that the BCL-xL/2 dual degrader could be an effective and safe therapeutic for a subset of SCLC patients warranting clinical trials in future.
Collapse
Affiliation(s)
- Sajid Khan
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lin Cao
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Janet Wiegand
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Maria Zajac-Kaye
- Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Frederic J. Kaye
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Daohong Zhou
- Department of Biochemistry & Structural Biology, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
13
|
Solta A, Ernhofer B, Boettiger K, Megyesfalvi Z, Heeke S, Hoda MA, Lang C, Aigner C, Hirsch FR, Schelch K, Döme B. Small cells - big issues: biological implications and preclinical advancements in small cell lung cancer. Mol Cancer 2024; 23:41. [PMID: 38395864 PMCID: PMC10893629 DOI: 10.1186/s12943-024-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
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.
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Thoracic Oncology, Mount Sinai Health System, Tisch Cancer Institute, New York, NY, USA.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balazs Döme
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Wyżewski Z, Stępkowska J, Kobylińska AM, Mielcarska A, Mielcarska MB. Mcl-1 Protein and Viral Infections: A Narrative Review. Int J Mol Sci 2024; 25:1138. [PMID: 38256213 PMCID: PMC10816053 DOI: 10.3390/ijms25021138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
MCL-1 is the prosurvival member of the Bcl-2 family. It prevents the induction of mitochondria-dependent apoptosis. The molecular mechanisms dictating the host cell viability gain importance in the context of viral infections. The premature apoptosis of infected cells could interrupt the pathogen replication cycle. On the other hand, cell death following the effective assembly of progeny particles may facilitate virus dissemination. Thus, various viruses can interfere with the apoptosis regulation network to their advantage. Research has shown that viral infections affect the intracellular amount of MCL-1 to modify the apoptotic potential of infected cells, fitting it to the "schedule" of the replication cycle. A growing body of evidence suggests that the virus-dependent deregulation of the MCL-1 level may contribute to several virus-driven diseases. In this work, we have described the role of MCL-1 in infections caused by various viruses. We have also presented a list of promising antiviral agents targeting the MCL-1 protein. The discussed results indicate targeted interventions addressing anti-apoptotic MCL1 as a new therapeutic strategy for cancers as well as other diseases. The investigation of the cellular and molecular mechanisms involved in viral infections engaging MCL1 may contribute to a better understanding of the regulation of cell death and survival balance.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University in Warsaw, Dewajtis 5, 01-815 Warsaw, Poland
| | - Justyna Stępkowska
- Institute of Family Sciences, Cardinal Stefan Wyszyński University in Warsaw, Dewajtis 5, 01-815 Warsaw, Poland;
| | - Aleksandra Maria Kobylińska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.M.K.); (M.B.M.)
| | - Adriana Mielcarska
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children’s Memorial Health Institute, Av. Dzieci Polskich 20, 04-730 Warsaw, Poland;
| | - Matylda Barbara Mielcarska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.M.K.); (M.B.M.)
| |
Collapse
|
15
|
Solta A, Boettiger K, Kovács I, Lang C, Megyesfalvi Z, Ferk F, Mišík M, Hoetzenecker K, Aigner C, Kowol CR, Knasmueller S, Grusch M, Szeitz B, Rezeli M, Dome B, Schelch K. Entinostat Enhances the Efficacy of Chemotherapy in Small Cell Lung Cancer Through S-phase Arrest and Decreased Base Excision Repair. Clin Cancer Res 2023; 29:4644-4659. [PMID: 37725585 PMCID: PMC10644001 DOI: 10.1158/1078-0432.ccr-23-1795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/10/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE Acquired chemoresistance is a frequent event in small cell lung cancer (SCLC), one of the deadliest human malignancies. Histone deacetylase inhibitors (HDACi) have been shown to synergize with different chemotherapeutic agents including cisplatin. Accordingly, we aimed to investigate the dual targeting of HDAC inhibition and chemotherapy in SCLC. EXPERIMENTAL DESIGN The efficacy of HDACi and chemotherapy in SCLC was investigated both in vitro and in vivo. Synergistic drug interactions were calculated based on the HSA model (Combenefit software). Results from the proteomic analysis were confirmed via ICP-MS, cell-cycle analysis, and comet assays. RESULTS Single entinostat- or chemotherapy significantly reduced cell viability in human neuroendocrine SCLC cells. The combination of entinostat with either cisplatin, carboplatin, irinotecan, epirubicin, or etoposide led to strong synergy in a subset of resistant SCLC cells. Combination treatment with entinostat and cisplatin significantly decreased tumor growth in vivo. Proteomic analysis comparing the groups of SCLC cell lines with synergistic and additive response patterns indicated alterations in cell-cycle regulation and DNA damage repair. Cell-cycle analysis revealed that cells exhibiting synergistic drug responses displayed a shift from G1 to S-phase compared with cells showing additive features upon dual treatment. Comet assays demonstrated more DNA damage and decreased base excision repair in SCLC cells more responsive to combination therapy. CONCLUSIONS In this study, we decipher the molecular processes behind synergistic interactions between chemotherapy and HDAC inhibition. Moreover, we report novel mechanisms to overcome drug resistance in SCLC, which may be relevant to increasing therapeutic success.
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Ildikó Kovács
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - Franziska Ferk
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Miroslav Mišík
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R. Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | | | - Michael Grusch
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, Hungary
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Balazs Dome
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- Department of Translational Medicine, Lund University, Lund, Sweden
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|