251
|
Mambetsariev I, Arvanitis L, Fricke J, Pharaon R, Baroz AR, Afkhami M, Koczywas M, Massarelli E, Salgia R. Small Cell Lung Cancer Transformation following Treatment in EGFR-Mutated Non-Small Cell Lung Cancer. J Clin Med 2022; 11:1429. [PMID: 35268520 PMCID: PMC8911080 DOI: 10.3390/jcm11051429] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 03/02/2022] [Indexed: 01/21/2023] Open
Abstract
EGFR-mutated lung adenocarcinoma patients who received tyrosine kinase inhibitors (TKIs) may initially respond to therapy, but over time, resistance eventually occurs. In a small population (5-10%), these patients can have a histological transformation to SCLC. Nine patients with EGFR-mutated lung adenocarcinoma who transformed to SCLC were evaluated at City of Hope. Patient clinical and pathology data, including multiple next-generation sequencing (NGS) results, clinical therapies, histology, and outcomes, were collected across multiple time points. Descriptive statistics were utilized to visualize and interpret the clinical therapeutic timeline and molecular transformation profiles for these patients. All patients received at least one line of EGFR TKI therapies prior to small cell lung cancer transformation, including erlotinib, afatinib, and osimertinib. Two patients also received chemotherapy prior to transformation (one with immunotherapy). The median months to small cell lung cancer transformation was 16 months, ranging from 4-49 months. The median overall survival (OS) was 29 months from diagnosis, with the minimum of 16 months and maximum of 62 months. The majority of patients had EGFR exon 19 deletion (n = 7, 77.8%), and no patients had a change of original oncogenic EGFR mutation over the different time points. Though a TP53 mutation was detected in eight patients (88.9%) either at the first biopsy or the subsequent biopsies, an RB1 alteration was only detected in one patient at presentation, and three patients upon subsequent biopsies (n = 4, 44.4%). Each patient had a unique molecular profile in the subsequent molecular testing post-transformation, but BRAF alterations occurred frequently, including BRAF rearrangement (n = 1), fusion (n = 1), and amplification (n = 1). Our results showed that EGFR-mutated lung adenocarcinoma to SCLC transformation patients have a unique histological, molecular, and clinical profile over multiple time points, with further heterogeneity that is not currently reported in the literature, and we suggest more work is required to better understand the molecular heterogeneity and clinical outcomes over time for this EGFR TKI resistance subtype.
Collapse
Affiliation(s)
- Isa Mambetsariev
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Leonidas Arvanitis
- Department of Pathology, City of Hope, Duarte, CA 91010, USA; (L.A.); (M.A.)
| | - Jeremy Fricke
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Rebecca Pharaon
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Angel R. Baroz
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Michelle Afkhami
- Department of Pathology, City of Hope, Duarte, CA 91010, USA; (L.A.); (M.A.)
| | - Marianna Koczywas
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Erminia Massarelli
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA 91010, USA; (I.M.); (J.F.); (R.P.); (A.R.B.); (M.K.); (E.M.)
| |
Collapse
|
252
|
Radfar P, Aboulkheyr Es H, Salomon R, Kulasinghe A, Ramalingam N, Sarafraz-Yazdi E, Thiery JP, Warkiani ME. Single-cell analysis of circulating tumour cells: enabling technologies and clinical applications. Trends Biotechnol 2022; 40:1041-1060. [DOI: 10.1016/j.tibtech.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/26/2022]
|
253
|
Abstract
Small cell lung cancer (SCLC) is a rapidly growing, highly metastatic, and relatively immune-cold lung cancer subtype. Historically viewed in the laboratory and clinic as a single disease, new discoveries suggest that SCLC comprises multiple molecular subsets. Expression of MYC family members and lineage-related transcription factors ASCL1, NEUROD1, and POU2F3 (and, in some studies, YAP1) define unique molecular states that have been associated with distinct responses to a variety of therapies. However, SCLC tumors exhibit a high degree of intratumoral heterogeneity, with recent studies suggesting the existence of tumor cell plasticity and phenotypic switching between subtype states. While SCLC plasticity is correlated with, and likely drives, therapeutic resistance, the mechanisms underlying this plasticity are still largely unknown. Subtype states are also associated with immune-related gene expression, which likely impacts response to immune checkpoint blockade and may reveal novel targets for alternative immunotherapeutic approaches. In this review, we synthesize recent discoveries on the mechanisms of SCLC plasticity and how these processes may impinge on antitumor immunity.
Collapse
Affiliation(s)
- Kate D Sutherland
- Australian Cancer Research Foundation (ACRF) Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Abbie S Ireland
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Trudy G Oliver
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| |
Collapse
|
254
|
Mao X, Liu J, Hu F, Niu Y, Pan F, Fu X, Jiang L. Serum NSE is Early Marker of Transformed Neuroendocrine Tumor After EGFR-TKI Treatment of Lung Adenocarcinoma. Cancer Manag Res 2022; 14:1293-1302. [PMID: 35386183 PMCID: PMC8978576 DOI: 10.2147/cmar.s349082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background Transformation to a lung neuroendocrine tumor (LNET) is a mechanism of resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI). Serum neuron-specific enolase (NSE) is a useful marker in the detection of LNET. Therefore, we explored the clinical significance of serum NSE levels in the detection of transformed neuroendocrine tumors after EGFR-TKI therapy. Methods We report a cohort of 5 cases in our treatment group. The characteristics of the patients, pathological diagnoses, immunohistochemistry with molecular detection, laboratory examination, and treatment histories are analyzed. The tumor markers of serum NSE were analyzed. Additionally, we reviewed the publications reporting the tumor markers before and after LNET transformation during EGFR-TKI therapy. Results Most patients are female (3/5), aged <60 years old (4/5), nonsmokers (4/5) and harbor the EGFR 19 exon deletion (4/5). The median time of LNET transformation was 19 months (range: 12–31 months). The clinical characteristics were similar to those reported in previous studies. Laboratory examination revealed an increased NSE level before the LNET is defined. Sixteen publications were reviewed. Of those, 86.67% (13/15) publications showed an increased level of NSE when the LNET transformation was defined. Conclusion Adenocarcinoma tumors in non-smokers, young patients harboring the EGFR 19 exon deletion tended to transform to LNETs after EGFR-TKI therapy. Combining our findings and a review of the literature, we suggest that serum NSE may be a useful tumor marker to predict neuroendocrine tumor transformation.
Collapse
Affiliation(s)
- Xiaowei Mao
- Pulmonary and Critical Care Medicine, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiabing Liu
- Department of Radiation Oncology, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
| | - Fang Hu
- Pulmonary and Critical Care Medicine, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
| | - Yanjie Niu
- Pulmonary and Critical Care Medicine, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
| | - Feng Pan
- Pulmonary and Critical Care Medicine, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
| | - Liyan Jiang
- Pulmonary and Critical Care Medicine, Shanghai Jiao Tong University, Shanghai Chest Hospital, Shanghai, People’s Republic of China
- Correspondence: Liyan Jiang; Xiaolong Fu, Email ;
| |
Collapse
|
255
|
Xu J, Xu L, Wang B, Kong W, Chen Y, Yu Z. Outcomes in Patients With Lung Adenocarcinoma With Transformation to Small Cell Lung Cancer After EGFR Tyrosine Kinase Inhibitors Resistance: A Systematic Review and Pooled Analysis. Front Oncol 2022; 11:766148. [PMID: 35223450 PMCID: PMC8867701 DOI: 10.3389/fonc.2021.766148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022] Open
Abstract
Background Lung adenocarcinoma can transform into small-cell lung cancer (SCLC) when resistance to tyrosine kinase inhibitors (TKIs) develops. Approximately 3% to 10% of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) could transform to SCLC. This phenomenon has been described in several case reports and small patient series. However, the characteristics and treatment outcomes of this population have not been comprehensively reported, and their clinical course is poorly characterized. Methods We performed a systematic review of the published literature to summarize the clinical and pathological features and prognosis of the reported cases and analyzed the demographics, disease features, and outcomes. Results A total of 72 patients (50 females and 22 males) initially diagnosed with lung adenocarcinoma were included. EGFR mutations included 19-deletion (75%), L858R (22%), and G719X (3%). All patients received EGFR-TKIs before SCLC transformation. The median time from diagnosis to transformation was 20.5 months (95% CI, 15.45 to 26.55 months). Of the 67 patients with post-translational gene test results, 58 maintained their EGFR mutation, and only 1 of 18 with prior T790M positivity retained T790M mutation. After the pathological transformation, both conventional chemotherapy regimen and chemotherapy combined targeted therapy yielded high response rates. The disease control rate of first-line therapy after transformation was 76%, while the objective response rate was 48%. The median overall survival (OS) since diagnosis was 27 months (95% CI, 22.90 to 31.10 months), whereas median OS since SCLC transformation was 8.5 months (95% CI, 5.50 to 11.60 months). Conclusion The prognosis of transformed SCLC is worse than primary SCLC. The response rate to conventional chemotherapy was high. However, the progression-free survival and OS after transformation were short and the prognosis was poor with first-line therapies. New therapies are needed in the management of transformed SCLC.
Collapse
Affiliation(s)
- Jinhe Xu
- Fu Zong Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Lihuan Xu
- Fu Zong Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Baoshan Wang
- Department of Gastroenterology, Dongfang Hospital of Xiamen University, Fuzhou General Hospital of Fujian Medical University, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Wencui Kong
- Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, Department of Respiratory and Critical Care Medicine, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Ying Chen
- Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, Department of Respiratory and Critical Care Medicine, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| | - Zongyang Yu
- Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, Department of Respiratory and Critical Care Medicine, The 900th Hospital of the Joint Logistic Support Force, PLA, Fuzhou, China
| |
Collapse
|
256
|
Chen Y, Yang H, Cheng Z, Chen L, Peng S, Wang J, Yang M, Lin C, Chen Y, Wang Y, Huang L, Chen Y, Li W, Ke Z. A whole-slide image (WSI)-based immunohistochemical feature prediction system improves the subtyping of lung cancer. Lung Cancer 2022; 165:18-27. [PMID: 35065344 DOI: 10.1016/j.lungcan.2022.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/21/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinically, accurate pathological diagnosis is often challenged by insufficient tissue amounts and the unaffordability of additional immunohistochemical or genetic tests; thus, there is an urgent need for a universal approach to improve the subtyping of lung cancer without the above limitations. Here we aimed to develop a deep learning system to predict the immunohistochemistry (IHC) phenotype directly from whole-slide images (WSIs) to improve the subtyping of lung cancer from surgical resection and biopsy specimens. METHODS A total of 1914 patients with lung cancer from three independent hospitals in China were enrolled for WSI-based immunohistochemical feature prediction system (WIFPS) development and validation. RESULTS The WIFPS could directly predict the IHC status of nine subtype-specific biomarkers, including CK7, TTF-1, Napsin A, CK5/6, P63, P40, CD56, Synaptophysin, and Chromogranin A, achieving average areas under the curve (AUCs) of 0.912, 0.906, and 0.888 and overall diagnostic accuracies of 0.925, 0.941, and 0.887 in the validation datasets of total, external surgical resection specimens and biopsy specimens, respectively. The histological subtyping performance of the WIFPS remained comparable with that of general pathologists (GPs), with Cohen's kappa values ranging from 0.7646 to 0.8282. Furthermore, the WIFPS could be trained to not only predict the IHC status of anaplastic lymphoma kinase (ALK), programmed death-1 (PD-1), and programmed death ligand 1 (PD-L1), but also predict EGFR and KRAS mutation status, with AUCs from 0.525 to 0.917, as detected in separate populations. CONCLUSIONS In this study, the WIFPS showed its proficiency as a useful complement to traditional histologic subtyping for integrated immunohistochemical spectrum prediction as well as potential in the detection of gene mutations.
Collapse
Affiliation(s)
- Yanyang Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huan Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Medical Big Data Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhiqiang Cheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Department of Pathology, Shenzhen People's Hospital, Shenzhen, China
| | - Lili Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sui Peng
- Molecular Diagnosis Center or Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jianbo Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Minglei Yang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chenghao Lin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yu Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuefeng Wang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Leilei Huang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yangshan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weizhong Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Molecular Diagnosis Center or Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
257
|
Shan G, Bi G, Bian Y, Valeria B, Zeng D, Zhang H, Yao G, Zhang Y, Fan H, Zhan C. Genomic And Tumor Microenvironment Differences Between Cell Cycle Progression Pathway Altered/Non-Altered Patients With Lung Adenocarcinoma. Front Oncol 2022; 12:843528. [PMID: 35296002 PMCID: PMC8919059 DOI: 10.3389/fonc.2022.843528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
BackgroundIdentified as a hallmark of cancer, the dysregulated cell cycle progression plays an important role in the promotion and progression of lung adenocarcinoma (LUAD). However, the genomic and microenvironment differences between cell cycle progression pathway altered/non-altered LUAD patients remain to be elucidated.Materials and MethodsData of this study were obtained from The Cancer Genome Atlas (TCGA), including simple nucleotide variation, copy number variation (CNV), RNA-seq gene expression, miRNA expression, survival, and clinical information. Besides, 34 LUAD samples from our institution were used as a validation cohort. Differentially expressed genes (DEGs), enrichment analysis, and immune cell infiltration were detected. At last, we built a LASSO-binary Logistic regression model to predict the cell-cycle-related gene mutation (CDKN2A, CCND1, CDK4, CCNE1, and RB1) in LUAD patients and further verified it in the samples from our institution.ResultsBased on the cell cycle progression pathway status, the LUAD patients were divided into the mutation (n=322) and wild (n=46) groups. Compared to the wild group, the mutation group had a higher mutational load and CNV. Among the 16684 protein-coding genes analyzed, 302 were upregulated, and 354 were downregulated in the mutation group. Enrichment analysis indicated that these DEGs were closely related to metabolism items. After performing immune cell infiltration analysis of 22 immune cells, we found the proportion of 5 immune cells such as monocytes (P<0.01) and dendritic cells (P<0.01) were higher in the wild group. Finally, a cell-cycle-related 15-signature model was built by LASSO-Logistic regression analysis, which could predict the cell cycle progression pathway-related gene mutation (CDKN2A, CCND1, CDK4, CCNE1, and RB1) in LUAD patients. The validation cohorts showed the sensitivity and specificity of this model were 0.667 and 0.929, respectively.ConclusionThe genomic and microenvironment characteristics differed between the cell cycle progression pathway altered/non-altered patients with LUAD. Our findings may provide new insight into personalized treatment for LUAD patients.
Collapse
Affiliation(s)
- Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Besskaya Valeria
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dejun Zeng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- *Correspondence: Yi Zhang, ; Hong Fan,
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
- *Correspondence: Yi Zhang, ; Hong Fan,
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
258
|
Kroschke J, von Stackelberg O, Heußel CP, Wielpütz MO, Kauczor HU. Imaging Biomarkers in Thoracic Oncology: Current Advances in the Use of Radiomics in Lung Cancer Patients and its Potential Use for Therapy Response Prediction and Monitoring. ROFO-FORTSCHR RONTG 2022; 194:720-727. [PMID: 35211928 DOI: 10.1055/a-1729-1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related deaths. The development of therapies targeting molecular alterations has significantly improved the treatment of NSCLC patients. To identify these targets, tumor phenotyping is required, with tissue biopsies and molecular pathology being the gold standard. Some patients do not respond to targeted therapies and many patients suffer from tumor recurrence, which can in part be explained by tumor heterogeneity. This points out the need for new biomarkers allowing for better tumor phenotyping and monitoring during treatment to assess patient outcome. METHOD The contents of this review are based on a literature search conducted using the PubMed database in March 2021 and the authors' experience. RESULTS AND CONCLUSION The use of radiomics and artificial intelligence-based approaches allows for the identification of imaging biomarkers in NSCLC patients for tumor phenotyping. Several studies show promising results for models predicting molecular alterations, with the best results being achieved by combining structural and functional imaging. Radiomics could help solve the pressing clinical need for assessing and predicting therapy response. To reach this goal, advanced tumor phenotyping, considering tumor heterogeneity, is required. This could be achieved by integrating structural and functional imaging biomarkers with clinical data sources, such as liquid biopsy results. However, to allow for radiomics-based approaches to be introduced into clinical practice, further standardization using large, multi-center datasets is required. KEY POINTS · Some NSCLC patients do not benefit from targeted therapies, and many patients suffer from tumor recurrence, pointing out the need for new biomarkers allowing for better tumor phenotyping and monitoring during treatment.. · The use of radiomics-based approaches allows for the identification of imaging biomarkers in NSCLC patients for tumor phenotyping.. · A multi-omics approach integrating not only structural and functional imaging biomarkers but also clinical data sources, such as liquid biopsy results, could further enhance the prediction and assessment of therapy response.. CITATION FORMAT · Kroschke J, von Stackelberg O, Heußel CP et al. Imaging Biomarkers in Thoracic Oncology: Current Advances in the Use of Radiomics in Lung Cancer Patients and its Potential Use for Therapy Response Prediction and Monitoring. Fortschr Röntgenstr 2022; DOI: 10.1055/a-1729-1516.
Collapse
Affiliation(s)
- Jonas Kroschke
- Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research, Giessen, Germany.,Department for Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik am Universitätsklinikum Heidelberg, Germany
| | - Oyunbileg von Stackelberg
- Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research, Giessen, Germany.,Department for Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik am Universitätsklinikum Heidelberg, Germany
| | - Claus Peter Heußel
- Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research, Giessen, Germany.,Department for Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik am Universitätsklinikum Heidelberg, Germany
| | - Mark Oliver Wielpütz
- Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research, Giessen, Germany.,Department for Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik am Universitätsklinikum Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Department for Diagnostic and Interventional Radiology, University Hospital Heidelberg, Germany.,Translational Lung Research Center (TLRC), German Center for Lung Research, Giessen, Germany.,Department for Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik am Universitätsklinikum Heidelberg, Germany
| |
Collapse
|
259
|
Kolesar J, Peh S, Thomas L, Baburaj G, Mukherjee N, Kantamneni R, Lewis S, Pai A, Udupa KS, Kumar An N, Rangnekar VM, Rao M. Integration of liquid biopsy and pharmacogenomics for precision therapy of EGFR mutant and resistant lung cancers. Mol Cancer 2022; 21:61. [PMID: 35209919 PMCID: PMC8867675 DOI: 10.1186/s12943-022-01534-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/07/2022] [Indexed: 11/22/2022] Open
Abstract
The advent of molecular profiling has revolutionized the treatment of lung cancer by comprehensively delineating the genomic landscape of the epidermal growth factor receptor (EGFR) gene. Drug resistance caused by EGFR mutations and genetic polymorphisms of drug metabolizing enzymes and transporters impedes effective treatment of EGFR mutant and resistant lung cancer. This review appraises current literature, opportunities, and challenges associated with liquid biopsy and pharmacogenomic (PGx) testing as precision therapy tools in the management of EGFR mutant and resistant lung cancers. Liquid biopsy could play a potential role in selection of precise tyrosine kinase inhibitor (TKI) therapies during different phases of lung cancer treatment. This selection will be based on the driver EGFR mutational status, as well as monitoring the development of potential EGFR mutations arising during or after TKIs treatment, since some of these new mutations may be druggable targets for alternative TKIs. Several studies have identified the utility of liquid biopsy in the identification of EGFR driver and acquired resistance with good sensitivities for various blood-based biomarkers. With a plethora of sequencing technologies and platforms available currently, further evaluations using randomized controlled trials (RCTs) in multicentric, multiethnic and larger patient cohorts could enable optimization of liquid-based assays for the detection of EGFR mutations, and support testing of CYP450 enzymes and drug transporter polymorphisms to guide precise dosing of EGFR TKIs.
Collapse
Affiliation(s)
- Jill Kolesar
- Department of Pharmacy Practice & Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Spencer Peh
- Department of Pharmacy Practice & Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Levin Thomas
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gayathri Baburaj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nayonika Mukherjee
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raveena Kantamneni
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shirley Lewis
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Ananth Pai
- Department of Medical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Karthik S Udupa
- Department of Medical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Naveena Kumar An
- Department of Surgical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vivek M Rangnekar
- Markey Cancer Centre and Department of Radiation Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
260
|
Xu C, Ju D, Zhang X. Chimeric antigen receptor T cell therapy: challenges and opportunities in lung cancer. Antib Ther 2022; 5:73-83. [PMID: 35372786 PMCID: PMC8972219 DOI: 10.1093/abt/tbac006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 11/23/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the paradigm in hematological malignancies treatment, driving an ever-expanding number of basic research and clinical trials of genetically engineering T cells to treat solid tumors. CAR T-cell therapies based on the antibodies targeting Mesothelin, CEA, EGFR, EGFR, MUC1, DLL3, and emerging novel targets provide promising efficacy for lung cancer patients. However, clinical application of CAR T-cell therapy against lung cancer remains limited on account of physical and immune barriers, antigen escape and heterogeneity, on-target off-tumor toxicity, and many other reasons. Understanding the evolution of CAR structure and the generalizable requirements for manufacturing CAR T cells as well as the interplay between lung tumor immunology and CAR T cells will improve clinical translation of this therapeutic modality in lung cancer. In this review, we systematically summarize the latest advances in CAR T-cell therapy in lung cancer, focusing on the CAR structure, target antigens, challenges, and corresponding new strategies.
Collapse
Affiliation(s)
- Caili Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
261
|
Qiao H, Feng Y, Tang H. COL6A6 inhibits the proliferation and metastasis of non-small cell lung cancer through the JAK signalling pathway. Transl Cancer Res 2022; 10:4514-4522. [PMID: 35116307 PMCID: PMC8798920 DOI: 10.21037/tcr-21-2002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/16/2021] [Indexed: 12/19/2022]
Abstract
Background Collagen type VI alpha 6 chain (COL6A6) plays a vital role in maintaining cell structural integrity and regulating cell function. It has been proven to be a tumor suppressor gene and molecular therapeutic target. However, the mechanism of COL6A6 in non-small cell lung cancer (NSCLC) has not been elucidated. The purpose of this study was to investigate the relationship between COL6A6 and NSCLC. Methods We analyzed the expression of COL6A6 in NSCLC using public databases and verified the findings in NSCLC tissues and cells. The protein expression of COL6A6 was evaluated by Western blot. The CCK8 and Transwell assays were used to assess the invasion ability of NSCLC cells after COL6A6 knockdown. At the same time, we discussed the role of the JAK signalling pathway that may be related to COL6A6. Results Bioinformatics analysis showed that COL6A6 expression was downregulated in NSCLC, and its high expression was associated with a better prognosis of NSCLC. In vitro, the expression of COL6A6 in NSCLC tissues was significantly lower than that in adjacent tissues. Furthermore, COL6A6 knockout accelerated the proliferation, invasion, and migration of NSCLC cells and activated the JAK signalling pathway. Conclusions Our study illustrates that COL6A6 is a tumor suppressor gene in NSCLC and is involved in NSCLC tumorigenesis by regulating the JAK signalling pathway. Therefore, COL6A6 holds promise as a molecular therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Han Qiao
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Feng
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Huaping Tang
- Department of Respiratory Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
262
|
Petranovic M, Raoof S, Digumarthy SR, Sharma A, Shepard JAO, Gainor JF, Pandharipande PV. Liquid Biopsy, Diagnostic Imaging, and Future Synergies. J Am Coll Radiol 2022; 19:336-343. [DOI: 10.1016/j.jacr.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022]
|
263
|
Wang Z, Zhang L, Xu W, Li J, Liu Y, Zeng X, Zhong M, Zhu Y. The Multi-Omics Analysis of Key Genes Regulating EGFR-TKI Resistance, Immune Infiltration, SCLC Transformation in EGFR-Mutant NSCLC. J Inflamm Res 2022; 15:649-667. [PMID: 35140497 PMCID: PMC8818984 DOI: 10.2147/jir.s341001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Background Lung cancer is a high-risk malignancy worldwide. The harboring of epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer (NSCLC) makes EGFR-tyrosine kinase inhibitor (EGFR-TKI) an attractive therapeutic option. However, patients usually suffer the primary and secondary resistance to EGFR-TKI. And the molecular alteration is still not fully clear and needs further study. Methods The GEO database was utilized to find the differentially expressed genes (DEGs) in NSCLC profiles resistant to the 1st or 2nd generation EGFR-TKI. We analyzed the expression and pathway enrichment of hub genes, and the prognosis of EGFR mutant/wild-type lung adenocarcinoma (LUAD). Moreover, small cell lung cancer (SCLC) and TKI-resistant profiles were used to find common DEGs, and construct miRNA regulatory network. Analysis was performed of hub genes' related immune infiltration, drug sensitivity, and methylation. Further, we analyzed hub gene expression in EGFR-mutant LUAD and paracancerous tissue by qRT-PCR. Results A total of 107 DEGs were found related to TKI resistance. Eleven hub genes were obtained after visualization, of which 5 hub genes were co-expressed in SCLC with common miRNAs. Lower expression of SPP1 (hub gene) was associated with better survival in NSCLC. The immune infiltration analysis showed more CD4+ T cells in the resistant group with high expression of SPP1. SPP1 and CD44 promoters’ methylations were independent prognostic factors of LUAD. And the expression level of SPP1 related to the sensitivity of EGFR-TKIs in multiple cancer cell lines. qRT-PCR validated the higher expression of SPP1 in EGFR-mutant LUAD than in normal tissue. Conclusion Our study suggested that the upregulation of SPP1 might induce resistance to the 1st and 2nd generation EGFR-TKI, and influence tumor immune infiltration, resulting in poor survival. ZEB1, SPP1, MUC1, CD44, and ESRP1 might be molecular drivers of SCLC transformation of TKI resistance.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Lingling Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Wenwen Xu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Yi Liu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Xiaozhu Zeng
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Maoxi Zhong
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
| | - Yuxi Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People’s Republic of China
- Correspondence: Yuxi Zhu, Tel +86-23-88955813, Fax +862368811487, Email
| |
Collapse
|
264
|
Ishii J, Sato-Yazawa H, Kashiwagi K, Nakadate K, Iwamoto M, Kohno K, Miyata-Hiramatsu C, Masawa M, Onozaki M, Noda S, Miyazawa T, Takagi M, Yazawa T. Endocrine secretory granule production is caused by a lack of REST and intragranular secretory content and accelerated by PROX1. J Mol Histol 2022; 53:437-448. [PMID: 35094211 PMCID: PMC9117388 DOI: 10.1007/s10735-021-10055-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/23/2021] [Indexed: 12/30/2022]
Abstract
Endocrine secretory granules (ESGs) are morphological characteristics of endocrine/neuroendocrine cells and store peptide hormones/neurotransmitters. ESGs contain prohormones and ESG-related molecules, mainly chromogranin/secretogranin family proteins. However, the precise mechanism of ESG formation has not been elucidated. In this study, we experimentally induced ESGs in the non-neuroendocrine lung cancer cell line H1299. Since repressive element 1 silencing transcription factor (REST) and prospero homeobox 1 (PROX1) are closely associated with the expression of ESG-related molecules, we edited the REST gene and/or transfected PROX1 and then performed molecular biology, immunocytochemistry, and electron and immunoelectron microscopy assays to determine whether ESG-related molecules and ESGs were induced in H1299 cells. Although chromogranin/secretogranin family proteins were induced in H1299 cells by knockout of REST and the induction was accelerated by the PROX1 transgene, the ESGs could not be defined by electron microscopy. However, a small number of ESGs were detected in the H1299 cells lacking REST and expressing pro-opiomelanocortin (POMC) by electron microscopy. Furthermore, many ESGs were produced in the REST-lacking and PROX1- and POMC-expressing H1299 cells. These findings suggest that a lack of REST and the expression of genes related to ESG content are indispensable for ESG production and that PROX1 accelerates ESG production. Trial registration: Not applicable.
Collapse
Affiliation(s)
- Jun Ishii
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Hanako Sato-Yazawa
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Korehito Kashiwagi
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Kazuhiko Nakadate
- Education Research Center, Meiji Pharmaceutical University, Kiyose-shi, Tokyo, Japan
| | - Masami Iwamoto
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
- Department of Pathology, The Jikei University, Minato-ku, Tokyo, Japan
| | - Kakeru Kohno
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
- Institute of Life Innovation Studies, Toyo University, Itakura-machi, Gunma, Japan
| | - Chie Miyata-Hiramatsu
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Meitetsu Masawa
- Department of Respiratory Medicine, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Masato Onozaki
- Department of Diagnostic Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Shuhei Noda
- Department of Diagnostic Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Tadasuke Miyazawa
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Megumi Takagi
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan
| | - Takuya Yazawa
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, Mibu-machi, Tochigi, Japan.
| |
Collapse
|
265
|
Circular RNA circZCCHC6 contributes to tumorigenesis by regulating LPCAT1 via miR-433-3p in non-small cell lung cancer. Clin Exp Med 2022; 22:647-659. [PMID: 35089454 DOI: 10.1007/s10238-021-00780-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 11/30/2021] [Indexed: 11/03/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-associated mortality worldwide. Circular RNA (circRNA) circZCCHC6 has been reported to be upregulated in the plasma from NSCLC patients. This study is designed to explore the role and mechanism of circZCCHC6 in NSCLC. CircZCCHC6, microRNA-433-3p (miR-433-3p), and lysophosphatidylcholine acyltransferase 1 (LPCAT1) level were determined by real-time quantitative polymerase chain reaction. Cell viability, cell cycle progression, migration, and invasion were assessed by 3-(4,5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT), flow cytometry, wound healing, and transwell assays, severally. The binding relationship between miR-433-3p and circZCCHC6 or LPCAT1 was predicted by Circinteractome or Starbase, and then verified by a dual-luciferase reporter, RNA pull-down, or RNA Immunoprecipitation (RIP) assays. Protein levels of LPCAT1, Cyclin D1, E-cadherin, and Vimentin were examined by western blot assay. The biological role of circZCCHC6 on NSCLC tumor growth and epithelial-mesenchymal transition (EMT) was examined by the xenograft tumor model in vivo. CircZCCHC6 was highly expressed in NSCLC serum, tissues, and cells. Moreover, circZCCHC6 knockdown could repress cell viability, cell cycle progression, migration, invasion, and EMT in NSCLC cells in vitro. The mechanical analysis suggested that circZCCHC6 acted as a sponge of miR-433-3p to regulate LPCAT1 expression. CircZCCHC6 silencing hindered cell growth and EMT of NSCLC in vivo. CircZCCHC6 inhibited the progression of NSCLC cells partly by regulating the miR-433-3p/LPCAT1 axis, implying a promising therapeutic target for the NSCLC treatment.
Collapse
|
266
|
Li YC. Durable response to durvalumab-based immunochemotherapy in small-cell lung carcinoma transformation from EGFR-mutant non-small cell lung cancer: A case report. Thorac Cancer 2022; 13:775-779. [PMID: 35088537 PMCID: PMC8888151 DOI: 10.1111/1759-7714.14325] [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: 09/28/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/30/2022] Open
Abstract
Combined small‐cell lung carcinoma (C‐SCLC) is small‐cell lung carcinoma (SCLC) with added non–small‐cell morphology. We report a case of epidermal growth factor receptor (EGFR) mutation‐positive C‐SCLC in an 84‐year‐old patient with metastatic brain lesions who developed intrinsic resistance to osimertinib, a tyrosine kinase inhibitor (TKI). The patient was diagnosed with small‐cell transformation of non–small‐cell lung carcinoma (NSCLC) and received 6 cycles of dose‐adjusted durvalumab with etoposide and carboplatin. In December 2021, the patient received the seventeenth cycle of maintenance durvalumab 19 months after diagnosis and showed continued treatment response and disease control. Comprehensive molecular profiling and repeated biopsies are recommended in NSCLC patients who progress on first‐line EGFR‐TKIs. Durvalumab in combination with chemotherapy appears to be beneficial for EGFR mutation‐positive C‐SCLC patients that are resistant to TKIs.
Collapse
Affiliation(s)
- Yu-Chung Li
- Hong Kong United Oncology Centre, Hong Kong, China
| |
Collapse
|
267
|
Kim M, Ly SH, Xie Y, Duronio GN, Ford-Roshon D, Hwang JH, Sulahian R, Rennhack JP, So J, Gjoerup O, Talamas JA, Grandclaudon M, Long HW, Doench JG, Sethi NS, Giannakis M, Hahn WC. YAP1 and PRDM14 converge to promote cell survival and tumorigenesis. Dev Cell 2022; 57:212-227.e8. [PMID: 34990589 PMCID: PMC8827663 DOI: 10.1016/j.devcel.2021.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/15/2021] [Accepted: 12/03/2021] [Indexed: 01/26/2023]
Abstract
The transcriptional co-activator YAP1 oncogene is the downstream effector of the Hippo pathway, which regulates tissue homeostasis, organ size, regeneration, and tumorigenesis. Multiple cancers are dependent on sustained expression of YAP1 for cell proliferation, survival, and tumorigenesis, but the molecular basis of this oncogene dependency is not well understood. To identify genes that can functionally substitute for YAP1, we performed a genome-scale genetic rescue screen in YAP1-dependent colon cancer cells expressing an inducible YAP1-specific shRNA. We found that the transcription factor PRDM14 rescued cell proliferation and tumorigenesis upon YAP1 suppression in YAP1-dependent cells, xenografts, and colon cancer organoids. YAP1 and PRDM14 individually activated the transcription of calmodulin 2 (CALM2) and a glucose transporter SLC2A1 upon YAP1 suppression, and CALM2 or SLC2A1 expression was required for the rescue of YAP1 suppression. Together, these findings implicate PRDM14-mediated transcriptional upregulation of CALM2 and SLC2A1 as key components of oncogenic YAP1 signaling and dependency.
Collapse
Affiliation(s)
- Miju Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Seav Huong Ly
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yingtian Xie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Gina N Duronio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dane Ford-Roshon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Justin H Hwang
- Masonic Cancer Center and Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Rita Sulahian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan P Rennhack
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan So
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ole Gjoerup
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jessica A Talamas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
268
|
Osorio JC, Blanco R, Corvalán AH, Muñoz JP, Calaf GM, Aguayo F. Epstein-Barr Virus Infection in Lung Cancer: Insights and Perspectives. Pathogens 2022; 11:132. [PMID: 35215076 PMCID: PMC8878590 DOI: 10.3390/pathogens11020132] [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: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 11/30/2022] Open
Abstract
Lung cancer (LC) is the leading cause of cancer death worldwide. Tobacco smoke is the most frequent risk factor etiologically associated with LC, although exposures to other environmental factors such as arsenic, radon or asbestos are also involved. Additionally, the involvement of some viral infections such as high-risk human papillomaviruses (HR-HPVs), Merkel cell polyomavirus (MCPyV), Jaagsiekte Sheep Retrovirus (JSRV), John Cunningham Virus (JCV), and Epstein-Barr virus (EBV) has been suggested in LC, though an etiological relationship has not yet been established. EBV is a ubiquitous gamma herpesvirus causing persistent infections and some lymphoid and epithelial tumors. Since EBV is heterogeneously detected in LCs from different parts of the world, in this review we address the epidemiological and experimental evidence of a potential role of EBV. Considering this evidence, we propose mechanisms potentially involved in EBV-associated lung carcinogenesis. Additional studies are warranted to dissect the role of EBV in this very frequent malignancy.
Collapse
Affiliation(s)
- Julio C. Osorio
- Population Registry of Cali, Department of Pathology, Universidad del Valle, Cali 760042, Colombia;
| | - Rancés Blanco
- Laboratorio de Oncovirología, Programa de Virología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Alejandro H. Corvalán
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago 8320000, Chile;
| | - Juan P. Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (J.P.M.); (G.M.C.)
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile; (J.P.M.); (G.M.C.)
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | | |
Collapse
|
269
|
Hu H, Tou FF, Mao WM, Xu YL, Jin H, Kuang YK, Han CB, Guo CY. microRNA-1321 and microRNA-7515 contribute to the progression of non-small cell lung cancer by targeting CDC20. Kaohsiung J Med Sci 2022; 38:425-436. [PMID: 35050556 DOI: 10.1002/kjm2.12500] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 12/10/2021] [Accepted: 12/17/2021] [Indexed: 11/10/2022] Open
Abstract
Cell division cycle 20 (CDC20) and microRNAs (miRNAs) are differentially expressed in non-small cell lung cancer (NSCLC). The current study aimed to investigate the role of miR-1321 and miR-7515 regulation in CDC20 during NSCLC development. CDC20 expression in paracancerous and tumor tissues was assessed using quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The relationship between CDC20 expression and prognosis of patients was analyzed using the TCGA database. The expression profile of CDC20 in healthy lung cells and NSCLC cells was detected using qRT-PCR and western blotting. After the knockdown of CDC20 in NSCLC cells, the cell proliferation, apoptosis, migration, invasion, and cell cycle changes were investigated by CCK8, EdU, flow cytometry, wound healing, and Transwell assays. The miRNAs targeting CDC20 were predicted using two bioinformatics websites and validated using dual-luciferase assays. CDC20 was enhanced in NSCLC tissues and cells, thus predicting the poor prognosis in NSCLC patients. After CDC20 inhibition, the malignant phenotype of NSCLC cells was reverted. miR-1321 and miR-7515 targeted CDC20 and exhibited the same anti-tumor effects as CDC20 silencing. Functional rescue experiments showed that CDC20 overexpression averted the anti-tumor effects of miR-1321 and miR-7515 on NSCLC cells. miR-1321 and miR-7515 inhibited NSCLC development by targeting CDC20. Thus, the current study has implications in NSCLC treatment and provides novel insights into NSCLC management.
Collapse
Affiliation(s)
- Hao Hu
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China.,Department of Radiation Therapy, General Hospital of Southern Theater Command of Chinese People's Liberation Army, Guangzhou, China
| | - Fang-Fang Tou
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Wei-Min Mao
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Yan-Liang Xu
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Hui Jin
- Department of Thoracic Surgery, Ji'an Central Hospital, Ji'an, China
| | - Yu-Kang Kuang
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Chun-Bin Han
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China
| | - Chang-Ying Guo
- Department of Thoracic Surgery, Jiangxi Cancer Hospital, Nanchang, China.,Department of Thoracic Surgery, Nanchang University, Nanchang, China
| |
Collapse
|
270
|
Quan Y, Li L, Yin Z, Chen S, Yi J, Lang J, Zhang L, Yue Q, Zhao J. Bulbus Fritillariae Cirrhosae as a Respiratory Medicine: Is There a Potential Drug in the Treatment of COVID-19? Front Pharmacol 2022; 12:784335. [PMID: 35126123 PMCID: PMC8811224 DOI: 10.3389/fphar.2021.784335] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023] Open
Abstract
Bulbus fritillariae cirrhosae (BFC) is one of the most used Chinese medicines for lung disease, and exerts antitussive, expectorant, anti-inflammatory, anti-asthmatic, and antioxidant effects, which is an ideal therapeutic drug for respiratory diseases such as ARDS, COPD, asthma, lung cancer, and pulmonary tuberculosis. Through this review, it is found that the therapeutic mechanism of BFC on respiratory diseases exhibits the characteristics of multi-components, multi-targets, and multi-signaling pathways. In particular, the therapeutic potential of BFC in terms of intervention of “cytokine storm”, STAT, NF-κB, and MAPK signaling pathways, as well as the renin-angiotensin system (RAS) that ACE is involved in. In the “cytokine storm” of SARS-CoV-2 infection there is an intense inflammatory response. ACE2 regulates the RAS by degradation of Ang II produced by ACE, which is associated with SARS-CoV-2. For COVID-19, may it be a potential drug? This review summarized the research progress of BFC in the respiratory diseases, discussed the development potentiality of BFC for the treatment of COVID-19, explained the chemical diversity and biological significance of the alkaloids in BFC, and clarified the material basis, molecular targets, and signaling pathways of BFC for the respiratory diseases. We hope this review can provide insights on the drug discovery of anti-COVID-19.
Collapse
Affiliation(s)
- Yunyun Quan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
- Department of Pharmacognosy, West China School of Pharmacy Sichuan University, Chengdu, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Shilong Chen
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Jing Yi
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Jirui Lang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Lu Zhang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Qianhua Yue
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
| | - Junning Zhao
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, China
- Department of Pharmacognosy, West China School of Pharmacy Sichuan University, Chengdu, China
- *Correspondence: Junning Zhao,
| |
Collapse
|
271
|
Ma Z, Chen G, Chen Y, Guo Z, Chai H, Tang Y, Zheng L, Wei K, Pan C, Ma Z, Xia Y, Zhang A. MiR-937-3p promotes metastasis and angiogenesis and is activated by MYC in lung adenocarcinoma. Cancer Cell Int 2022; 22:31. [PMID: 35033084 PMCID: PMC8761314 DOI: 10.1186/s12935-022-02453-w] [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: 11/04/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is still one of the diseases with the highest mortality and morbidity, and lung adenocarcinoma (LUAD) accounts for more than half of all NSCLC cases in most countries. miRNA can be used as a potential biological marker and treatment for lung adenocarcinoma. However, the effect of miR-937-3p to the invasion and metastasis of LUAD cells is not clear. Methods miRNA microarray is used to analyze the expression of miRNA in lung adenocarcinoma tissue. Transwell migration, Wound-healing assay and Western blot analysis are used to analyze cell migration, invasion and epithelial-mesenchymal transition (EMT) capabilities. Tube formation is used to assess angiogenesis ability. In addition, dual luciferase reporter gene detection is used to identify the potential binding between miRNA and target mRNA. In vivo experiments were performed on male NOD/SCID nude mice by tail vein injection to establish a transplanted tumor model. The CHIP experiment is used to verify the transcription factors of miRNA. Result In our study, miR-937-3p was high-regulated in LUAD cell lines and tissues, and its expression level was related to tumor progression. We found that miR-937-3p high-expression has an effect on cell invasion and metastasis. In molecular mechanism, miR-937-3p causes SOX11 reduction by directly binding to the 3′-UTR of SOX11.In addition, MYC affects miR-937-3p transcription by binding to its promoter region. Conclusions Our research shows that miR-937-3p is mediated by MYC and can control the angiogenesis, invasion and metastasis of LUAD by regulating SOX11, thereby promoting the progress of LUAD. We speculate that miR-937-3p can be used as a therapeutic target and potential biomarker for LUAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02453-w.
Collapse
Affiliation(s)
- Zijian Ma
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ganyi Chen
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yiqian Chen
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zizhang Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chai
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu Tang
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lin Zheng
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ke Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chunfeng Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhifei Ma
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Aiping Zhang
- Department of Thoracic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China. .,Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210029, China.
| |
Collapse
|
272
|
Screening of Apoptosis Pathway-Mediated Anti-Proliferative Activity of the Phytochemical Compound Furanodienone against Human Non-Small Lung Cancer A-549 Cells. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010114. [PMID: 35054507 PMCID: PMC8779876 DOI: 10.3390/life12010114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 01/11/2023]
Abstract
Furanodienone (FDN), a major bioactive component of sesquiterpenes produced from Rhizoma Curcumae, has been repeatedly acknowledged for its intrinsic anticancer efficacy against different types of cancer. In this study, we aimed to investigate the cytotoxic potential of furanodienone against human lung cancer (NSCLC A549) cells in vitro, as well as its underlying molecular mechanisms in the induction of apoptosis. Herein, we found that FDN significantly inhibited the proliferation of A549 cells in a dose-dependent manner. In addition, treatment with FDN potentially triggered apoptosis in A549 cells via not only disrupting the nuclear morphology, but by activating capsase-9 and caspase-3 with concomitant modulation of the pro- and antiapoptotic gene expression as well. Furthermore, FDN revealed its competence in inducing cell cycle arrest at G0/G1 phase in A549 cells, which was associated with decreased expression of cyclin D1 and cyclin-dependent kinase 4 (CDK4), along with increased expression of CDK inhibitor p21Cip1. Intriguingly, FDN treatment efficiently downregulated the Wnt signaling pathway, which was correlated with increased apoptosis, as well as cell cycle arrest, in A549 cells. Collectively, FDN might represent a promising adjuvant therapy for the management of lung cancer.
Collapse
|
273
|
Hwang S, Hong TH, Park S, Jung HA, Sun JM, Ahn JS, Ahn MJ, Park K, Choi YL, Lee SH. Molecular subtypes of small cell lung cancer transformed from adenocarcinoma after EGFR tyrosine kinase inhibitor treatment. Transl Lung Cancer Res 2022; 10:4209-4220. [PMID: 35004251 PMCID: PMC8674595 DOI: 10.21037/tlcr-21-691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/09/2021] [Indexed: 01/22/2023]
Abstract
Background A certain proportion of non-small cell lung cancer (NSCLC) with activating EGFR mutations showed resistance to tyrosine kinase inhibitors (TKIs) by transforming their histology into small cell lung cancer (SCLC). In this study, we evaluated the molecular characteristics of transformed SCLCs. Methods Eighteen SCLC tissue samples transformed after EGFR TKI treatment were used for the analysis. Immunohistochemistry was conducted to evaluate the molecular subtype using antibodies representative of the major transcriptional factor-based molecular subtypes, ASCL1 (SCLC-A), NEUROD1 (SCLC-N), POU2F3 (SCLC-P), and YAP1. Subtypes were categorized based on a predefined criteria. Results Among the study population (n=18), most of the patients were initially diagnosed with adenocarcinoma (n=17), and one patient was diagnosed with adenosquamous histology. Eight patients (44.4%) were never-smokers, and nine patients were women (50.0%). Staining of pre-transformation sample was conducted in six patients, and five of them showed no discernible expression for ASCL1, NEUROD1, or POU2F3. However, the proportion of molecular subtypes after SCLC transformation was predominantly SCLC-N (n=9, 50.0%), followed by SCLC-Triple Negative (SCLC-TN; n=5, 27.8%) and SCLC-A (n=4, 22.2%). The median overall survival from TKI initiation was longer in patients who transformed to SCLC-A (P=0.009) than in those who transformed to either SCLC-N or SCLC-TN. However, the overall survival difference since SCLC transformation was not significant (P=0.370). Conclusions In our series, SCLC-N subtype was prevalent in SCLC transformed after EGFR TKI treatment. In addition, overall survival and the time to SCLC transformation from the EGFR TKI treatment were longer in patients who transformed to the SCLC-A type. Large-scale data will be required to confirm our findings.
Collapse
Affiliation(s)
- Soohyun Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tae Hee Hong
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Digital Health, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
274
|
Pathak AK, Husain N, Shukla S, Pandey RK, Kant S, Bala L. Impact of glutathione S transferases P1 (Ile105Val) variants on the risk of GSTp, phosphorylated c-Jun kinase, and P53 phenotypic expression and their implications on overall survival outcomes in non-small cell lung cancer patients treated with chemotherapy. Mutat Res 2022; 824:111775. [PMID: 35124341 DOI: 10.1016/j.mrfmmm.2022.111775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/18/2021] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
AIM This study focused on GST-M1, T1 null, and P1 Ile105Val variant genotypes associated with the risk of altered expression of GSTp, pJNK, and P53 in NSCLC patients. These markers and overall survival (OS) were correlated with a key set of clinicopathological characteristics. METHODS Genotyping of GST- M1, T1 (+/-), and P1 (Ile105Val) was performed using PCR-RFLP.The expression of GSTp, pJNK, and P53 phenotypes was assessed by immunohistochemistry. The Spearman test was used to examine the correlation between GSTp, pJNK, and P53. Kaplan-Meier test was used for OS analysis. RESULTS GSTP1 Val/Val and Ile/Val genotypes notably increased GSTp expression by 1.8 and 1.7 fold, respectively (p = 0.04,p = 0.06). GSTP1 Val/Val and Ile/Val genotypes considerably reduced P53 expression by 0.61 and 0.57 fold, respectively (p = 0.03& p = 0.05), respectively. GSTp, pJNK, and P53 were significantly co-expressed (p < 0.001). GSTp and pJNK expression showed a moderate negative correlation (ρ = -0.32, p = 0.046). In contrast, GSTp and P53 expression exhibited a strong negative correlation (ρ = -0.53, p < 0.0001). There was no correlation between P53 and pJNK expression(ρ = 0.07, p = 0.54). The patient's median OS was 8.9 months, and it was significantly related to pack-years, stage, metastasis, and GSTM1(-/-) genotypes (p > 0.05). SQCLC showed poor OS than ADC (5.7 months vs.9.1 months, p = 0.2). Stage IV and metastasis significantly reduced the OS (p = 0.001). The tumour size and lymph nodes reflected poor OS (p = 0.07&p = 0.06). Gemcitabine+Cisplatin and Gefitinib showed a slightly higher rate of survival (9.3 months and 8.1 months) than Pemtrexe+Cisplatin treatment (7.0 months,p = 0.8). Multivariate analysis revealed that pack-years and GSTp were independent predictors for OS (p = 0.03). CONCLUSION GSTp, pJNK, and P53 showed interconnected cascading. Age, pack-year, stage, and GSTp were found to be significant predictive factors for OS.Pack-years, GSTp independent OS predictor.
Collapse
Affiliation(s)
- Anumesh K Pathak
- Department of Pathology, Dr. Ram ManoharLohia Institute of Medical Sciences, Lucknow 226010, India; Department of Biochemistry, Babu Banarasi Das University, Lucknow 226018, India
| | - Nuzhat Husain
- Department of Pathology, Dr. Ram ManoharLohia Institute of Medical Sciences, Lucknow 226010, India.
| | - Saumya Shukla
- Department of Pathology, Dr. Ram ManoharLohia Institute of Medical Sciences, Lucknow 226010, India
| | - Rahul Kumar Pandey
- Department of Pathology, Dr. Ram ManoharLohia Institute of Medical Sciences, Lucknow 226010, India
| | - Surya Kant
- Department of Respiratory Medicine, King George's Medical University, Lucknow 226003, India
| | - Lakshmi Bala
- Department of Biochemistry, Babu Banarasi Das University, Lucknow 226018, India
| |
Collapse
|
275
|
Wang J, Feng X, Li Z, Chen Y, Huang W. Patient-derived organoids as a model for tumor research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:259-326. [PMID: 35595351 DOI: 10.1016/bs.pmbts.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer represents a leading cause of death, despite the rapid progress of cancer research, leading to urgent need for accurate preclinical model to further study of tumor mechanism and accelerate translational applications. Cancer cell lines cannot fully recapitulate tumors of different patients due to the lack of tumor complexity and specification, while the high technical difficulty, long time, and substantial cost of patient-derived xenograft model makes it unable to be used extensively for all types of tumors and large-scale drug screening. Patient-derived organoids can be established rapidly with a high success rate from many tumors, and precisely replicate the key histopathological, genetic, and phenotypic features, as well as therapeutic response of patient tumor. Therefore, they are extensively used in cancer basic research, biobanking, disease modeling and precision medicine. The combinations of cancer organoids with other advanced technologies, such as 3D bio-printing, organ-on-a-chip, and CRISPR-Cas9, contributes to the more complete replication of complex tumor microenvironment and tumorigenesis. In this review, we discuss the various methods of the establishment and the application of patient-derived organoids in diverse tumors as well as the limitations and future prospects of these models. Further advances of tumor organoids are expected to bridge the huge gap between bench and bedside and provide the unprecedented opportunities to advance cancer research.
Collapse
Affiliation(s)
- Jia Wang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Xiaoying Feng
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China
| | - Yongsong Chen
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Weiren Huang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China; Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
276
|
Arman T, Nelson PS. Endocrine and paracrine characteristics of neuroendocrine prostate cancer. Front Endocrinol (Lausanne) 2022; 13:1012005. [PMID: 36440195 PMCID: PMC9691667 DOI: 10.3389/fendo.2022.1012005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2022] Open
Abstract
Prostate cancer is a common malignancy affecting men worldwide. While the vast majority of newly diagnosed prostate cancers are categorized as adenocarcinomas, a spectrum of uncommon tumor types occur including those with small cell and neuroendocrine cell features. Benign neuroendocrine cells exist in the normal prostate microenvironment, and these cells may give rise to primary neuroendocrine carcinomas. However, the more common development of neuroendocrine prostate cancer is observed after therapeutics designed to repress the signaling program regulated by the androgen receptor which is active in the majority of localized and metastatic adenocarcinomas. Neuroendocrine tumors are identified through immunohistochemical staining for common markers including chromogranin A/B, synaptophysin and neuron specific enolase (NSE). These markers are also common to neuroendocrine tumors that arise in other tissues and organs such as the gastrointestinal tract, pancreas, lung and skin. Notably, neuroendocrine prostate cancer shares biochemical features with nerve cells, particularly functions involving the secretion of a variety of peptides and proteins. These secreted factors have the potential to exert local paracrine effects, and distant endocrine effects that may modulate tumor progression, invasion, and resistance to therapy. This review discusses the spectrum of factors derived from neuroendocrine prostate cancers and their potential to influence the pathophysiology of localized and metastatic prostate cancer.
Collapse
Affiliation(s)
- Tarana Arman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, United States
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, United States
- *Correspondence: Peter S. Nelson,
| |
Collapse
|
277
|
Fang G, Liu W, Shang Y, Huo R, Shi X, Wang Y, Li J. Characterization of non-small cell lung cancer transforming to small cell lung cancer and its response to EGFR-TKI: a case report. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:115. [PMID: 35282065 PMCID: PMC8848416 DOI: 10.21037/atm-21-6991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/14/2022] [Indexed: 11/06/2022]
Abstract
Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have demonstrated significant survival benefits for advanced non-small cell lung cancer (NSCLC) patients with sensitive EGFR mutations. However, patients with EGFR-TKI treatment often develop acquired resistance subsequently. Transformation from NSCLC to small cell lung cancer (SCLC) is a rare EGFR-TKI resistance mechanism for patients with sensitive EGFR mutations. Herein, we report a NSCLC patient with EGFR exon 19 deletion treated with EGFR-TKI. During treatment, the pathological type of tumor showed transformation from NSCLC to combined SCLC and then to pure SCLC after acquiring EGFR-TKI resistance. Genomic analysis revealed that the EGFR exon 19 deletion, TP53 Y220H mutation, and retinoblastomal transcriptional corepressor 1 (RB1) F755V mutation existed persistently. Immunohistochemical results showed the loss of EGFR and RB1 expression in SCLC. The patient received multi-line chemotherapy with platinum agents and experienced a briefly effective window, but died of aggressive tumor progression. We profiled the transformation from NSCLC to SCLC of this case and pointed out the importance of repeat biopsy in response to EGFR-TKI resistance. Our results showed a novel RB1 F755V mutation which may be associated with RB1 loss. This report summarized the clinical characteristics, mechanisms, and predictors of SCLC transformation, and discussed the treatment after transformation.
Collapse
Affiliation(s)
- Guotao Fang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Weiwei Liu
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Yanhong Shang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China.,Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, China
| | - Ran Huo
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | | | - Yanan Wang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Jinghua Li
- Medical Office, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
278
|
Li J, Wei B, Feng J, Wu X, Chang Y, Wang Y, Yang X, Zhang H, Han S, Zhang C, Zheng J, Groen H, van den Berg A, Ma J, Li H, Guo Y. Case report: TP53 and RB1 loss may facilitate the transformation from lung adenocarcinoma to small cell lung cancer by expressing neuroendocrine markers. Front Endocrinol (Lausanne) 2022; 13:1006480. [PMID: 36583000 PMCID: PMC9792468 DOI: 10.3389/fendo.2022.1006480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Transformation from lung adenocarcinoma (LUAD) to small cell lung cancer (SCLC) is one of the mechanisms responsible for acquired EGFR-TKIs resistance. Although it rarely happens this event determines a rapid disease deterioration and needs specific treatment. PATIENT AND METHOD We report a case of 75-year-old LUAD female with a p.L858R mutation in Epidermal Growth Factor Receptor (EGFR) who presented with SCLC transformation after responding to first line osimertinib treatment for only 6 months. To understand the underlying molecular mechanism, we retrospectively sequenced the first (LUAD) and the second (SCLC) biopsy using a 56 multi-gene panel. Immunohistochemistry (IHC) staining and Fluorescence In Situ Hybridization (FISH) was applied to confirm the genetic aberrations identified. RESULTS EGFR p.E709A and p.L858R, Tumor Protein p53 (TP53) p.A159D and Retinoblastoma 1 (RB1) c.365-1G>A were detected in both the diagnostic LUAD and transformed SCLC samples. A high copy number gain for Proto-Oncogene C-Myc (MYC) and a Phosphoinositide 3-Kinase Alpha (PIK3CA) p.E545K mutation were found in the transformed sample specifically. Strong TP53 staining and negative RB1 staining were observed in both LUAD and SCLC samples, but FISH only identified MYC amplification in SCLC tissue. CONCLUSION We consider the combined presence of MYC amplification with mutations in TP53 and RB1 as drivers of SCLC transformation. Our results highlight the need to systematically evaluate TP53 and RB1 status in LUAD patients to offer a different therapeutic strategy.
Collapse
Affiliation(s)
- Jun Li
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Bing Wei
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Junnan Feng
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Xinxin Wu
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Yuxi Chang
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Yi Wang
- Department of Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Xiuli Yang
- Department of Oncology, First Affiliated Hospital of Nanyang Medical College, Nanyang, China
| | - Haiyan Zhang
- Department of Pathology, First Affiliated Hospital of Nanyang Medical College, Nanyang, China
| | - Sile Han
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Cuiyun Zhang
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Jiawen Zheng
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Harry J. M. Groen
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jie Ma
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
| | - Hongle Li
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
- *Correspondence: Yongjun Guo, ; Hongle Li,
| | - Yongjun Guo
- Department of Molecular Pathology, Clinical Pathology Center, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Henan Key Laboratory of Molecular Pathology, Zhengzhou, China
- Henan International Joint Laboratory of Cancer Molecular Genetics, Zhengzhou, China
- *Correspondence: Yongjun Guo, ; Hongle Li,
| |
Collapse
|
279
|
Nuclear medicine therapy of lung cancer, breast cancer and colorectal cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00172-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
280
|
Ando T, Kage H, Shinozaki-Ushiku A, Tatsuno K, Tsutsumi S, Nagayama K, Nakajima J, Kohsaka S, Miyagawa K, Aburatani H, Mano H, Nagase T. Composite clonal analysis reveals transition of non-small cell lung cancer subtypes through accumulation of gene mutations: a case report. JTO Clin Res Rep 2022; 3:100277. [PMID: 35199052 PMCID: PMC8844245 DOI: 10.1016/j.jtocrr.2022.100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/22/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Takahiro Ando
- Department of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
- Corresponding author. Address for correspondence: Hidenori Kage, MD, PhD, Department of Respiratory Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | - Kenji Tatsuno
- Genome Science Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shuichi Tsutsumi
- Genome Science Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiro Nagayama
- Department of Cardiac & Thoracic Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Jun Nakajima
- Department of Cardiac & Thoracic Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Shinji Kohsaka
- Department of Cellular Signaling, The University of Tokyo, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Department of Cellular Signaling, The University of Tokyo, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
281
|
Zheng Y, Tang L, Liu Z. Multi-omics analysis of an immune-based prognostic predictor in non-small cell lung cancer. BMC Cancer 2021; 21:1322. [PMID: 34893051 PMCID: PMC8662860 DOI: 10.1186/s12885-021-09044-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/23/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Inhibitors targeting immune checkpoints, such as PD-1/PD-L1 and CTLA-4, have prolonged survival in small groups of non-small cell lung cancer (NSCLC) patients, but biomarkers predictive of the response to the immune checkpoint inhibitors (ICIs) remain rare. METHODS The nonnegative matrix factorization (NMF) was performed for TCGA-NSCLC tumor samples based on the LM22 immune signature to construct subgroups. Characterization of NMF subgroups involved the single sample gene set variation analysis (ssGSVA), and mutation/copy number alteration and methylation analyses. Construction of RNA interaction network was based on the identification of differentially expressed RNAs (DERs). The prognostic predictor was constructed by a LASSO-Cox regression model. Four GEO datasets were used for the validation analysis. RESULTS Four immune based NMF subgroups among NSCLC patients were identified. Genetic and epigenetic analyses between subgroups revealed an important role of somatic copy number alterations in determining the immune checkpoint expression on specific immune cells. Seven hub genes were recognized in the regulatory network closely related to the immune phenotype, and a three-gene prognosis predictor was constructed. CONCLUSIONS Our study established an immune-based prognosis predictor, which might have the potential to select subgroups benefiting from the ICI treatment, for NSCLC patients using publicly available databases.
Collapse
Affiliation(s)
- Yang Zheng
- Jilin University First Hospital, Changchun, Jilin, People's Republic of China
| | - Lili Tang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, People's Republic of China
| | - Ziling Liu
- Jilin University First Hospital, Changchun, Jilin, People's Republic of China.
| |
Collapse
|
282
|
Ju Q, Wu YT, Zhang Y, Yang WH, Zhao CL, Zhang J. Histology transformation-mediated pathological atypism in small-cell lung cancer within the presence of chemotherapy: A case report. World J Clin Cases 2021; 9:10652-10658. [PMID: 35004997 PMCID: PMC8686138 DOI: 10.12998/wjcc.v9.i34.10652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/27/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The treatment of small-cell lung cancer (SCLC) has progressed little in recent years because of its unique biological activities and complex genomic alterations. Chemotherapy combined with radiotherapy has been widely accepted as the first-line treatment for SCLC.
CASE SUMMARY Here, we present a 68-year-old male smoker who was diagnosed with SCLC of the right lung. After several cycles of concurrent chemoradiotherapy, the tumor progressed with broad metastasis to liver and bone. Histopathological examination showed an obvious transformation to adenocarcinoma, probably a partial recurrence mediated by the chemotherapy-based regimen. A mixed tumor as the primary lesion and transformation from SCLC or/and tumor stem cells may have accounted for the pathology conversion. We adjusted the treatment schedule in accord with the change in phenotype.
CONCLUSION Although diffuse skeletal and hepatic metastases were seen on a recent computed tomography scan, the patient is alive, with intervals of progression and shrinkage of his cancer.
Collapse
Affiliation(s)
- Qing Ju
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Xi'an 710000, Shaanxi Province, China
| | - Ying-Tong Wu
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Xi'an 710000, Shaanxi Province, China
| | - Yong Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Xi'an 710000, Shaanxi Province, China
| | - Wen-Hui Yang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Xi'an 710000, Shaanxi Province, China
| | - Cheng-Lei Zhao
- Department of Dermatology, Southwest Hospital, Chongqing 404100, China
| | - Jian Zhang
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, Xi'an 710000, Shaanxi Province, China
| |
Collapse
|
283
|
Cheng N, Cui X, Chen C, Li C, Huang J. Exploration of Lung Cancer-Related Genetic Factors via Mendelian Randomization Method Based on Genomic and Transcriptomic Summarized Data. Front Cell Dev Biol 2021; 9:800756. [PMID: 34938740 PMCID: PMC8686495 DOI: 10.3389/fcell.2021.800756] [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: 10/24/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Lung carcinoma is one of the most deadly malignant tumors in mankind. With the rising incidence of lung cancer, searching for the high effective cures become more and more imperative. There has been sufficient research evidence that living habits and situations such as smoking and air pollution are associated with an increased risk of lung cancer. Simultaneously, the influence of individual genetic susceptibility on lung carcinoma morbidity has been confirmed, and a growing body of evidence has been accumulated on the relationship between various risk factors and the risk of different pathological types of lung cancer. Additionally, the analyses from many large-scale cancer registries have shown a degree of familial aggregation of lung cancer. To explore lung cancer-related genetic factors, Genome-Wide Association Studies (GWAS) have been used to identify several lung cancer susceptibility sites and have been widely validated. However, the biological mechanism behind the impact of these site mutations on lung cancer remains unclear. Therefore, this study applied the Summary data-based Mendelian Randomization (SMR) model through the integration of two GWAS datasets and four expression Quantitative Trait Loci (eQTL) datasets to identify susceptibility genes. Using this strategy, we found ten of Single Nucleotide Polymorphisms (SNPs) sites that affect the occurrence and development of lung tumors by regulating the expression of seven genes. Further analysis of the signaling pathway about these genes not only provides important clues to explain the pathogenesis of lung cancer but also has critical significance for the diagnosis and treatment of lung cancer.
Collapse
Affiliation(s)
- Nitao Cheng
- Department of Thoracic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Xinran Cui
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chen Chen
- Department of Biological Repositories, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Changsheng Li
- Department of Thoracic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Jingyu Huang
- Department of Thoracic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
284
|
Skurikhin E, Pershina O, Zhukova M, Widera D, Ermakova N, Pan E, Pakhomova A, Morozov S, Kubatiev A, Dygai A. Potential of Stem Cells and CART as a Potential Polytherapy for Small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:778020. [PMID: 34926461 PMCID: PMC8678572 DOI: 10.3389/fcell.2021.778020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
Despite the increasing urgency of the problem of treating small cell lung cancer (SCLC), information on the causes of its development is fragmentary. There is no complete understanding of the features of antitumor immunity and the role of the microenvironment in the development of SCLC resistance. This impedes the development of new methods for the diagnosis and treatment of SCLC. Lung cancer and chronic obstructive pulmonary disease (COPD) have common pathogenetic factors. COPD is a risk factor for lung cancer including SCLC. Therefore, the search for effective approaches to prevention, diagnosis, and treatment of SCLC in patients with COPD is an urgent task. This review provides information on the etiology and pathogenesis of SCLC, analyses the effectiveness of current treatment options, and critically evaluates the potential of chimeric antigen receptor T cells therapy (CART therapy) in SCLC. Moreover, we discuss potential links between lung cancer and COPD and the role of endothelium in the development of COPD. Finally, we propose a new approach for increasing the efficacy of CART therapy in SCLC.
Collapse
Affiliation(s)
- Evgenii Skurikhin
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Olga Pershina
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Mariia Zhukova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading, United Kingdom
| | - Natalia Ermakova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Edgar Pan
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Angelina Pakhomova
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
| | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Aslan Kubatiev
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Alexander Dygai
- Laboratory of Regenerative Pharmacology, Goldberg ED Research Institute of Pharmacology and Regenerative Medicine, Tomsk National Research Medical Centre of the Russian Academy of Sciences, Tomsk, Russia
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
285
|
Malinova A, Veghini L, Real FX, Corbo V. Cell Lineage Infidelity in PDAC Progression and Therapy Resistance. Front Cell Dev Biol 2021; 9:795251. [PMID: 34926472 PMCID: PMC8675127 DOI: 10.3389/fcell.2021.795251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022] Open
Abstract
Infidelity to cell fate occurs when differentiated cells lose their original identity and either revert to a more multipotent state or transdifferentiate into a different cell type, either within the same embryonic lineage or in an entirely different one. Whilst in certain circumstances, such as in wound repair, this process is beneficial, it can be hijacked by cancer cells to drive disease initiation and progression. Cell phenotype switching has been shown to also serve as a mechanism of drug resistance in some epithelial cancers. In pancreatic ductal adenocarcinoma (PDAC), the role of lineage infidelity and phenotype switching is still unclear. Two consensus molecular subtypes of PDAC have been proposed that mainly reflect the existence of cell lineages with different degrees of fidelity to pancreatic endodermal precursors. Indeed, the classical subtype of PDAC is characterised by the expression of endodermal lineage specifying transcription factors, while the more aggressive basal-like/squamous subtype is defined by epigenetic downregulation of endodermal genes and alterations in chromatin modifiers. Here, we summarise the current knowledge of mechanisms (genetic and epigenetic) of cell fate switching in PDAC and discuss how pancreatic organoids might help increase our understanding of both cell-intrinsic and cell-extrinsic factors governing lineage infidelity during the distinct phases of PDAC evolution.
Collapse
Affiliation(s)
- Antonia Malinova
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Francisco X. Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre, Madrid, Spain
- CIBERONC, Madrid, Spain
- Department de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
286
|
Nussinov R, Tsai CJ, Jang H. Anticancer drug resistance: An update and perspective. Drug Resist Updat 2021; 59:100796. [PMID: 34953682 PMCID: PMC8810687 DOI: 10.1016/j.drup.2021.100796] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022]
Abstract
Driver mutations promote initiation and progression of cancer. Pharmacological treatment can inhibit the action of the mutant protein; however, drug resistance almost invariably emerges. Multiple studies revealed that cancer drug resistance is based upon a plethora of distinct mechanisms. Drug resistance mutations can occur in the same protein or in different proteins; as well as in the same pathway or in parallel pathways, bypassing the intercepted signaling. The dilemma that the clinical oncologist is facing is that not all the genomic alterations as well as alterations in the tumor microenvironment that facilitate cancer cell proliferation are known, and neither are the alterations that are likely to promote metastasis. For example, the common KRasG12C driver mutation emerges in different cancers. Most occur in NSCLC, but some occur, albeit to a lower extent, in colorectal cancer and pancreatic ductal carcinoma. The responses to KRasG12C inhibitors are variable and fall into three categories, (i) new point mutations in KRas, or multiple copies of KRAS G12C which lead to higher expression level of the mutant protein; (ii) mutations in genes other than KRAS; (iii) original cancer transitioning to other cancer(s). Resistance to adagrasib, an experimental antitumor agent exerting its cytotoxic effect as a covalent inhibitor of the G12C KRas, indicated that half of the cases present multiple KRas mutations as well as allele amplification. Redundant or parallel pathways included MET amplification; emerging driver mutations in NRAS, BRAF, MAP2K1, and RET; gene fusion events in ALK, RET, BRAF, RAF1, and FGFR3; and loss-of-function mutations in NF1 and PTEN tumor suppressors. In the current review we discuss the molecular mechanisms underlying drug resistance while focusing on those emerging to common targeted cancer drivers. We also address questions of why cancers with a common driver mutation are unlikely to evolve a common drug resistance mechanism, and whether one can predict the likely mechanisms that the tumor cell may develop. These vastly important and tantalizing questions in drug discovery, and broadly in precision medicine, are the focus of our present review. We end with our perspective, which calls for target combinations to be selected and prioritized with the help of the emerging massive compute power which enables artificial intelligence, and the increased gathering of data to overcome its insatiable needs.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, MD, 21702, USA
| |
Collapse
|
287
|
El-Kalyoubi S, Agili F, Adel I, Tantawy MA. Novel Uracil Derivatives Depicted Potential Anticancer Agents: In Vitro, Molecular Docking, and ADME Study. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
288
|
Takemura C, Kashima J, Hashimoto T, Ichikawa H, Honma Y, Goto Y, Watanabe SI, Yatabe Y. A mimic of lung adenocarcinoma: a case report of histological conversion of metastatic thyroid papillary carcinoma. Histopathology 2021; 80:1004-1007. [PMID: 34843113 DOI: 10.1111/his.14607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
In a patient with a history of cancer, distinguishing a metastasis of the prior tumour from a new primary tumour relies on the comparison of morphological features between the tumour of concern and the prior tumour, often with the aid of immunohistochemistry. This approach is the basis of pathological diagnosis, but recent advances in precision cancer medicine and cancer treatments change the situation. We here present a case report of lung cancer, mimicking primary lung adenocarcinoma, with minute foci of metastatic thyroid papillary carcinoma, in a 34-year-old man with a history of papillary thyroid carcinoma. Through the analysis of thyroid tumor and lung tumor at the age of 4 and 34 years in this patient, identical NCOA4-RET translation suggested histological conversion, which was induced probably due to high selective pressure by the treatment. As histological transformation of non-small cell lung cancer is well documented as a form of recurrence after EGFR-tyrosine kinase inhibitor treatment, the lineage conversion in association with effective treatments may be a diagnostic pitfall in the era of precision cancer medicine.
Collapse
Affiliation(s)
- Chihiro Takemura
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Jumpei Kashima
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Taiki Hashimoto
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Hitoshi Ichikawa
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Honma
- Division of Clinical Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Department of Respiratory Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
289
|
Fang D, Jin H, Huang X, Shi Y, Liu Z, Ben S. PPy@Fe 3O 4 Nanoparticles Inhibit Tumor Growth and Metastasis Through Chemodynamic and Photothermal Therapy in Non-small Cell Lung Cancer. Front Chem 2021; 9:789934. [PMID: 34820358 PMCID: PMC8606671 DOI: 10.3389/fchem.2021.789934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is considered to be a principal cause of cancer death across the world, and nanomedicine has provided promising alternatives for the treatment of NSCLC in recent years. Photothermal therapy (PTT) and chemodynamic therapy (CDT) have represented novel therapeutic modalities for cancer treatment with excellent performance. The purpose of this research was to evaluate the effects of PPy@Fe3O4 nanoparticles (NPs) on inhibiting growth and metastasis of NSCLC by combination of PTT and CDT. In this study, we synthesized PPy@Fe3O4 NPs through a very facile electrostatic absorption method. And we detected reactive oxygen species production, cell apoptosis, migration and protein expression in different groups of A549 cells and established xenograft models to evaluate the effects of PPy@Fe3O4 NPs for inhibiting the growth of NSCLC. The results showed that the PPy@Fe3O4 NPs had negligible cytotoxicity and could efficiently inhibit the cell growth and metastasis of NSCLC in vitro. In addition, the PPy@Fe3O4 NPs decreased tumor volume and growth in vivo and endowed their excellent MRI capability of observing the location and size of tumor. To sum up, our study displayed that the PPy@Fe3O4 NPs had significant synergistic effects of PTT and CDT, and had good biocompatibility and safety in vivo and in vitro. The PPy@Fe3O4 NPs may be an effective drug platform for the treatment of NSCLC.
Collapse
Affiliation(s)
- Danruo Fang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hansong Jin
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiulin Huang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongxin Shi
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zeyu Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Suqin Ben
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
290
|
Xue C, Li G, Lu J, Li L. Crosstalk between circRNAs and the PI3K/AKT signaling pathway in cancer progression. Signal Transduct Target Ther 2021; 6:400. [PMID: 34815385 PMCID: PMC8611092 DOI: 10.1038/s41392-021-00788-w] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 02/06/2023] Open
Abstract
Circular RNAs (circRNAs), covalently closed noncoding RNAs, are widely expressed in eukaryotes and viruses. They can function by regulating target gene expression, linear RNA transcription and protein generation. The phosphoinositide 3-kinase (PI3K)/AKT signaling pathway plays key roles in many biological and cellular processes, such as cell proliferation, growth, invasion, migration, and angiogenesis. It also plays a pivotal role in cancer progression. Emerging data suggest that the circRNA/PI3K/AKT axis modulates the expression of cancer-associated genes and thus regulates tumor progression. Aberrant regulation of the expression of circRNAs in the circRNA/PI3K/AKT axis is significantly associated with clinicopathological characteristics and plays an important role in the regulation of biological functions. In this review, we summarized the expression and biological functions of PI3K-AKT-related circRNAs in vitro and in vivo and assessed their associations with clinicopathological characteristics. We also further discussed the important role of circRNAs in the diagnosis, prognostication, and treatment of cancers.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Ganglei Li
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
291
|
Protective effects of piperlongumin in the prevention of inflammatory damage caused by pulmonary exposure to benzopyrene carcinogen. Int Immunopharmacol 2021; 101:108285. [PMID: 34802947 DOI: 10.1016/j.intimp.2021.108285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022]
Abstract
Benzopyrene is one of the main polycyclic aromatic hydrocarbons with carcinogenic capacity. Research has shown that anti-inflammatory drugs can reduce the incidence of lung cancer. In this scenario, we highlight piperlongumin (PL), an alkaloid from Piper longum with anti-inflammatory properties. Therefore, our aim was to study the effect of PL administration in a model of pulmonary carcinogenesis induced by benzopyrene in Balb/c mice. Animals were divided into 3 groups (n = 10/group): sham (10% DMSO), induced by benzopyrene (100 mg/kg, diluted in DMSO) without treatment (BaP) for 12 weeks and induced by benzopyrene and treated with PL (BaP/PL) (2 mg/kg in 10% DMSO) from the eighth week post-induction. Animals were weighed daily and pletsmography was performed in the 12th week. Genotoxicity and hemoglobin levels were analyzed in blood and quantification of leukocytes in bronchoalveolar lavage (BAL). Lungs were collected for histopathological evaluation, immunohistochemical studies of annexin A1 (AnxA1), cyclooxygenase 2 (COX-2), anti-apoptotic protein Bcl-2 and nuclear transcription factor (NF-kB) and also the measurement of interleukin cytokines (IL)-1β, IL-17 and tumor necrosis factor (TNF) -α. Treatment with PL reduced the pulmonary parameters (p < 0,001) of frequency, volume and pulmonary ventilation, decreased lymphocytes, monocytes and neutrophils in BAL (p < 0,05) as well as blood hemoglobin levels (p < 0,01). PL administration also reduced DNA damage and preserved the pulmonary architecture compared to the BaP group. Moreover, the anti-inflammatory effect of PL was evidenced by the maintenance of AnxA1 levels, reduction of COX-2 (p < 0,05), Bcl-2 (p < 0,01) and NF-kB (p < 0,001) expressions and decreased IL-1β, IL-17 (p < 0,01) and TNF-α (p < 0,05) levels. The results show the therapeutic potential of PL in the treatment of pulmonary anti-inflammatory and anti-tumor diseases with promising therapeutic implications.
Collapse
|
292
|
Challenge and countermeasures for EGFR targeted therapy in non-small cell lung cancer. Biochim Biophys Acta Rev Cancer 2021; 1877:188645. [PMID: 34793897 DOI: 10.1016/j.bbcan.2021.188645] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/18/2021] [Accepted: 11/03/2021] [Indexed: 12/25/2022]
Abstract
Lung cancer causes the highest mortality compared to other cancers in the world according to the latest WHO reports. Non-small cell lung cancer (NSCLC) contributes about 85% of total lung cancer cases. An extensive number of risk factors are attributed to the progression of lung cancer. Epidermal growth factor receptor (EGFR), one of the most frequently mutant driver genes, is closely involved in the development of lung cancer through regulation of the PI3K/AKT and MAPK pathways. As a representative of precision medicine, EGFR-tyrosine kinase inhibitors (TKIs) targeted therapy significantly relieves the development of activating mutant EGFR-driven NSCLC. However, treatment with TKIs facilitates the emergence of acquired resistance that continues to pose a significant hurdle with respect to EGFR targeted therapy. In this review, the development of current approved EGFR-TKIs as well as the related supporting clinical trials are summarized and discussed. Mechanisms of action and resistance were addressed respectively, which serve as important guides to understanding acquired resistance. We also explored the corresponding combination treatment options according to different resistance mechanisms. Future challenges include more comprehensive characterization of unclear resistance mechanisms in different populations and the development of more efficient and precision synthetic therapeutic strategies.
Collapse
|
293
|
Lei J, Yang D, Li R, Dai Z, Zhang C, Yu Z, Wu S, Pang L, Liang S, Zhang Y. Label-free surface-enhanced Raman spectroscopy for diagnosis and analysis of serum samples with different types lung cancer. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 261:120021. [PMID: 34116414 DOI: 10.1016/j.saa.2021.120021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/07/2021] [Accepted: 05/23/2021] [Indexed: 05/20/2023]
Abstract
Screening and detection of early lung cancer is important for diagnosis and prognosis. Intervention in early stage of lung cancer can significantly improve the cure and survival of patients. Surface-enhanced Raman spectroscopy (SERS) is an increasingly popular method of diagnosing cancer. We used silver nanoparticles (AgNPs) as the Raman-enhanced substrate to increase Raman signals, which contributes to the subsequent classification of lung cancer and normal serum. SERS acquired from the serum indicated the difference in biochemical components between cancerous (n = 51) lung serum and normal (n = 18) serum. Principal component analysis (PCA) and partial least-squares discriminant analysis (PLS-DA) were utilized to establish the identification model, and the various indicators of PLS-DA were all superior to those of the PLS model. Our study offers a new proposal for the universal applicability of analysis and identification with SERS of serum samples in clinical diagnosis.
Collapse
Affiliation(s)
- Jia Lei
- School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Dafu Yang
- The Second Department of Thoracic Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Rui Li
- School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China.
| | - ZhaoXia Dai
- The Second Department of Thoracic Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, People's Republic of China.
| | - Chenlei Zhang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China
| | - Zhanwu Yu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Shenyang 110042, People's Republic of China
| | - Shifa Wu
- School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China
| | - Lu Pang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Shanshan Liang
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Oncology Department, Affiliated Zhongshan Hospital of Dalian University, Dalian 116023, People's Republic of China
| | - Yi Zhang
- School of Physics, Dalian University of Technology, Dalian, 116023, People's Republic of China
| |
Collapse
|
294
|
Wu S, Zhu C, Tang D, Dou QP, Shen J, Chen X. The role of ferroptosis in lung cancer. Biomark Res 2021; 9:82. [PMID: 34742351 PMCID: PMC8572460 DOI: 10.1186/s40364-021-00338-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022] Open
Abstract
Lung cancer is one of the most common cancers in the world. Although medical treatment has made impressive progress in recent years, it is still one of the leading causes of cancer-related deaths in men and women. Ferroptosis is a type of non-apoptotic cell death modality, usually characterized by iron-dependent lipid peroxidation, rather than caspase-induced protein cleavage. Excessive or lack of ferroptosis is associated with a variety of diseases, including cancer and ischaemia-reperfusion injury. Recent preclinical evidence suggests that targeting ferroptotic pathway is a potential strategy for the treatment of lung cancer. In this review, we summarize the core mechanism and regulatory network of ferroptosis in lung cancer cells, and highlight ferroptosis induction-related tumor therapies. The reviewed information may provide new insights for targeted lung cancer therapy.
Collapse
Affiliation(s)
- Sikai Wu
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
| | - Chengchu Zhu
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Q Ping Dou
- Department of Oncology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Departments of Pharmacology & Pathology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Jianfei Shen
- Department of Thoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai, China.
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Linhai, China.
| | - Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
295
|
Xie Q, Chu H, Yi J, Yu H, Gu T, Guan Y, Liu X, Liang J, Li Y, Wang J. Identification of a prognostic immune-related signature for small cell lung cancer. Cancer Med 2021; 10:9115-9128. [PMID: 34741430 PMCID: PMC8683526 DOI: 10.1002/cam4.4402] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 01/14/2023] Open
Abstract
Purpose As a subgroup of lung cancer, small cell lung cancer (SCLC) is characterized by a short tumor doubling time, high rates of early occurred distant cancer spread, and poor outcomes. Despite its exquisite sensitivity to chemotherapy and radiotherapy, acquired drug resistance and tumor progression are typical. This study aimed to develop a robust signature based on immune‐related genes to predict the outcome of patients with SCLC. Methods The expression data of 77 SCLC patients from George's cohort were divided into training set and testing set, and 1534 immune‐related genes from ImmPort database were used to generate and validate the signature. Cox proportional hazards and the Kaplan–Meier analysis were used for developing and testing the prognostic signature. Single‐sample gene set enrichment analysis was used to determine immune cell infiltration phenotypes. Results A 10‐gene model comprising NR3C1, NR1D2, TANK, ARAF, HDGF, INHBE, LRSAM1, PLXNA1, PML, and SP1 with the highest frequency after 1000 interactions, was chosen to construct immune‐related signature. This signature showed robust predictive value for SCLC patients’ survival in both training and testing sets. This signature was weakly associated with the clinic pathological values like TNM stage. Furthermore, patients with low risk presented with activation of immune signal pathways, and specific immune cell infiltration with high levels of CD56bright NK cells but low levels of CD8+ T cells, mast cells, and helper T cells. Conclusion The present study developed immune‐related signature that may help predict the prognosis of SCLC patients, which reflects an unappreciated level of heterogeneity of immunophenotype associated with diverse prognosis for specific subsets in this highly lethal cancer type.
Collapse
Affiliation(s)
- Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Huili Chu
- Department of Oncology, No. 960 Hospital, The People's Liberation Army of China, Jinan, China
| | - Jian Yi
- YuceBio Technology Co., Ltd., Shenzhen, China
| | - Hui Yu
- YuceBio Technology Co., Ltd., Shenzhen, China
| | - Tiantian Gu
- YuceBio Technology Co., Ltd., Shenzhen, China
| | - Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Xiaolin Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Yan Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| |
Collapse
|
296
|
Xia Z, Qing B, Wang W, Gu L, Chen H, Yuan Y. Formation, contents, functions of exosomes and their potential in lung cancer diagnostics and therapeutics. Thorac Cancer 2021; 12:3088-3100. [PMID: 34734680 PMCID: PMC8636224 DOI: 10.1111/1759-7714.14217] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide due to diagnosis in the advanced stage and drug resistance in the subsequent treatments. Development of novel diagnostic and therapeutic methods is urged to improve the disease outcome. Exosomes are nano-sized vehicles which transport different types of biomolecules intercellularly, including DNA, RNA and proteins, and are implicated in cross-talk between cells and their surrounding microenvironment. Tumor-derived exosomes (TEXs) have been revealed to strongly influence the tumor microenvironment, antitumor immunoregulatory activities, tumor progression and metastasis. Potential of TEXs as biomarkers for lung cancer diagnosis, prognosis and treatment prediction is supported by numerous studies. Moreover, exosomes have been proposed to be promising drug carriers. Here, we review the mechanisms of exosomal formation and uptake, the functions of exosomes in carcinogenesis, and potential clinical utility of exosomes as biomarkers, tumor vaccine and drug delivery vehicles in the diagnosis and therapeutics of lung cancer.
Collapse
Affiliation(s)
- Zhenkun Xia
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Qing
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Linguo Gu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hongzuo Chen
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunchang Yuan
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
297
|
Makarem M, García-Pardo M, Leighl NB. Plasma-Based Genotyping in Advanced Solid Tumors: A Comprehensive Review. Cancers (Basel) 2021; 13:5299. [PMID: 34771462 PMCID: PMC8582457 DOI: 10.3390/cancers13215299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Molecular genotyping for advanced solid malignancies has transformed the clinical management of patients with metastatic disease. Treatment decisions in a growing number of tumors require knowledge of molecularly driven alterations in order to select optimal targeted therapy. Although genomic testing of tumor tissue is the gold standard for identifying targetable genomic alterations, biopsy samples are often limited or difficult to access. This has paved the way for the development of plasma-based approaches for genomic profiling. Recent advances in the detection of plasma-circulating tumor DNA (ctDNA) have enabled the integration of plasma-based molecular profiling into clinical practice as an alternative or complementary tool for genomic testing in the setting of advanced cancer, to facilitate the identification of driver mutations to guide initial treatment and diagnose resistance. Several guidelines now recommend the use of plasma where tumor tissue is limited to identify a targetable genomic alteration. Current plasma-based assays can evaluate multiple genes in comprehensive panels, and their application in advanced disease will be increasingly incorporated into standard practice. This review focuses on current and future applications of plasma ctDNA-based assays in advanced solid malignancies, while highlighting some limitations in implementing this technology into clinical practice.
Collapse
Affiliation(s)
| | | | - Natasha B. Leighl
- Princess Margaret Cancer Center, Department of Medical Oncology, Toronto, ON M5G 2C1, Canada; (M.M.); (M.G.-P.)
| |
Collapse
|
298
|
Wang Y, Jiang F, Xia R, Li M, Yao C, Li Y, Li H, Zhao Q, Shi M, Yu Y, Shao YW, Zhou G, Xia H, Miao L, Cai H. Unique Genomic Alterations of Cerebrospinal Fluid Cell-Free DNA Are Critical for Targeted Therapy of Non-Small Cell Lung Cancer With Leptomeningeal Metastasis. Front Oncol 2021; 11:701171. [PMID: 34671549 PMCID: PMC8522975 DOI: 10.3389/fonc.2021.701171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022] Open
Abstract
We reported unique molecular features of cerebrospinal fluid (CSF) of nonsmall cell lung cancer (NSCLC) patients with leptomeningeal metastasis (LM), suggesting establishing CSF as a better liquid biopsy in clinical practices. We performed next-generation panel sequencing of primary tumor tissue, plasma, and CSF from 131 NSCLC patients with LM and observed high somatic copy number variations (CNV) in CSF of NSCLC patients with LM. The status of EGFR-activating mutations was highly concordant between CSF, plasma, and primary tumors. ALK translocation was detected in 8.3% of tumor tissues but only 2.4% in CSF and 2.7% in plasma. Others such as ROS1 rearrangement, RET fusion, HER2 mutation, NTRK1 fusion, and BRAF V600E mutation were detected in 7.9% of CSF and 11.1% of tumor tissues but only 4% in plasma. Our study has shed light on the unique genomic variations of CSF and demonstrated that CSF might represent better liquid biopsy for NSCLC patients with LM.
Collapse
Affiliation(s)
- Yongsheng Wang
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Feng Jiang
- The First Affiliated Hospital/Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Ruixue Xia
- Department of Respiratory and Critical Care Medicine, Henan University Huaihe Hospital, Kaifeng, China
| | - Ming Li
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Chengyun Yao
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yan Li
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Hui Li
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Qi Zhao
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Mingke Shi
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Yanzhe Yu
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Yang W Shao
- Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Hongping Xia
- Department of Pathology, School of Basic Medical Sciences & Sir Run Run Hospital & Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
| | - Liyun Miao
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Hourong Cai
- Department of Respiratory Medicine & Radiology & Cardiothoracic Surgery, Nanjing Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
299
|
Zhang S, Li S, Cui Y, Cheng Y. [Research Advances on Transformation to Small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:729-733. [PMID: 34696545 PMCID: PMC8560981 DOI: 10.3779/j.issn.1009-3419.2021.101.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
小细胞肺癌(small cell lung cancer, SCLC)转化是非小细胞肺癌尤其是表皮生长因子受体(epidermal growth factor receptor, EGFR)突变患者的重要耐药机制之一。目前发现转化性SCLC具有与原发性SCLC相似的临床特征,对化疗短期有效,中位生存期仅1年左右。RB1缺失、体细胞拷贝数改变与SCLC转化有关,但发生SCLC转化的确切分子机制仍不完全清楚。转化性SCLC的治疗也面临巨大的挑战,针对SCLC的化疗方案是目前的主要治疗选择,联合治疗、局部治疗以及预防SCLC转化的策略也在探索。本文将转化性SCLC的临床特征、分子机制和治疗选择进行综述。
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130000, China
| | - Shuang Li
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130000, China
| | - Yanan Cui
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130000, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130000, China
| |
Collapse
|
300
|
Du M, Liang Y, Liu Z, Li X, Liang M, Zhou B, Gao Y. Identification of Key Genes Related to CD8+ T-Cell Infiltration as Prognostic Biomarkers for Lung Adenocarcinoma. Front Oncol 2021; 11:693353. [PMID: 34650911 PMCID: PMC8505972 DOI: 10.3389/fonc.2021.693353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 09/06/2021] [Indexed: 01/11/2023] Open
Abstract
Background CD8+ T cells are one of the central effector cells in the immune microenvironment. CD8+ T cells play a vital role in the development and progression of lung adenocarcinoma (LUAD). This study aimed to explore the key genes related to CD8+ T-cell infiltration in LUAD and to develop a novel prognosis model based on these genes. Methods With the use of the LUAD dataset from The Cancer Genome Atlas (TCGA), the differentially expressed genes (DEGs) were analyzed, and a co-expression network was constructed by weighted gene co-expression network analysis (WGCNA). Combined with the CIBERSORT algorithm, the gene module in WGCNA, which was the most significantly correlated with CD8+ T cells, was selected for the subsequent analyses. Key genes were then identified by co-expression network analysis, protein–protein interactions network analysis, and least absolute shrinkage and selection operator (Lasso)-penalized Cox regression analysis. A risk assessment model was built based on these key genes and then validated by the dataset from the Gene Expression Omnibus (GEO) database and multiple fluorescence in situ hybridization experiments of a tissue microarray. Results Five key genes (MZT2A, ALG3, ATIC, GPI, and GAPDH) related to prognosis and CD8+ T-cell infiltration were identified, and a risk assessment model was established based on them. We found that the risk score could well predict the prognosis of LUAD, and the risk score was negatively related to CD8+ T-cell infiltration and correlated with the advanced tumor stage. The results of the GEO database and tissue microarray were consistent with those of TCGA. Furthermore, the risk score was higher significantly in tumor tissues than in adjacent lung tissues and was correlated with the advanced tumor stage. Conclusions This study may provide a novel risk assessment model for prognosis prediction and a new perspective to explore the mechanism of tumor immune microenvironment related to CD8+ T-cell infiltration in LUAD.
Collapse
Affiliation(s)
- Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixu Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingkai Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|