1
|
Shukla S, Pandey RK, Gupta V, Husain N, Gupta A, Kant S. Programmed Death Ligand-1 Testing in Adenocarcinoma Lung: A Comparative Study of Cell Block versus Biopsy. J Cytol 2023; 40:165-168. [PMID: 38058664 PMCID: PMC10697314 DOI: 10.4103/joc.joc_33_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/24/2023] [Accepted: 08/16/2023] [Indexed: 12/08/2023] Open
Abstract
Background Immunotherapy currently stands as a novel treatment option, specifically in cases of advanced non-small cell lung carcinoma (NSCLC). Expression of programmed death ligand-1 (PD-L1) in tumor cells forms the mainstay for the use of anti-PD-L1 monoclonal antibodies in the treatment of NSCLC. Aims The objectives of the study were to assess utility of cell blocks for testing of PD-L1 in adenocarcinoma lung and to compare the expression of PD-L1 in cell blocks and the corresponding biopsy specimens. Materials and Methods The current study was a prospective case series that included 20 cases of NSCLC-adenocarcinoma lung. Cases included in the study had biopsies performed from lung masses, along with which cell blocks were prepared from fine needle aspiration cytology (FNAC) samples. Testing for PD-L1 was done using the monoclonal PD-L1 antibody, SP-263 clone on the Ventana Benchmark XT system. PD-L1 expression was assessed only in the tumor cells, and cases with >1% expression, cytoplasmic or membranous, in tumor cells were categorized as positive. Results PD-L1 expression was identified in the biopsy samples of tumor cells of 20% of cases (n = 4/20). In the corresponding cell blocks, PD-L1 expression was identified in the tumor cells of 15% of cases (n = 3/20). Sensitivity and specificity of cell blocks were 75% and 100%, respectively. Positive and negative predictive values were 100% and 94.12%, respectively. Conclusion PD-L1 testing has both predictive and prognostic implications. PD-L1 testing in cell block samples is a potential alternative, specifically in cases where biopsy tissue is minimal or unavailable.
Collapse
Affiliation(s)
- Saumya Shukla
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Rahul K. Pandey
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Vani Gupta
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Nuzhat Husain
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Anurag Gupta
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University Lucknow, Uttar Pradesh, India
| |
Collapse
|
2
|
A pharmacological exploration of targeted drug therapy in non-small cell lung cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:147. [PMID: 35834033 DOI: 10.1007/s12032-022-01744-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/30/2022] [Indexed: 10/17/2022]
Abstract
Lung cancer is the prime cause of cancer-related deaths globally, with a contribution of 85% from non-small cell lung cancer. Before a few decades back, conventional chemotherapy was the most chosen treatment option for NSCLC but with side effects. Now, the treatment approaches have shifted to a new trend, targeted therapy, and a better treatment strategy with minimal side effects compared to chemotherapy. Advances in technologies and understanding the pathways lead to the discovery of new targets and through which it is possible to improve treatment outcomes and patient compliance. Unlike chemotherapy, targeted therapy focuses on the tumor cells and does not produce toxicity to healthy cells. The last two decades were very crucial in the development of many small molecules with the capability to target-specific proteins or genes in the disease progression pathway. Although the targeted therapy approach was a gemstone with many successful drugs for the treatment of NSCLC, various resistance mechanisms and activation of bypass signaling pathways put many of these drugs in the trash. In this review, we will discuss the major targeted proteins involved in NSCLC as well as the inhibitor drugs developed to target them for now and along with the future directions.
Collapse
|
3
|
Musashi-2 (MSI2) regulates epidermal growth factor receptor (EGFR) expression and response to EGFR inhibitors in EGFR-mutated non-small cell lung cancer (NSCLC). Oncogenesis 2021; 10:29. [PMID: 33723247 PMCID: PMC7961039 DOI: 10.1038/s41389-021-00317-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/04/2021] [Accepted: 02/19/2021] [Indexed: 01/21/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) has limited treatment options. Expression of the RNA-binding protein (RBP) Musashi-2 (MSI2) is elevated in a subset of non-small cell lung cancer (NSCLC) tumors upon progression, and drives NSCLC metastasis. We evaluated the mechanism of MSI2 action in NSCLC to gain therapeutically useful insights. Reverse phase protein array (RPPA) analysis of MSI2-depleted versus control KrasLA1/+; Trp53R172HΔG/+ NSCLC cell lines identified EGFR as a MSI2-regulated protein. MSI2 control of EGFR expression and activity in an NSCLC cell line panel was studied using RT-PCR, Western blots, and RNA immunoprecipitation. Functional consequences of MSI2 depletion were explored for cell growth and response to EGFR-targeting drugs, in vitro and in vivo. Expression relationships were validated using human tissue microarrays. MSI2 depletion significantly reduced EGFR protein expression, phosphorylation, or both. Comparison of protein and mRNA expression indicated a post-transcriptional activity of MSI2 in control of steady state levels of EGFR. RNA immunoprecipitation analysis demonstrated that MSI2 directly binds to EGFR mRNA, and sequence analysis predicted MSI2 binding sites in the murine and human EGFR mRNAs. MSI2 depletion selectively impaired cell proliferation in NSCLC cell lines with activating mutations of EGFR (EGFRmut). Further, depletion of MSI2 in combination with EGFR inhibitors such as erlotinib, afatinib, and osimertinib selectively reduced the growth of EGFRmut NSCLC cells and xenografts. EGFR and MSI2 were significantly co-expressed in EGFRmut human NSCLCs. These results define MSI2 as a direct regulator of EGFR protein expression, and suggest inhibition of MSI2 could be of clinical value in EGFRmut NSCLC.
Collapse
|
4
|
Kagawa Y, Furuta H, Uemura T, Watanabe N, Shimizu J, Horio Y, Kuroda H, Inaba Y, Kodaira T, Masago K, Fujita S, Niimi A, Hida T. Efficacy of local therapy for oligoprogressive disease after programmed cell death 1 blockade in advanced non-small cell lung cancer. Cancer Sci 2020; 111:4442-4452. [PMID: 32770608 PMCID: PMC7734009 DOI: 10.1111/cas.14605] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/10/2020] [Accepted: 07/23/2020] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically changed the strategy used to treat patients with non-small-cell lung cancer (NSCLC); however, the vast majority of patients eventually develop progressive disease (PD) and acquire resistance to ICIs. Some patients experience oligoprogressive disease. Few retrospective studies have evaluated clinical efficacy in patients with oligometastatic progression who received local therapy after ICI treatment. We conducted a retrospective analysis of advanced NSCLC patients who received PD-1 inhibitor monotherapy with nivolumab or pembrolizumab to evaluate the effects of ICIs on the patterns of progression and the efficacy of local therapy for oligoprogressive disease. Of the 307 patients treated with ICIs, 148 were evaluated in our study; 42 were treated with pembrolizumab, and 106 were treated with nivolumab. Thirty-eight patients showed oligoprogression. Male sex, a lack of driver mutations, and smoking history were significantly correlated with the risk of oligoprogression. Primary lesions were most frequently detected at oligoprogression sites (15 patients), and 6 patients experienced abdominal lymph node (LN) oligoprogression. Four patients showed evidence of new abdominal LN oligometastases. There was no significant difference in overall survival (OS) between the local therapy group and the switch therapy group (reached vs. not reached, P = .456). We summarized clinical data on the response of oligoprogressive NSCLC to ICI therapy. The results may help to elucidate the causes of ICI resistance and indicate that the use of local therapy as the initial treatment in this setting is feasible treatment option.
Collapse
Affiliation(s)
- Yusuke Kagawa
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan.,Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medicine Sciences, Nagoya, Japan
| | - Hiromi Furuta
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takehiro Uemura
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan.,Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medicine Sciences, Nagoya, Japan
| | - Naohiro Watanabe
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Junichi Shimizu
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yoshitsugu Horio
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yoshitaka Inaba
- Department of Diagnostic and Interventional Radiology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takeshi Kodaira
- Department of Radiation Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Shiro Fujita
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medicine Sciences, Nagoya, Japan
| | - Toyoaki Hida
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
5
|
Révész D, Engelhardt EG, Tamminga JJ, Schramel FMNH, Onwuteaka-Philipsen BD, van de Garde EMW, Steyerberg EW, de Vet HC, Coupé VMH. Needs with Regard to Decision Support Systems for Treating Patients with Incurable Non-small Cell Lung Cancer. JOURNAL OF CANCER EDUCATION : THE OFFICIAL JOURNAL OF THE AMERICAN ASSOCIATION FOR CANCER EDUCATION 2020; 35:345-351. [PMID: 30685832 PMCID: PMC7075822 DOI: 10.1007/s13187-019-1471-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Treatment decision-making for patients with incurable non-small cell lung cancer (NSCLC) is complex due to the rapidly increasing number of treatments and discovery of new biomarkers. Decision support systems (DSS) could assist thoracic oncologists (TO) weighing of the pros and cons of treatments in order to arrive at an evidence-based and personalized treatment advice. Our aim is to inventory (1) TO's needs with regard to DSS in the treatment of incurable (stage IIIB/IV) NSCLC patients, and (2) preferences regarding the development of future tools in this field. We disseminated an online inventory questionnaire among all members of the Section of Oncology within the Society of Physicians in Chest Medicine and Tuberculosis. Telephone interviews were conducted to better contextualize the findings from the questionnaire. In total, 58 TO completed the questionnaire and expressed a need for new DSS. They reported that it is important for tools to include genetic and immune markers, to be sufficiently validated, regularly updated, and time-efficient. Also, future DSS should incorporate multiple treatment options, integrate estimates of toxicity, quality of life and cost-effectiveness of treatments, enhance communication between caregivers and patients, and use IT solutions for a clear interface and continuous updating of tools. With this inventory among Dutch TO, we summarized the need for new DSS to aid treatment decision-making for patients with incurable NSCLC. To meet the expressed needs, substantial additional efforts will be required by DSS developers, above already existing tools.
Collapse
Affiliation(s)
- Dóra Révész
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Ellen G. Engelhardt
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Johannes J. Tamminga
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Franz M. N. H. Schramel
- Department of Lung Diseases and Treatment, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Bregje D. Onwuteaka-Philipsen
- Department of Public and Occupational Health, Amsterdam Public Health Research Institute, VU University Medical Center, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Ewoudt M. W. van de Garde
- Department of Clinical Pharmacy, St. Antonius Hospital, Koekoekslaan 1, 3435 CM Nieuwegein, The Netherlands
| | - Ewout W. Steyerberg
- Center for Medical Decision Sciences, Department of Public Health, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Henrica C.W. de Vet
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| | - Veerle M. H. Coupé
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, VU University Medical Center, De Boelelaan 1089a, PO Box 7057, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
6
|
Efficacy of a small molecule inhibitor of the transcriptional cofactor PC4 in prevention and treatment of non-small cell lung cancer. PLoS One 2020; 15:e0230670. [PMID: 32231397 PMCID: PMC7108703 DOI: 10.1371/journal.pone.0230670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
The human positive coactivator 4 (PC4) was originally identified as a multi-functional cofactor capable of mediating transcription activation by diverse gene- and tissue-specific activators. Recent studies suggest that PC4 might also function as a novel cancer biomarker and therapeutic target for different types of cancers. siRNA knockdown studies indicated that down-regulation of PC4 expression could inhibit tumorigeneicity of A549 non-small cell lung cancer tumor model in nude mice. Here we show that AG-1031, a small molecule identified by high throughput screening, can inhibit the double-stranded DNA binding activity of PC4, more effectively than its single-stranded DNA binding activity. AG-1031 also specifically inhibited PC4-dependent transcriptional activation in vitro using purified transcription factors. AG-1031 inhibited proliferation of several cultured cell lines derived from non-small cell lung cancers (NSCLC) and growth of tumors that formed from A549 cell xenografts in immuno-compromised mice. Moreover, pre-injection of AG-1031 in these mice not only reduced tumor size, but also prevented tumor formation in 20% of the animals. AG-1031 treated A549 cells and tumors from AG-1031 treated animals showed a significant decrease in the levels of both PC4 and VEGFC, a key mediator of angiogenesis in cancer. On the other hand, all tested mice remained constant weight during animal trials. These results demonstrated that AG-1031 could be a potential therapy for PC4-positive NSCLC.
Collapse
|
7
|
Wang S, Jiang W, Zhang X, Lu Z, Geng Q, Wang W, Li N, Cai X. LINC-PINT alleviates lung cancer progression via sponging miR-543 and inducing PTEN. Cancer Med 2020; 9:1999-2009. [PMID: 31981466 PMCID: PMC7064031 DOI: 10.1002/cam4.2822] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Long intergenic nonprotein coding RNA p53‐induced transcript (LINC‐PINT) has been reported to participate in various cancers. Here, we investigated the effects of LINC‐PINT on lung cancer progression. Firstly, in our study, we implied that LINC‐PINT was obviously decreased in NSCLC. Thereafter, in A549 and H1299 cells, LINC‐PINT was upregulated via transfecting LV‐LINC‐PINT. As exhibited, LINC‐PINT repressed cell proliferation and cell colony formation of A549 and H1299 cells. Subsequently, flow cytometry evidenced that A549 and H1299 cell apoptosis was obviously triggered and the cell cycle was arrested in G1 phase. Then, migration and transwell invasion experiments were carried out to detect the cell migration and invasion capacity. We found A549 and H1299 cell migration and invasion capacity were restrained by the upregulation of LINC‐PINT. Meanwhile, we predicted that miR‐543 could function as the target of LINC‐PINT and the association was verified. Moreover, we exhibited that miR‐543 was remarkably increased in lung cancer, which could be regulated by LINC‐PINT negatively. Furthermore, PTEN could act as the downstream target of miR‐543 and upregulation of miR‐543 repressed PTEN, which was reversed by LV‐PINT in A549 and H1299 cells. Finally, xenografts were utilized to confirm the function of LINC‐PINT on lung cancer. All these findings concluded that LINC‐PINT exerted crucial biological roles in NSCLC through sponging miR‐543 and inducing PTEN.
Collapse
Affiliation(s)
- Shu Wang
- Department of Gerontology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenyang Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinghua Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zilong Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Nan Li
- Department of Cardiology, Shangrao People's Hospital, Shangrao, China
| | - Xinyong Cai
- Department of Cardiology, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Domagala-Kulawik J. New Frontiers for Molecular Pathology. Front Med (Lausanne) 2019; 6:284. [PMID: 31867335 PMCID: PMC6904313 DOI: 10.3389/fmed.2019.00284] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Lung cancer remains a serious oncological problem worldwide. The delayed diagnosis and a prevalence of advanced stages in up to 70% of cases at recognition are still observed. Thanks to targeted therapies and immunotherapy a significant progress in achieving prolonged survival in some lung cancer patients is reported. A precise histopathological diagnosis, especially the recognition of adenocarcinoma, and a progress in the methods of clinical staging underlie the proper qualification of patients for a tailored therapy. The deep molecular characteristics of lung cancer in liquid biopsy, for example blood, bronchoalveolar lavage fluid (BALF), cell suspension from needle aspirates, are currently available. The molecular characteristic has recently been extended with molecular aberrations of BRAF, KRAS, MET, ERBB2, RET, NTRK next to the well-known EGFR mutations and ALK, ROS-1 relocation. The present paper discusses the usefulness of adequate pathological methods and molecular testing for the identification of a broad spectrum of predictive biomarkers for a molecular-directed lung cancer therapy. Immunotherapy with immune checkpoint inhibitors (ICIs) is approved in the first line therapy of advanced non-small-cell lung cancer. To date only PD-L1 expression on tumor cells has been found to be a marker of response to ICIs. The efficacy of ICIs as well as the susceptibility to immune-related adverse events are highly individual, so immune biomarkers are widely investigated. The candidates for predictive factors for ICIs immunotherapy include cancer cell antigenicity, presence of regulatory/suppressory molecules on cancer cells, cancer stem cells or on exosomes, and, on the other hand, an immune status of the patient. Cancers with high immune infiltration in the tumor milieu, referred to as “hot” tumors, seem to ensure a better response to ICIs than the “cold” ones. BALF analysis may replace cancer tissue examination, which is of limited access in advanced stages, for the recognition of the nature of immune response in the tumor environment. Tumor mutational burden (TMB) was shown to correlate with a good response to ICIs, especially when combined with other anticancer therapies. The present paper demonstrates the results of recent studies on lung cancer characteristics which bring us closer to the definition of useful prognostic/predictive factors.
Collapse
Affiliation(s)
- Joanna Domagala-Kulawik
- Department of Internal Medicine, Pulmonary Diseases and Allergy Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
9
|
Ząbczyk M, Królczyk G, Czyżewicz G, Plens K, Prior S, Butenas S, Undas A. Altered fibrin clot properties in advanced lung cancer: strong impact of cigarette smoking. Med Oncol 2019; 36:37. [PMID: 30891644 DOI: 10.1007/s12032-019-1262-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Dense fibrin networks resistant to lysis have been reported in patients at high risk of thromboembolism. Little is known about fibrin clot properties in cancer. We investigated fibrin clot properties and their determinants in patients with inoperable lung cancer. METHODS We enrolled 150 patients with advanced lung cancer prior to therapy and 90 control subjects matched by age, sex, cardiovascular disease, and diabetes. Plasma clot permeability (Ks), turbidimetric analysis of clot formation, clot lysis time (CLT), microparticle-associated tissue factor (MP-TF) activity, thrombin generation, and serum cotinine levels were determined. RESULTS Lung cancer patients, compared with controls, formed at a faster rate (- 8.1% lag phase) denser plasma fibrin networks (- 27.2% Ks) that displayed impaired lysis (+ 26.5% CLT), along with 19.5% higher MP-TF activity and 100% higher peak thrombin generated, also after adjustment for potential confounders. Cotinine levels were associated with fibrin maximum absorbance (r = 0.20, p = 0.016) and Ks (r = - 0.50, p < 0.0001) in cancer patients. On multivariate regression analysis, an increase in cotinine levels was a predictor of low Ks (the lower quartile, < 5.8 × 10-9 cm2; odds ratio = 1.21 per 10 ng/ml, 95% confidence interval 1.02-1.46), but not CLT. CONCLUSION Advanced lung cancer is associated with the prothrombotic plasma clot phenotype largely driven by smoking.
Collapse
Affiliation(s)
- Michał Ząbczyk
- Institute of Cardiology, Jagiellonian University Medical College, 80 Prądnicka Str. 31-202, Kraków, Poland.,Center for Research and Medical Technology, John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland
| | - Grzegorz Królczyk
- Institute of Cardiology, Jagiellonian University Medical College, 80 Prądnicka Str. 31-202, Kraków, Poland.,Oncology Ward, John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland
| | - Grzegorz Czyżewicz
- Oncology Ward, John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland
| | | | - Shannon Prior
- Department of Biochemistry, University of Vermont, 360 South Park Drive, Colchester, VT, USA
| | - Saulius Butenas
- Department of Biochemistry, University of Vermont, 360 South Park Drive, Colchester, VT, USA
| | - Anetta Undas
- Institute of Cardiology, Jagiellonian University Medical College, 80 Prądnicka Str. 31-202, Kraków, Poland. .,Center for Research and Medical Technology, John Paul II Hospital, Pradnicka 80, 31-202, Kraków, Poland. .,Faculty of Medicine and Health Sciences, Jan Kochanowski University, IX Wiekow Kielc 19A, 25-317, Kielce, Poland.
| |
Collapse
|
10
|
Choi JH, Choi J, Chung SM, Oh JY, Lee YS, Min KH, Hur GY, Shim JJ, Kang KH, Lee HK, Lee SY. The Clinical Efficacy and Safety of Four-Weekly Docetaxel as First-Line Therapy in Elderly Lung Cancer Patients with Squamous Cell Carcinoma. Tuberc Respir Dis (Seoul) 2018; 82:211-216. [PMID: 30841020 PMCID: PMC6609518 DOI: 10.4046/trd.2018.0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/15/2018] [Accepted: 10/12/2018] [Indexed: 11/24/2022] Open
Abstract
Background Docetaxel is one of the standard treatments for advanced non-small cell lung cancer. Docetaxel is usually administered in a 3-week schedule, but there is significant toxicity. In this phase II clinical study, we investigated the efficacy and safety of a 4-weekly schedule of docetaxel monotherapy, as first-line chemotherapy for advanced squamous cell carcinoma in elderly lung cancer patients. Methods Patients with stage IIIB/ IV lung squamous-cell carcinoma age 70 or older, that had not undergone cytotoxic chemotherapy were enrolled. Patients received docetaxel 25 mg/m2 on days 1, 8, and 15, every 4 weeks. Primary endpoint was the objective response rate (ORR). Secondary endpoints were progression-free survival (PFS), overall survival (OS), and toxicity profiles. Results A total of 19 patients were enrolled. Among 19 patients, 17 were for evaluated efficacy and safety. In the intent-to-treat population, ORR and disease control rate (DCR) were 11.8% and 47.1%, respectively. In the response evaluable population, ORR was 16.7% and DCR was 66.7%. Median PFS and OS were 3.1 months and 3.3 months, respectively. There were three adverse grade 3/4 events. Grade 1 neutropenia was reported in one patient. Conclusion Our data failed to demonstrate efficacy of a 4-weekly docetaxel regimen, in elderly patients with a poor performance status. However, incidence of side effects, including neutropenia, was lower than with a 3-week docetaxel regimen, as previously reported.
Collapse
Affiliation(s)
- Jong Hyun Choi
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Juwhan Choi
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Sang Mi Chung
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Jee Youn Oh
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Young Seok Lee
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Kyung Hoon Min
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Gyu Young Hur
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Jae Jeong Shim
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Kyung Ho Kang
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Hyun Kyung Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Korea
| | - Sung Yong Lee
- Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea.
| |
Collapse
|
11
|
Lee SH. Chemotherapy for Lung Cancer in the Era of Personalized Medicine. Tuberc Respir Dis (Seoul) 2018; 82:179-189. [PMID: 30841023 PMCID: PMC6609523 DOI: 10.4046/trd.2018.0068] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/05/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
Although recent advances in molecular targeted therapy and immuno-oncology have revolutionized the landscape of lung cancer therapeutics, cytotoxic chemotherapy remains an essential component of lung cancer treatment. Extensive evidence has demonstrated the clinical benefit of chemotherapy, either alone or in combination with other treatment modalities, on survival and quality of life of patients with early and advanced lung cancer. Combinational approaches with other classes of anti-neoplastic agents and new drug-delivery systems have revealed promising data and are areas of active investigation. Chemotherapy is recommended as a standard of care in patients that have progressed after tyrosine kinase inhibitors or immune checkpoint inhibitors. Chemotherapy remains the fundamental means of lung cancer management and keeps expanding its clinical implication. This review will discuss the current position and future role of chemotherapy, and specific consideration for its clinical application in the era of precision medicine.
Collapse
Affiliation(s)
- Seung Hyeun Lee
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Korea.
| |
Collapse
|
12
|
Królczyk G, Ząbczyk M, Czyżewicz G, Plens K, Prior S, Butenas S, Undas A. Altered fibrin clot properties in advanced lung cancer: impact of chemotherapy. J Thorac Dis 2018; 10:6863-6872. [PMID: 30746232 DOI: 10.21037/jtd.2018.11.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Faster formation of dense and poorly lyzable fibrin networks have been reported in patients at risk of thromboembolism, including cancer patients. We sought to investigate whether chemotherapy affects plasma fibrin clot properties and their determinants in lung cancer patients. Methods In this observational study we enrolled 83 consecutive patients with advanced inoperable lung cancer. Plasma fibrin clot permeability (Ks), turbidimetric analysis of clot formation, clot lysis time (CLT), microparticle-associated tissue factor (MP-TF) activity, and thrombin generation parameters were investigated at enrolment and 3-4 months after standard chemotherapy. Results Lung cancer patients after 4 (range, 4-5) cycles of chemotherapy had 35.6% higher D-dimer, 22.1% lower MP-TF activity, and unaltered fibrinogen compared with baseline. Chemotherapy resulted also in 7.5% increased Ks, 8.6% prolonged lag phase, and 5.4% shortened CLT, while thrombin generation was unchanged. Chemotherapy-related differences in clot structure were confirmed by scanning electron microscopy images. Fibrin clot properties after chemotherapy did not differ among histological types of lung cancer, cancer stages or chemotherapy regimens. Interestingly, never smoking (n=13, 16%) was associated with looser post-treatment fibrin structure as reflected by 12.3% higher Ks. Multiple linear regression showed that more advanced cancer stage, higher peak thrombin generation, and higher white blood cell count determined post-treatment change in Ks, while active smoking was associated with change in CLT. Conclusions Three-month chemotherapy in lung cancer patients improves clot properties despite unaffected thrombin generation, suggesting that anticancer treatment might quickly produce antithrombotic actions.
Collapse
Affiliation(s)
- Grzegorz Królczyk
- Oncology Ward, John Paul II Hospital, Cracow, Poland.,Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, Cracow, Poland
| | - Michał Ząbczyk
- Institute of Cardiology, Jagiellonian University Medical College and John Paul II Hospital, Cracow, Poland
| | | | | | - Shannon Prior
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Saulius Butenas
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Anetta Undas
- Institute of Cardiology, Jagiellonian University Medical College and John Paul II Hospital, Cracow, Poland.,Faculty of Medicine and Health Sciences, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
13
|
Stratifin regulates stabilization of receptor tyrosine kinases via interaction with ubiquitin-specific protease 8 in lung adenocarcinoma. Oncogene 2018; 37:5387-5402. [PMID: 29880877 DOI: 10.1038/s41388-018-0342-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 04/09/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022]
Abstract
Previously we have reported that stratifin (SFN, 14-3-3 sigma) acts as a novel oncogene, accelerating the tumor initiation and progression of lung adenocarcinoma. Here, pull-down assay and LC-MS/MS analysis revealed that ubiquitin-specific protease 8 (USP8) specifically bound to SFN in lung adenocarcinoma cells. Both USP8 and SFN showed higher expression in human lung adenocarcinoma than in normal lung tissue, and USP8 expression was significantly correlated with SFN expression. Expression of SFN, but not of USP8, was associated with histological subtype, pathological stage, and poor prognosis. USP8 stabilizes receptor tyrosine kinases (RTKs) such as EGFR and MET by deubiquitination, contributing to the proliferative activity of many human cancers including non-small cell lung cancer. In vitro, USP8 binds to SFN and they co-localize at the early endosomes in lung adenocarcinoma cells. Moreover, USP8 or SFN knockdown leads to downregulation of tumor cellular proliferation and upregulation of apoptosis, p-EGFR or p-MET, which are related to the degradation pathway, and accumulation of ubiquitinated RTKs, leading to lysosomal degradation. Additionally, mutant USP8, which is unable to bind to SFN, reduces the expression of RTKs and p-STAT3. We also found that interaction with SFN is critical for USP8 to exert its autodeubiquitination function and avoid dephosphorylation by PP1. Our findings demonstrate that SFN enhances RTK stabilization through abnormal USP8 regulation in lung adenocarcinoma, suggesting that SFN could be a more suitable therapeutic target for lung adenocarcinoma than USP8.
Collapse
|
14
|
Paik SS, Hwang IK, Park MJ, Lee SH. Pemetrexed Continuation Maintenance versus Conventional Platinum-Based Doublet Chemotherapy in EGFR-Negative Lung Adenocarcinoma: Retrospective Analysis. Tuberc Respir Dis (Seoul) 2018. [PMID: 29527843 PMCID: PMC5874144 DOI: 10.4046/trd.2017.0090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Although targeted therapy and immuno-oncology have shifted the treatment paradigm for lung cancer, platinum-based combination is still the standard of care for advanced non-small cell lung cancer (NSCLC). Pemetrexed continuation maintenance therapy has been approved and increasingly used for patients with nonsquamous NSCLC. However, the efficacy of this strategy has not been proven in patients without driving mutations. The objective of this study was to compare the clinical benefit of pemetrexed continuation maintenance to conventional platinum-based doublet in epidermal growth factor receptor (EGFR)-negative lung adenocarcinoma. Methods A total of 114 patients with EGFR-negative lung adenocarcinoma who were treated with platinum doublet were retrospectively enrolled. We compared the survival rates between patients received pemetrexed maintenance after four-cycled pemetrexed/cisplatin and those received at least four-cycled platinum doublet without maintenance chemotherapy as a first-line treatment. Results Forty-one patients received pemetrexed maintenance and 73 received conventional platinum doublet. Median progression-free survival (PFS), which was defined as the time from the day of response evaluation after four cycles of chemotherapy to disease progression or death, was significantly higher in the pemetrexed maintenance group compared to conventional group (5.8 months vs. 2.2 months, p<0.001). Median overall survival showed an increasing trend in the pemetrexed maintenance group (22.3 months vs. 16.1 months, p=0.098). Multivariate analyses showed that pemetrexed maintenance chemotherapy was associated with better PFS (hazard ratio, 0.73; 95% confidence interval, 0.15–0.87). Conclusion Compared to conventional platinum-based chemotherapy, premetrexed continuation maintenance treatment is associated with better clinical outcome for the patients with EGFR wild-type lung adenocarcinoma.
Collapse
Affiliation(s)
- Seung Sook Paik
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongnam Institute of Radiological & Medical Sciences, Busan, Korea
| | - In Kyoung Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Dongnam Institute of Radiological & Medical Sciences, Busan, Korea
| | - Myung Jae Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea
| | - Seung Hyeun Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University School of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Kuo CHS, Liu CY, Pavlidis S, Lo YL, Wang YW, Chen CH, Ko HW, Chung FT, Lin TY, Wang TY, Lee KY, Guo YK, Wang TH, Yang CT. Unique Immune Gene Expression Patterns in Bronchoalveolar Lavage and Tumor Adjacent Non-Neoplastic Lung Tissue in Non-Small Cell Lung Cancer. Front Immunol 2018; 9:232. [PMID: 29483918 PMCID: PMC5816075 DOI: 10.3389/fimmu.2018.00232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Background The immune cells in the local environments surrounding non-small cell lung cancer (NSCLC) implicate the balance of pro- and antitumor immunity; however, their transcriptomic profiles remain poorly understood. Methods A transcriptomic microarray study of bronchoalveolar lavage (BAL) cells harvested from tumor-bearing lung segments was performed in a discovery group. The findings were validated (1) in published microarray datasets, (2) in an independent group by RT-qPCR, and (3) in non-diseased and tumor adjacent non-neoplastic lung tissue by immunohistochemistry and in BAL cell lysates by immunoblotting. Result The differential expression of 129 genes was identified in the discovery group. These genes revealed functional enrichment in Fc gamma receptor-dependent phagocytosis and circulating immunoglobulin complex among others. Microarray datasets analysis (n = 607) showed that gene expression of BAL cells of tumor-bearing lung segment was also the unique transcriptomic profile of tumor adjacent non-neoplastic lung of early stage NSCLC and a significantly gradient increase of immunoglobulin genes’ expression for non-diseased lungs, tumor adjacent non-neoplastic lungs, and tumors was identified (ANOVA, p < 2 × 10−16). A 53-gene signature was determined with significant correlation with inhibitory checkpoint PDCD1 (r = 0.59, p = 0.0078) among others, where the nine top genes including IGJ and IGKC were RT-qPCR validated with high diagnostic performance (AUC: 0.920, 95% CI: 0.831–0.985, p = 2.98 × 10−7). Increased staining and expression of IGKC revealed by immunohistochemistry and immunoblotting in tumor adjacent non-neoplastic lung tissues (Wilcoxon signed-rank test, p < 0.001) and in BAL cell lysates (p < 0.01) of NSCLC, respectively, were noted. Conclusion The BAL cells of tumor-bearing lung segments and tumor adjacent non-neoplastic lung tissues present a unique gene expression characterized by IGKC in relation to inhibitory checkpoints. Further study of humoral immune responses to NSCLC is warranted.
Collapse
Affiliation(s)
- Chih-Hsi Scott Kuo
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Chien-Ying Liu
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Stelios Pavlidis
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Yu-Lun Lo
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yen-Wen Wang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Hung Chen
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - How-Wen Ko
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Fu-Tsai Chung
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tin-Yu Lin
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tsai-Yu Wang
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Thoracic Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Ke Guo
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Tzu-Hao Wang
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Cheng-Ta Yang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Liu SV, Macke LA, Colton BS, Imran SS, Christiansen J, Chow-Maneval E, Hornby Z, Multani PS. Response to Entrectinib in Differentiated Thyroid Cancer With a ROS1 Fusion. JCO Precis Oncol 2017; 1:PO.17.00105. [PMID: 32913977 PMCID: PMC7446518 DOI: 10.1200/po.17.00105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Stephen V. Liu
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Laura A. Macke
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Bradley S. Colton
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Sikandar S. Imran
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Jason Christiansen
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Edna Chow-Maneval
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Zachary Hornby
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| | - Pratik S. Multani
- Stephen V. Liu, Laura A. Macke, Bradley S. Colton, and Sikander S. Imran, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and Jason Christiansen, Edna Chow-Maneval, Zachary Hornby, and Pratik S. Multani, Ignyta, San Diego, CA
| |
Collapse
|
17
|
Dai Q, Li N, Zhou X. Increased miR-21a provides metabolic advantages through suppression of FBP1 expression in non-small cell lung cancer cells. Am J Cancer Res 2017; 7:2121-2130. [PMID: 29218237 PMCID: PMC5714742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 09/07/2017] [Indexed: 06/07/2023] Open
Abstract
Lung cancer is the most common solid tumor and the leading cause of cancer-related mortality worldwide. miR-21 is one of the most commonly observed aberrant miRNAs in human cancers. However, the biological roles of miR-21 in glucose metabolism of non-small cell lung cancer (NSCLC) cells remain unknown. In the present study, our findings demonstrated that miR-21 promoted glucose uptake and increased TXNIP expression. miR-21 increased lactate generation and decreased oxygen consumption in NSCLC cells. Moreover, we found that miR-21 promoted glycolysis and decreased OXPHOS. Mechanistically, fructose-1,6-biphosphatase (FBP1) was a direct target of miR-21 and observed a negative correlation between miR-21 and FBP1 in NSCLC samples. Restoring FBP1 expression reversed the effects induced by miR-21 overexpression in NSCLC cells. Together, our findings suggest the critical role of miR-21 in glucose metabolism through suppression of FBP1 in NSCLC cells. miR-21 may be a potential target of NSCLC treatment.
Collapse
Affiliation(s)
- Qingchun Dai
- Department of Critical Care Medicine, Cangzhou Central HospitalCangzhou, Hebei Province, China
| | - Na Li
- Outpatient Department Hebei Cangzhou People’s HospitalCangzhou, Hebei Province, China
| | - Xiaohong Zhou
- Pathophysiological Research Group, Hebei Medical UniversityHebei Province, China
| |
Collapse
|
18
|
Inamura K. Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Front Oncol 2017; 7:193. [PMID: 28894699 PMCID: PMC5581350 DOI: 10.3389/fonc.2017.00193] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 08/11/2017] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide due to late diagnoses and limited treatment interventions. Recently, comprehensive molecular profiles of lung cancer have been identified. These novel characteristics have enhanced the understanding of the molecular pathology of lung cancer. The identification of driver genetic alterations and potential molecular targets has resulted in molecular-targeted therapies for an increasing number of lung cancer patients. Thus, the histopathological classification of lung cancer was modified in accordance with the increased understanding of molecular profiles. This review focuses on recent developments in the molecular profiling of lung cancer and provides perspectives on updated diagnostic concepts in the new 2015 WHO classification. The WHO classification will require additional revisions to allow for reliable, clinically meaningful tumor diagnoses as we gain a better understanding of the molecular characteristics of lung cancer.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
19
|
The between Now and Then of Lung Cancer Chemotherapy and Immunotherapy. Int J Mol Sci 2017; 18:ijms18071374. [PMID: 28653990 PMCID: PMC5535867 DOI: 10.3390/ijms18071374] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/23/2017] [Accepted: 06/25/2017] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the most common cancer worldwide. Disappointingly, despite great effort in encouraging screening or, at least, a close surveillance of high-risk individuals, most of lung cancers are diagnosed when already surgically unresectable because of local advancement or metastasis. In these cases, the treatment of choice is chemotherapy, alone or in combination with radiotherapy. Here, we will briefly review the most successful and recent advances in the identification of novel lung cancer genetic lesions and in the development of new drugs specifically targeting them. However, lung cancer is still the leading cause of cancer-related mortality also because, despite impressive initial responses, the patients often develop resistance to novel target therapies after a few months of treatment. Thus, it is literally vital to continue the search for new therapeutic options. So, here, on the basis of our recent findings on the role of the tumor suppressor CCDC6 protein in lung tumorigenesis, we will also discuss novel therapeutic approaches we envision for lung cancer.
Collapse
|
20
|
Hmmier A, O'Brien ME, Lynch V, Clynes M, Morgan R, Dowling P. Proteomic analysis of bronchoalveolar lavage fluid (BALF) from lung cancer patients using label-free mass spectrometry. BBA CLINICAL 2017; 7:97-104. [PMID: 28331811 PMCID: PMC5357681 DOI: 10.1016/j.bbacli.2017.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/08/2017] [Accepted: 03/01/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related mortality in both men and women throughout the world. The need to detect lung cancer at an early, potentially curable stage, is essential and may reduce mortality by 20%. The aim of this study was to identify distinct proteomic profiles in bronchoalveolar fluid (BALF) and plasma that are able to discriminate individuals with benign disease from those with non-small cell lung cancer (NSCLC). METHODS Using label-free mass spectrometry analysis of BALF during discovery-phase analysis, a significant number of proteins were found to have different abundance levels when comparing control to adenocarcinoma (AD) or squamous cell lung carcinoma (SqCC). Validation of candidate biomarkers identified in BALF was performed in a larger cohort of plasma samples by detection with enzyme-linked immunoassay. RESULTS Four proteins (Cystatin-C, TIMP-1, Lipocalin-2 and HSP70/HSPA1A) were selected as a representative group from discovery phase mass spectrometry BALF analysis. Plasma levels of TIMP-1, Lipocalin-2 and Cystatin-C were found to be significantly elevated in AD and SqCC compared to control. CONCLUSION The results presented in this study indicate that BALF is an important proximal biofluid for the discovery and identification of candidate lung cancer biomarkers. GENERAL SIGNIFICANCE There is good correlation between the trend of protein abundance levels in BALF and that of plasma which validates this approach to develop a blood biomarker to aid lung cancer diagnosis, particularly in the era of lung cancer screening. The protein signatures identified also provide insight into the molecular mechanisms associated with lung malignancy.
Collapse
Affiliation(s)
- Abduladim Hmmier
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland; BioNano Integration Research Group, Biotechnology Research Centre, Tripoli, Libya
| | | | - Vincent Lynch
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Martin Clynes
- National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Ross Morgan
- Department of Respiratory Medicine, Beaumont Hospital, Dublin 9, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| |
Collapse
|
21
|
Jia X, Wang Z, Qiu L, Yang Y, Wang Y, Chen Z, Liu Z, Yu L. Upregulation of LncRNA-HIT promotes migration and invasion of non-small cell lung cancer cells by association with ZEB1. Cancer Med 2016; 5:3555-3563. [PMID: 27790864 PMCID: PMC5224854 DOI: 10.1002/cam4.948] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the most common solid tumor and the leading cause of cancer‐related mortality worldwide. Non‐small cell lung cancer (NSCLC) accounts for approximately 80% of all lung cancer cases. The main reason of lung cancer‐related deaths is due to tumor metastasis. But, the mechanisms of NSCLC metastasis remains poorly understood. LncRNAs play pivotal roles in multiple biological processes. LncRNA‐HIT (HOXA transcript induced by TGFβ) was recently identified. LncRNA‐HIT promotes cell migration, invasion, tumor growth, and metastasis. However, the detailed role of lncRNA‐HIT in NSCLC remains unknown. In this study, for the first time, we revealed a novel role of lncRNA‐HIT in the migration and invasion of NSCLC cells. The expression of lncRNA‐HIT was significantly upregulated in NSCLC tissues and cell lines, and the expression level of lncRNA‐HIT correlates with advanced disease stage and predicts unfavorable prognosis of NSCLC patients. Functional assays demonstrated that lncRNA‐HIT markedly increased the ability of NSCLC cells to migrate and invade. Furthermore, the molecular mechanism by which lncRNA‐HIT affects NSCLC cells was associated with regulation of ZEB1 stability. LncRNA‐HIT functions as a prometastasis oncogene by directly associating with ZEB1 to regulate NSCLC. The interaction of lncRNA‐HIT and ZEB1 may be a potential target for NSCLC therapy.
Collapse
Affiliation(s)
- Xiaojing Jia
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhicheng Wang
- Key Laboratory of Radiobiology, Ministry of Health, School of Public Health, Jilin University, Changchun, 130021, China
| | - Ling Qiu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yanming Yang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yunlong Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhishen Chen
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhongshan Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Lei Yu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130041, China
| |
Collapse
|