1
|
Cui B, Cheng X, Zhang X, Chen L, Pang W, Liu Y, Yang Z, Li H, He X, Li X, Bi X. Anti-cancer activity and mechanism of flurbiprofen organoselenium compound RY-1-92 in non-small cell lung cancer. RSC Med Chem 2024; 15:1737-1745. [PMID: 38784458 PMCID: PMC11110739 DOI: 10.1039/d4md00058g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/15/2024] [Indexed: 05/25/2024] Open
Abstract
Lung cancer is one of the malignancies with the highest incidence and mortality rates worldwide, and non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer types. In this study, the anti-cancer activities of a novel flurbiprofen organic selenium compound, RY-1-92, on NSCLC cells and a mouse model and the underlying molecular mechanisms were explored. We found that compound RY-1-92 can significantly inhibit the viability, colony formation and migration of A549, NCI-H460 lung cancer cells. Flow cytometry analysis showed that RY-1-92 also can lead to G2/M cell cycle arrest and apoptosis induced in lung cancer cells. Further, RY-1-92 can decrease the tumor size in the Lewis lung cancer tumor-bearing mouse model. The protein levels of cell cycle-related proteins CDK1/cyclinB1 were decreased, while the apoptosis-related protein BAX was increased dramatically after RY-1-92 treatment in vitro and in vivo. Impressively, it was found that TRPV1 might act as a potential molecular target of RY-1-92 using the SEA search server. Furthermore, down-regulation on TRPV1 and its downstream associated factors including p-AKT protein and MAPK signaling pathway-related proteins after RY-1-92 treatment was observed in A549, NCI-H460 lung cancer cells. Taken together, our findings shed light on the potential of RY-1-92 as a novel small molecular drug for NSCLC, and it is of great significance for its further in-depth research and development.
Collapse
Affiliation(s)
- Bo Cui
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xianda Cheng
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xin Zhang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Lili Chen
- College of Life Science, Liaoning University Shenyang 110036 China
- Shenyang Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, College of Life Sciences, Liaoning University Shenyang 110036 China
- College of Mathematics and Statistics, Liaoning University Shenyang 110036 China
| | - Wenqian Pang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Yue Liu
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Zhe Yang
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Hui Li
- College of Life Science, Liaoning University Shenyang 110036 China
| | - Xianran He
- Institute for Interdisciplinary Research, Jianghan University Wuhan Economic and Technological Development Zone Wuhan 430056 China
| | - Xiaolong Li
- Shenzhen Fushan Biological Technology Co., Ltd Kexing Science Park A1 1005, Nanshan Zone Shenzhen 518057 China
| | - Xiuli Bi
- College of Life Science, Liaoning University Shenyang 110036 China
- Shenyang Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, College of Life Sciences, Liaoning University Shenyang 110036 China
| |
Collapse
|
2
|
Zhao C, Xiong K, Bi D, Zhao F, Lan Y, Jin X, Li X. Redox-associated messenger RNAs identify novel prognostic values and influence the tumor immune microenvironment of lung adenocarcinoma. Front Genet 2023; 14:1079035. [PMID: 36873939 PMCID: PMC9977811 DOI: 10.3389/fgene.2023.1079035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Background: An imbalance of redox homeostasis participates in tumorigenesis, proliferation, and metastasis, which results from the production of reactive oxygen species (ROS). However, the biological mechanism and prognostic significance of redox-associated messenger RNAs (ramRNAs) in lung adenocarcinoma (LUAD) still remain unclear. Methods: Transcriptional profiles and clinicopathological information were retrieved from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) of LUAD patients. A total of 31 overlapped ramRNAs were determined, and patients were separated into three subtypes by unsupervised consensus clustering. Biological functions and tumor immune-infiltrating levels were analyzed, and then, differentially expressed genes (DEGs) were identified. The TCGA cohort was divided into a training set and an internal validation set at a ratio of 6:4. Least absolute shrinkage and selection operator regression were used to compute the risk score and determine the risk cutoff in the training set. Both TCGA and GEO cohort were distinguished into a high-risk or low-risk group at the median cutoff, and then, relationships of mutation characteristics, tumor stemness, immune differences, and drug sensitivity were investigated. Results: Five optimal signatures (ANLN, HLA-DQA1, RHOV, TLR2, and TYMS) were selected. Patients in the high-risk group had poorer prognosis, higher tumor mutational burden, overexpression of PD-L1, and lower immune dysfunction and exclusion score compared with the low-risk group. Cisplatin, docetaxel, and gemcitabine had significantly lower IC50 in the high-risk group. Conclusion: This study constructed a novel predictive signature of LUAD based on redox-associated genes. Risk score based on ramRNAs served as a promising biomarker for prognosis, TME, and anti-cancer therapies of LUAD.
Collapse
Affiliation(s)
- Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kewei Xiong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.,School of Mathematics and Statistics, Central China Normal University, Wuhan, China
| | - Dong Bi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangrui Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanfang Lan
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaorui Jin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangpan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Abbasian MH, Ardekani AM, Sobhani N, Roudi R. The Role of Genomics and Proteomics in Lung Cancer Early Detection and Treatment. Cancers (Basel) 2022; 14:5144. [PMID: 36291929 PMCID: PMC9600051 DOI: 10.3390/cancers14205144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 08/17/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, with non-small-cell lung cancer (NSCLC) being the primary type. Unfortunately, it is often diagnosed at advanced stages, when therapy leaves patients with a dismal prognosis. Despite the advances in genomics and proteomics in the past decade, leading to progress in developing tools for early diagnosis, targeted therapies have shown promising results; however, the 5-year survival of NSCLC patients is only about 15%. Low-dose computed tomography or chest X-ray are the main types of screening tools. Lung cancer patients without specific, actionable mutations are currently treated with conventional therapies, such as platinum-based chemotherapy; however, resistances and relapses often occur in these patients. More noninvasive, inexpensive, and safer diagnostic methods based on novel biomarkers for NSCLC are of paramount importance. In the current review, we summarize genomic and proteomic biomarkers utilized for the early detection and treatment of NSCLC. We further discuss future opportunities to improve biomarkers for early detection and the effective treatment of NSCLC.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Ali M. Ardekani
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Seven ZGT, Özen D, Özyazgan S. Pharmacogenomic Biomarkers. Biomark Med 2022. [DOI: 10.2174/9789815040463122010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Why does the usual dose of medication work for a person while another
individual cannot give the expected response to the same drug? On the other hand, how
come half of the usual dose of an analgesic relieves an individual’s pain immediately,
as another man continue to suffer even after taking double dose? Although a treatment
method has been successfully used in majority of the population for many years, why
does the same therapy cause serious side effects in another region of the world? Most
presently approved therapies are not effective in all patients. For example, 20-40% of
patients with depression respond poorly or not at all to antidepressant drug therapy.
Many patients are resistant to the effects of antiasthmatics and antiulcer drugs or drug
treatment of hyperlipidemia and many other diseases. The reason for all those is
basically interindividual differences in genomic structures of people, which are
explained in this chapter in terms of the systems and the most frequently used drugs in
clinical treatment.
Collapse
Affiliation(s)
- Zeynep Gizem Todurga Seven
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-
Cerrahpasa, Istanbul, Turkey
| | - Deniz Özen
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-
Cerrahpasa, Istanbul, Turkey
| | - Sibel Özyazgan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-
Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
5
|
Sun XY, Lin Y, Guo W, Yin XM. Prognostic Value of the Geriatric Nutritional Risk Index in Patients with Non-Small Cell Lung Cancer: A Meta-Analysis. Nutr Cancer 2022; 74:3623-3633. [PMID: 35838018 DOI: 10.1080/01635581.2022.2096244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study aimed to quantitatively identify the prognostic and clinicopathological value of the geriatric nutritional risk index (GNRI) in non-small cell lung cancer (NSCLC) through a meta-analysis. The electronic databases PubMed, Web of Science, Embase, and Cochrane Library were thoroughly searched from inception to December 14, 2021. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to evaluate the prognostic value of GNRI. Odds ratios (ORs) and 95%CIs were combined to estimate the clinicopathological significance of the GNRI in NSCLC. Seven studies with 2,023 patients were included in the meta-analysis. A low GNRI score was significantly associated with poor overall survival (OS) (HR = 2.01, 95%CI = 1.65-2.44, p < 0.001) and worse progression-free survival (PFS), recurrence-free survival (RFS), and cancer-specific survival (CSS) (HR = 1.81, 95%CI = 1.48-2.22, p < 0.001) in NSCLC. Furthermore, a low GNRI score was significantly associated with the histological type of non-adenocarcinoma (OR= 1.55, 95%CI = 1.19-2.03, p = 0.001) and Eastern Cooperative Oncology Group performance status (ECOG PS) ≥2 (OR= 2.81, 95%CI= 1.49-5.32, p = 0.001). A low GNRI score is a significant and effective prognostic marker for poor survival outcomes in patients with NSCLC. In addition, low GNRI score was correlated with higher ECOG PS scores.
Collapse
Affiliation(s)
- Xin-Yu Sun
- Department of Radiation Oncology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yu Lin
- Department of Radiation Oncology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wei Guo
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, Hebei, China
| | - Xiao-Ming Yin
- Department of Radiation Oncology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, Hebei, China
| |
Collapse
|
6
|
Wang Y, He H. Prognostic value of soluble programmed cell death ligand-1 in patients with non-small-cell lung cancer: a meta-analysis. Immunotherapy 2022; 14:945-956. [PMID: 35822688 DOI: 10.2217/imt-2021-0238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Previously published data was collected and a meta-analysis was conducted to precisely identify the prognostic and clinicopathological significance of soluble programmed cell death ligand-1 (sPD-L1) in patients with non-small-cell lung cancer (NSCLC). Materials & methods: Combined hazard ratios (HRs), odds ratios and 95% confidence intervals were used to assess the correlation between sPD-L1 expression and prognosis in patients with NSCLC. Results: A total of 11 studies with 976 patients were included in this meta-analysis. High levels of sPD-L1 were associated with poor overall and progression-free survival (HR: 2.65, 95% CI: 2.32-3.02; p < 0.001 vs HR: 2.02, 95% CI: 1.24-3.29; p = 0.005). sPD-L1 level was not significantly correlated with sex, smoking status, age, Eastern Cooperative Oncology Group performance status, subtype or EGFR mutation. Conclusion: High levels of sPD-L1 are a prognostic marker for poor survival in patients with NSCLC.
Collapse
Affiliation(s)
- Yan Wang
- Clinical Laboratory, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Haiyun He
- Department of Respiration, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| |
Collapse
|
7
|
Yang Q, Yang M, Zhang J, Ma Y. SENP1 Aberrance and Its Linkage to Clinical Features, Adjuvant Regimen, and Prognosis in Patients With Surgical Non-small Cell Lung Cancer Receiving Adjuvant Chemotherapy. Front Surg 2022; 8:771785. [PMID: 35782332 PMCID: PMC9247181 DOI: 10.3389/fsurg.2021.771785] [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: 09/07/2021] [Accepted: 11/26/2021] [Indexed: 11/26/2022] Open
Abstract
Background Small ubiquitin-like modifier-specific protease 1 (SENP1) plays vital roles in cancer progression and chemoresistance, but its prognostic value in non-small cell lung cancer (NSCLC) is vague. This study aimed to explore the correlation of SENP1 with clinical features, adjuvant chemotherapy regimen, and prognosis in patients with surgical NSCLC receiving adjuvant chemotherapy. Methods Tumor and adjacent tissues were collected from 157 patients with surgical NSCLC receiving adjuvant chemotherapy. Meanwhile, tumor tissue and paired adjacent tissue specimens were obtained to evaluate SENP1 protein expression by immunohistochemistry (IHC) assay; among which, 102 pairs were used to detect SENP1 messenger RNA (mRNA) by reverse transcription quantitative PCR. Results SENP1 IHC score and SENP1 mRNA expression were increased in tumor tissue than adjacent tissue (p < 0.001). Besides, elevated SENP1 IHC score was correlated with > 5 cm tumor size (p = 0.045), lymph node metastasis occurrence (p = 0.003), and advanced tumor-node-metastasis (TNM) stage (p = 0.012); meanwhile, increased SENP1 mRNA expression was associated with histopathological subtype (p = 0.011), lymph node metastasis occurrence (p = 0.008), and higher TNM stage (p = 0.015). Besides, no correlation was found in SENP1 IHC score (p = 0.424) or mRNA expression (p = 0.927) with specific adjuvant chemotherapy regimen. Additionally, both the SENP1 protein (high) (p = 0.003) and mRNA high (p = 0.028) were correlated with poor disease-free survival (DFS), while SENP1 protein high was also associated with shorter overall survival (OS) (p = 0.029). Furthermore, SENP1 protein (high vs. low) was independently associated with unsatisfying DFS [p = 0.009, hazard ratio (HR) = 1.798] and OS (p = 0.049, HR = 1.735). Conclusion SENP1 may serve as a potential biomarker to improve the management of patients with surgical NSCLC receiving adjuvant chemotherapy.
Collapse
|
8
|
Alsagaby S, Ahmed AA, Rasheed Z, Althwab SA, Aljohani ASM, Alhumaydhi FA, Alhomaidan HT, Alkhamiss AS, Alkhowailed M, Alaqeel A, Alblihed MA, Alrehaili J, Fernández N, Abdulmonem WA. Association of genetic polymorphisms in DNA repair genes ERCC2 Asp312Asn (rs1799793), ERCC2 Lys 751 Gln (rs13181), XRCC1 Arg399 Gln (rs25487) and XRCC3 Thr 241Met (rs861539) with the susceptibility of lung cancer in Saudi population. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:530-554. [PMID: 35319340 DOI: 10.1080/15257770.2022.2052317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
This study demonstrated the association of polymorphisms in ERCC2 (Asp312Asn) rs1799793, ERCC2 (Lys751Gln) rs13181, XRCC1 (Arg399Gln) rs25487 and XRCC3(Thr241Met) rs861539 polymorphisms with a susceptibility of lung cancer (LC) onset in the Saudi population. The study was performed on 134 LC patients and 270 controls. The data revealed that there was no significant association of LC with subtype squamous cell carcinoma (SCC), small cell lung cancer (SCLC) and adenocarcinoma with the ERCC2 rs1799793 polymorphism. The data showed that the CC genotype for ERCC2 rs13181, the AA genotype for XRCC1 rs25487, and the genotype TT for XRCC3 rs861539 were significantly associated with SCC susceptibility (p < 0.05). Similarly, the CC genotype for ERCC2 rs13181 and the AA genotype for XRCC1 rs25487 were significantly associated with adenocarcinoma susceptibility (p < 0.05). Whereas, the TT genotype for XRCC3 rs861539 was significantly associated with SCLC susceptibility (p = 0.005). In total, significant association of LC susceptibility was found in the following combination models of recessive genotypes: AC heterozygous for ERCC2 rs13181 + AA homozygous for XRCC1 rs25487, CC homozygous for ERCC2 rs13181 + GA heterozygous for rs25487, CC homozygous for rs13181 + AA homozygous for XRCC1 rs25487, CC homozygous for ERCC2 rs13181 + TT homozygous for XRCC3 rs861539, GA heterozygous for XRCC1 rs25487 + CT heterozygous for XRCC3 rs861539, GA heterozygous for XRCC1 rs25487 + TT homozygous for XRCC3 rs861539, AA homozygous for XRCC1 rs25487 + CT heterozygous for XRCC3 rs861539, AA homozygous for XRCC1 rs25487+ TT homozygous for XRCC3 rs861539. These data clearly demonstrated that the combination of recessive genotypes may be associated with susceptibility of LC onset (p < 0.05). In short, the data indicated that DNA repair genes increase LC risk via gene-gene interaction rather than independent variants.
Collapse
Affiliation(s)
- Suliman Alsagaby
- Department of Medical Laboratories Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | - Ahmed A Ahmed
- Research Center, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Sami A Althwab
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agricultural and Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Homaidan T Alhomaidan
- Department of Family and Community Medicine, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Abdullah S Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Mohammad Alkhowailed
- Department of Dermatology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Aqeel Alaqeel
- Department of Pediatrics, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Mohamd A Alblihed
- Department of Medical Microbiology, School of Medicine, Taif University, Taif, Saudi Arabia
| | - Jihad Alrehaili
- Pathology Department, Imam Mohammad Ibn Saud University, Riyadh, Saudi Arabia
| | | | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Qassim, Saudi Arabia
| |
Collapse
|
9
|
Su Y, Chen R, Han Z, Xu R, Ma L, Wufuli R, Liu H, Wang F, Ma L, Chen R, Liu J. Clinical and Prognostic Significance of CD117 in Non-Small Cell Lung Cancer: A Systemic Meta-Analysis. Pathobiology 2021; 88:267-276. [PMID: 34107476 DOI: 10.1159/000514386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/24/2020] [Indexed: 12/09/2022] Open
Abstract
The aim of this study was to assess the relationship of cluster of differentiation 117 (CD117) expression with the clinicopathological characteristics and the prognosis in patients with non-small cell lung cancer (NSCLC). No meta-analysis concerning the correlation of CD117 expression with clinical and prognostic values of the patients with NSCLC is reported. A systematic literature search was conducted to achieve eligible studies. The combined odds ratios (ORs) or hazard ratios (HRs: multivariate Cox analysis) with their 95% confidence intervals (CIs) were calculated in this analysis. Final 17 eligible studies with 4,893 NSCLC patients using immunohistochemical detection were included in this meta-analysis. CD117 expression was not correlated with gender (male vs. female), clinical stage (stages 3-4 vs. stages 1-2), tumor grade (grade 3 vs. grades 1-2), T-stage (T-stages 3-4 vs. T-stages 0-2), distal metastasis, and disease-free survival (DFS) of NSCLC (all p values >0.05). CD117 expression was associated with lymph node metastasis (positive vs. negative: OR = 0.74, 95% CI = 0.56-0.97, p = 0.03), histological type (adenocarcinoma (AC) versus squamous cell carcinoma (SCC): OR = 1.74, 95% CI = 1.26-2.39, p = 0.001), and a worse overall survival (OS) (HR = 1.89, 95% CI = 1.22-2.92, p = 0.004). The expression of CD117 was significantly higher in AC than in SCC. CD117 may be an independent prognostic indicator for worse OS in NSCLC.
Collapse
Affiliation(s)
- Ying Su
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Ru Chen
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Zhongcheng Han
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Rong Xu
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Lili Ma
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Reyina Wufuli
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Hongbo Liu
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Fang Wang
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Lei Ma
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Rui Chen
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| | - Jiang Liu
- Department of Oncology, People's Hospital of Xinjiang Uygur, Urumqi, China
| |
Collapse
|
10
|
Jia B, Zhao X, Wu D, Dong Z, Chi Y, Zhao J, Wu M, An T, Wang Y, Zhuo M, Li J, Chen X, Tian G, Long J, Yang X, Chen H, Wang J, Zhai X, Li S, Li J, Ma M, He Y, Kong L, Brcic L, Fang J, Wang Z. Identification of serum biomarkers to predict pemetrexed/platinum chemotherapy efficacy for advanced lung adenocarcinoma patients by data-independent acquisition (DIA) mass spectrometry analysis with parallel reaction monitoring (PRM) verification. Transl Lung Cancer Res 2021; 10:981-994. [PMID: 33718037 PMCID: PMC7947410 DOI: 10.21037/tlcr-21-153] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Pemetrexed/platinum chemotherapy has been the standard chemotherapy regimen for lung adenocarcinoma patients, but the efficacy varies considerably. Methods To discover new serum biomarkers to predict the efficacy of pemetrexed/platinum chemotherapy, we analyzed 20 serum samples from advanced lung adenocarcinoma patients who received pemetrexed/platinum chemotherapy with the data-independent acquisition (DIA) quantitative mass spectrometry (MS). Results The 20 patients were categorized as “good response” [12 patients achieving partial response (PR)] and “poor response” [8 patients with progressive disease (PD)] groups. Altogether 23 significantly different expressed proteins were identified, which had relative ratios higher than 1.2 or lower than –0.83, with 7 proteins having an area under the curve (AUC) above 0.8. To further validate the DIA results, we used the parallel reaction monitoring (PRM) method to examine 16 candidate serum biomarkers in the study cohort of 20 patients and another cohort of 22 advanced lung adenocarcinoma patients (16 PR and 6 PD). Quantitative validation using PRM correlated well with the DIA results, and 10 promising proteins exhibited a similar up- or downregulation. It is worth noting that glutathione peroxidase 3 (GPX3) exhibits significant upregulation in the poor response group compared with the good response group, which was validated by both DIA and PRM methods. Conclusions Our study confirmed that combined DIA MS and PRM approaches were effective in identifying serum predictive biomarkers for advanced lung adenocarcinoma patients. Further studies are needed to explore the potential biological mechanism underlying these biomarkers.
Collapse
Affiliation(s)
- Bo Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinghui Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Di Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi Dong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of GI Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yujia Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Meina Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Tongtong An
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoling Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Guangming Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jieran Long
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hanxiao Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jingjing Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoyu Zhai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Junfeng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Menglei Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuling He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lingdong Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jian Fang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Oncology II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziping Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
11
|
Pharmacogenetic Association between XRCC1 Polymorphisms and Response to Platinum-Based Chemotherapy in Asian Patients with NSCLC: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3520764. [PMID: 33150172 PMCID: PMC7603545 DOI: 10.1155/2020/3520764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/25/2020] [Indexed: 01/18/2023]
Abstract
Background Platinum-based chemotherapy plays an antitumor role by damaging DNA. X-ray repair crosscomplementing protein 1 (XRCC1) participates in DNA repair and thus affects the sensitivity to platinum drugs. Two polymorphisms of XRCC1, rs25487 (Arg399Gln) and rs1799782 (Arg194Trp), have been widely studied for the association with clinical outcomes of platinum-based chemotherapy in Asian patients with non-small-cell lung cancer (NSCLC), but the results remain inconclusive. Thus, we performed the present meta-analysis. Methods Literature search was performed in PubMed, Web of Science, and EMBASE up to June 2019. Odds ratios (ORs) for objective response ratio (ORR), Cox proportional hazard ratios (HRs) of overall survival (OS) and progression-free survival (PFS), and the corresponding 95% confidence intervals (95% CIs) were calculated to assess the association strengths between XRCC1 polymorphisms and clinical outcomes. Comparisons were performed in homozygous, heterozygous, dominant, and recessive models. Results Finally, a total of 23 studies involving 5567 patients were included in the meta-analysis. Compared to ArgArg of rs25487, GlnGln (OR = 1.71, 95% CI: 1.16-2.52, p = .007, I2 = 56.8%) and GlnArg (OR = 1.23, 95% CI: 1.07-1.40, p = .003, I2 = 29.0%) were associated with higher ORR. Meanwhile, GlnGln indicated a favorable OS (HR = 0.60, 95% CI: 0.40-0.88) and PFS (HR = 0.64, 95% CI: 0.46-0.90). We also found positive associations between rs1799782 and ORR in all comparison models with low between-study heterogeneity. The association strength increased with the number of variant alleles (TrpTrp vs. ArgArg: OR = 1.73, 95% CI:1.31-2.27; TrpArg vs. ArgArg: OR = 1.28, 95% CI: 1.06-1.55), suggesting a gene dosage effect. In addition, TrpTrp predicted a longer OS. Conclusion Our results showed that rs25487 and rs1799782 of XRCC1 are potential markers to predict clinical outcomes of platinum-based chemotherapy in Asian patients with NSCLC.
Collapse
|
12
|
Zeng B, Ge C, Li R, Zhang Z, Fu Q, Li Z, Lin Z, Liu L, Xue Y, Xu Y, He J, Guo H, Li C, Huang W, Song X, Huang Y. Knockdown of microsomal glutathione S-transferase 1 inhibits lung adenocarcinoma cell proliferation and induces apoptosis. Biomed Pharmacother 2020; 121:109562. [DOI: 10.1016/j.biopha.2019.109562] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/04/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022] Open
|
13
|
Capula M, Mantini G, Funel N, Giovannetti E. New avenues in pancreatic cancer: exploiting microRNAs as predictive biomarkers and new approaches to target aberrant metabolism. Expert Rev Clin Pharmacol 2019; 12:1081-1090. [PMID: 31721608 DOI: 10.1080/17512433.2019.1693256] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Introduction: Most pancreatic cancer patients are diagnosed at advanced-stages and first-line regimens (FOLFIRINOX and gemcitabine/nab-paclitaxel) provide limited survival advantage and are associated with considerable toxicities. In this grim scenario, novel treatments and biomarkers are warranted.Areas covered: MicroRNAs (miRNAs) emerged as biomarkers for cancer prognosis and chemoresistance and blood-based miRNAs are being evaluated as indicators of therapeutic activity. Moreover, aberrant metabolism, such as aerobic glycolysis, has been correlated to tumor aggressiveness and poor prognosis. Against this background, innovative approaches to tackle metabolic aberrations are being implemented and glycolytic inhibitors targeting lactate dehydrogenase-A (LDH-A) showed promising effects in preclinical models. A PubMed search was used to compile relevant publications until February 2019.Expert opinion: Analysis of tissue/circulating miRNA might improve selection for optimal treatment regimens. For instance, miR-181a modulation seems to predict response to FOLFIRINOX. However, we need further studies to validate predictive miRNA profiles, as well as to exploit miRNAs for treatment-tailoring. Several miRNAs have also a key role in regulating metabolic aberrations. Since preliminary evidence supports the development of new agents targeting these aberrations, such as LDH-A inhibitors, the identification of biomarkers for these treatments, including the above-mentioned miRNAs, should shorten the gap between preclinical studies and personalized therapies.
Collapse
Affiliation(s)
- Mjriam Capula
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza Pisa, Pisa, Italy
| | - Giulia Mantini
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza Pisa, Pisa, Italy
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza Pisa, Pisa, Italy
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisa per la Scienza Pisa, Pisa, Italy
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
El Hassouni B, Li Petri G, Liu DSK, Cascioferro S, Parrino B, Hassan W, Diana P, Ali A, Frampton AE, Giovannetti E. Pharmacogenetics of treatments for pancreatic cancer. Expert Opin Drug Metab Toxicol 2019; 15:437-447. [PMID: 31100206 DOI: 10.1080/17425255.2019.1620731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Despite clinical efforts, pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis. The scarcity of effective therapies can be reflected by the lack of reliable biomarkers to adapt anticancer drugs prescription to tumors' and patients' features. Areas covered: Pharmacogenetics should provide the way to select patients who may benefit from a specific therapy that best matches individual and tumor genetic profile, but it has not yet led to gains in outcome. This review describes PDAC pharmacogenetics findings, critically reappraising studies on polymorphisms and -omics profiles correlated to response to gemcitabine, FOLFIRINOX, and nab-paclitaxel combinations, as well as limitations of targeted therapies. Further, we question whether personalized approaches will benefit patients to any significant degree, supporting the need of new strategies within well-designed trials and validated genomic tests for treatment decision-making. Expert opinion: A major challenge in PDAC is the identification of subgroups of patients who will benefit from treatments. Minimally-invasive tests to analyze biomarkers of drug sensitivity/toxicity should be developed alongside anticancer treatments. However, progress might fall below expectations because of tumor heterogeneity and clonal evolution. Whole-genome sequencing and liquid biopsies, as well as prospective validation in selected cohorts, should overcome the limitations of traditional pharmacogenetic approaches.
Collapse
Affiliation(s)
- Btissame El Hassouni
- a Department of Medical Oncology , Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands
| | - Giovanna Li Petri
- a Department of Medical Oncology , Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands.,b Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche , Università degli Studi di Palermo , Palermo , Italy
| | - Daniel S K Liu
- c Department of Surgery and Cancer , Imperial College , London , UK
| | - Stella Cascioferro
- b Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche , Università degli Studi di Palermo , Palermo , Italy
| | - Barbara Parrino
- b Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche , Università degli Studi di Palermo , Palermo , Italy
| | - Waqar Hassan
- a Department of Medical Oncology , Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands
| | - Patrizia Diana
- b Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche , Università degli Studi di Palermo , Palermo , Italy
| | - Asif Ali
- d Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences , University of Glasgow , Glasgow UK.,e Institute of Basic Medical Sciences , Khyber Medical University , Peshawar , Pakistan
| | - Adam E Frampton
- c Department of Surgery and Cancer , Imperial College , London , UK
| | - Elisa Giovannetti
- a Department of Medical Oncology , Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands.,f Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza , Pisa , Italy
| |
Collapse
|
15
|
Pan C, Zhang Y, Meng Q, Dai G, Jiang Z, Bao H. Down Regulation of the Expression of ELMO3 by COX2 Inhibitor Suppresses Tumor Growth and Metastasis in Non-Small-Cell Lung Cancer. Front Oncol 2019; 9:363. [PMID: 31134158 PMCID: PMC6515945 DOI: 10.3389/fonc.2019.00363] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/18/2019] [Indexed: 01/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignancies. Studies have shown that engulfment and cell motility 3 (ELMO3) is highly expressed in NSCLC and can be used as a novel biomarker, but its underlying mechanism remains to be explored. The aim of this study was to investigate the mechanism by which ELMO3 may be down-regulated by COX-2 inhibitors to inhibit NSCLC. NSCLC tissue and adjacent normal lung tissue from 24 patients were used to detect the mRNA and protein expression of ELMO3, COX-2, and other related proteins by Western blot, RT-PCR, and Immunohistochemical analysis. Lewis Lung carcinoma (LLC) cells were used to investigate the effects and the mechanism of siELMO3 and COX-2 inhibitor. C57BL/6 mice inoculated with LLC cells by subcutaneous (s.c.) injection were used to detect the in vivo effects of cox-2 inhibitor. The expression of ELMO3 and cyclooxygenase-2 (COX-2) in human NSCLC tissues was significantly increased compared with that in the adjacent normal tissues. ELMO3 exhibited a positive correlation with COX-2 expression. Moreover, knockdown of ELMO3 suppressed the epithelial-mesenchymal transition (EMT), adhesion, and metastasis of Lewis lung carcinoma (LLC) cells. Importantly, Parecoxib, a selective inhibitor of COX-2, significantly reduced the expression of ELMO3 and EMT in LLC cells and LLC-bearing mice. Furthermore, it could inhibit the growth, adhesion and metastasis of LLC cells in vitro. Our results demonstrate that down regulation of ELMO3 suppressed growth and metastasis of lung cancer by inhibiting EMT. Parecoxib could reduce ELMO3 expression and suppress growth and metastasis of lung cancer, which might be a useful chemotherapeutic agent for inhibiting metastasis and recurrence of NSCLC.
Collapse
Affiliation(s)
- Cailong Pan
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qinghai Meng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoliang Dai
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhitao Jiang
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Barati Bagherabad M, Afzaljavan F, ShahidSales S, Hassanian SM, Avan A. Targeted therapies in pancreatic cancer: Promises and failures. J Cell Biochem 2018; 120:2726-2741. [PMID: 28703890 DOI: 10.1002/jcb.26284] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/11/2018] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an incidence rate nearly equal to its mortality rate. The poor prognosis of the disease can be explained by the absence of effective biomarkers for screening and early detection, together with the aggressive behavior and resistance to the currently available chemotherapy. The therapeutic failure can also be attributed to the inter-/intratumor genetic heterogeneity and the abundance of tumor stroma that occupies the majority of the tumor mass. Gemcitabine is used in the treatment of PDAC; however, the response rate is less than 12%. A recent phase III trial revealed that the combination of oxaliplatin, irinotecan, fluorouracil, and leucovorin could be an option for the treatment of metastatic PDAC patients with good performance status, although these approaches can result in high toxicity level. Further investigations are required to develop innovative anticancer agents that either improve gemcitabine activity, within novel combinatorial approaches or acts with a better efficacy than gemcitabine. The aim of the current review is to give an overview of preclinical and clinical studies targeting key dysregulated signaling pathways in PDAC.
Collapse
Affiliation(s)
- Matineh Barati Bagherabad
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Afzaljavan
- Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research center, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine group, Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Zeng Z, Li D, Yu T, Huang Y, Yan H, Gu L, Yuan J. Association and clinical implication of the USP10 and MSH2 proteins in non-small cell lung cancer. Oncol Lett 2018; 17:1128-1138. [PMID: 30655874 PMCID: PMC6312927 DOI: 10.3892/ol.2018.9702] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 10/12/2018] [Indexed: 12/22/2022] Open
Abstract
Ubiquitin-specific protease 10 (USP10) is involved in a number of biological processes by stabilizing several proteins, which have been implicated in multiple stages of tumorigenesis and progression. Previous studies have indicated that USP10 stabilizes and deubiquitinates MutS homolog 2 (MSH2) in in vitro and in vivo models. The level of MSH2 protein has been positively correlated with that of the USP10 protein in a panel of lung cancer cell lines. Furthermore, depletion of USP10 in lung cancer cells causes decreased apoptosis and increased cell survival upon treatment with DNA-damaging agents. However, the expression and clinical implication of USP10 protein in lung cancer tissues is not clear. Additionally, whether the level of MSH2 protein is positively correlated with that of the USP10 protein in lung cancer tissues also remains unresolved. Therefore, USP10 protein expression was detected in 148 human non-small cell lung cancer (NSCLC) and 139 non-cancerous lung tissues using immunohistochemistry, whereas mRNA was investigated by Gene Expression Omnibus dataset and The Cancer Genome Atlas database analyses. It was identified that USP10 protein expression was significantly downregulated in NSCLC tissues compared with in normal lung tissues (P<0.05). However, no significant difference in USP10 mRNA expression between the two tissues was identified. In addition, a positive correlation was observed between the USP10 and MSH2 proteins in NSCLC tissues (P<0.05). However, the clinicopathological features and survival analysis indicated that the USP10 and MSH2 proteins were not associated with clinical features, including age, sex, tumor size, Tumor-Node-Metastasis stage and tumor cell differentiation, along with the prognosis of NSCLC. Collectively, these results suggest that downregulation of USP10 protein serves an important function in the tumorigenesis of NSCLC, and the level of USP10 protein is positively correlated with that of MSH2 protein in NSCLC tissues, which may indicate that USP10 also stabilizes the MSH2 protein in patients with lung cancer.
Collapse
Affiliation(s)
- Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Dan Li
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Yu
- Integrated Traditional Chinese and Western Medicine Ward, Oncology Department, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Yabing Huang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
18
|
Li S, Shi D, Zhang L, Yang F, Cheng G. Oridonin enhances the radiosensitivity of lung cancer cells by upregulating Bax and downregulating Bcl-2. Exp Ther Med 2018; 16:4859-4864. [PMID: 30546402 DOI: 10.3892/etm.2018.6803] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/22/2017] [Indexed: 12/17/2022] Open
Abstract
Oridonin is an active component of the traditional Chinese herb Rabdosia rubescens. The present study aimed to evaluate the antitumor effects of oridonin on human non-small cell lung cancer (NSCLC) cells and explore whether oridonin could enhance their radiosensitivity. Oridonin was demonstrated to inhibit the proliferation of SPC-A-1 and HCC827 lung cancer cells in a time- and dose-dependent manner, which was detected using the MTT assay. In addition, pretreatment with oridonin for 24 h prior to irradiation was identified to enhance the radiosensitivity of SPC-A-1 cells. Furthermore, the levels of apoptosis regulator BAX (Bax) and apoptosis regulator Bcl-2 (Bcl-2) were detected by western blotting analysis. The results demonstrated that the level of Bax was increased and the level of Bcl-2 was decreased in SPC-A-1 cells treated with oridonin and irradiation compared with the group that received irradiation alone. These results indicate that oridonin may have a novel application as a radiosensitizing agent for the treatment of human NSCLC.
Collapse
Affiliation(s)
- Sirui Li
- Department of Medical Oncology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163111, P.R. China
| | - Dan Shi
- Department of Radiation, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Liangyu Zhang
- Department of Medical Oncology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163111, P.R. China
| | - Fang Yang
- Department of Medical Oncology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163111, P.R. China
| | - Guanghui Cheng
- Department of Radiation, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
19
|
Petri GL, Cascioferro S, Parrino B, Peters GJ, Diana P, Giovannetti E. Proton-coupled folate transporter as a biomarker of outcome to treatment for pleural mesothelioma. Pharmacogenomics 2018; 19:811-814. [PMID: 29916298 DOI: 10.2217/pgs-2018-0071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Giovanna Li Petri
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, via Paradisa, 56100, Pisa, Italy
| |
Collapse
|
20
|
Toffalorio F, Santarpia M, Radice D, Jaramillo CA, Spitaleri G, Manzotti M, Catania C, Jordheim LP, Pelosi G, Peters GJ, Tibaldi C, Funel N, Spaggiari L, de Braud F, De Pas T, Giovannetti E. 5'-nucleotidase cN-II emerges as a new predictive biomarker of response to gemcitabine/platinum combination chemotherapy in non-small cell lung cancer. Oncotarget 2018; 9:16437-16450. [PMID: 29662657 PMCID: PMC5893252 DOI: 10.18632/oncotarget.24505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 02/02/2018] [Indexed: 02/04/2023] Open
Abstract
A number of pharmacogenetic studies have been carried out in non-small-cell lung cancer (NSCLC) to identify and characterize genes involved in chemotherapy activity. However, the results obtained so far are controversial and no reliable biomarker is currently used to predict clinical benefit from platinum-based chemotherapy, which represents the cornerstone of treatment of advanced NSCLC. This study investigated the expression levels of ERCC1 and of six genes (RRM1, RRM2, hENT1, dCK, cN-II and CDA) involved in gemcitabine metabolism in locally/advanced NSCLC patients treated with gemcitabine/platinum combination. Gene expression was assessed by quantitative-PCR in laser-microdissected specimens and correlated with tumor response. Frequency distribution of responses above and below the median expression level of biomarkers was compared using a two-sided Fisher's test. 5'-nucleotidase (cN-II) was the only gene differently expressed (p = 0.016) in the responders (complete/partial-response) compared to non-responders (stable/progressive disease). In the multivariate analysis, overexpression of this catabolic enzyme of gemcitabine remained a significant negative predictive factor. Patients with low cN-II had a modest trend toward increased survival, while both survival and progression-free survival were significantly longer in a more homogenous validation cohort of 40 advanced NSCLC (8.0 vs. 5.1 months, p = 0.026). Moreover, in vitro studies showed that silencing or pharmacological inhibition of cN-II increased the cytotoxicity of gemcitabine. This is the first study demonstrating the role of cN-II as a predictor of response to gemcitabine/platinum combinations in NSCLC. Its validation in prospective studies may improve clinical outcome of selected patients.
Collapse
Affiliation(s)
- Francesca Toffalorio
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
- Medical Affairs, Roche Spa, Monza, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
- Medical Oncology Unit, Department of Human Pathology, University of Messina, Messina, Italy
| | - Davide Radice
- Epidemiology and Biostatistics Division, European Institute of Oncology, Milan, Italy
| | | | - Gianluca Spitaleri
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
- Thoracic Oncology Division, European Institute of Oncology, Milan, Italy
| | - Michela Manzotti
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Chiara Catania
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
- Thoracic Oncology Division, European Institute of Oncology, Milan, Italy
| | - Lars Petter Jordheim
- Centre de Recherche en Cancérologie de Lyon, INSERM 1052/CNRS UMR 5286, Lyon, France
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Inter-Hospital Pathology Division, Science and Technology Park, IRCCS MultiMedica, Milan, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Carmelo Tibaldi
- Division of Oncology, Department of Oncology, S. Luca Hospital, Lucca, Italy
| | - Niccola Funel
- CNR-Nano, Institute of Nanoscience and Nanotechnology, Pisa, Italy
- Cancer Pharmacology Laboratory, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Lorenzo Spaggiari
- Thoracic Surgery Division, European Institute of Oncology, Milan, Italy
| | - Filippo de Braud
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Inter-Hospital Pathology Division, Science and Technology Park, IRCCS MultiMedica, Milan, Italy
| | - Tommaso De Pas
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
- CNR-Nano, Institute of Nanoscience and Nanotechnology, Pisa, Italy
- Cancer Pharmacology Laboratory, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
21
|
Kukula-Koch W, Grabarska A, Łuszczki J, Czernicka L, Nowosadzka E, Gumbarewicz E, Jarząb A, Audo G, Upadhyay S, Głowniak K, Stepulak A. Superior anticancer activity is demonstrated by total extract of Curcuma longa L. as opposed to individual curcuminoids separated by centrifugal partition chromatography. Phytother Res 2018; 32:933-942. [PMID: 29368356 DOI: 10.1002/ptr.6035] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 02/01/2023]
Abstract
Three curcuminoids: bisdemethoxycurcumin, demethoxycurcumin, and curcumin from turmeric were successfully separated by a high capacity solvent system composed of heptane: chloroform: methanol: water mixture (5: 6: 3: 2 v/v/v/v) tailored for centrifugal partition chromatographs at K-values of 0.504, 1.057, 1.644, respectively. These three ferulic acid derivatives obtained at a purity rate exceeding 95% were analysed by an HPLC-MS spectrometer. Turmeric extract inhibited the proliferation/viability of A549 human lung cancer, HT29 colon cancer, and T98G glioblastoma cell lines in (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) tetrazolium reduction assay (MTT). Single curcuminoids significantly decreased the viability/proliferation of lung cancer cells in a dose-dependent manner. However, total extract displayed the superior anticancer activity in the investigated cell lines. Crude extract in combination with cisplatin augmented the decrease in the viability of cancer cells compared with single compound treatment in A549 lung cancer cells. Total extract of Curcuma longa could be regarded as being more effective against lung cancer cells in vitro than its separated compounds.
Collapse
Affiliation(s)
- Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| | - Aneta Grabarska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| | - Jarogniew Łuszczki
- Department of Pathophysiology, Medical University of Lublin, 8 Jaczewskiego, 20-090, Lublin, Poland
| | - Lidia Czernicka
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodźki., 20-093, Lublin, Poland
| | - Ewa Nowosadzka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| | - Ewelina Gumbarewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| | - Agata Jarząb
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| | - Gregoire Audo
- Armen Instrument Application Laboratory, ZI Kermelin, 56890, Saint Avé, France
| | - Shakti Upadhyay
- Immunocon Biotech Sp. z o. o., 39A Zana, 20-601, Lublin, Poland
| | - Kazimierz Głowniak
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland.,Department of Cosmetology, University of Information Technology and Management, 2 Sucharskiego, 35-225, Rzeszów, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki, 20-093, Lublin, Poland
| |
Collapse
|
22
|
Cao Y, Wei M, Li B, Liu Y, Lu Y, Tang Z, Lu T, Yin Y, Qin Z, Xu Z. Functional role of eukaryotic translation initiation factor 4 gamma 1 (EIF4G1) in NSCLC. Oncotarget 2018; 7:24242-51. [PMID: 27003362 PMCID: PMC5029698 DOI: 10.18632/oncotarget.8168] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/02/2016] [Indexed: 01/04/2023] Open
Abstract
Eukaryotic translation initiation factor 4 gamma 1(EIF4G1) is related to tumorigenesis and tumor progression. However, its role and the underlying mechanisms in the regulation of tumor development in non-small cell lung cancers (NSCLC) remain largely unknown. Here we report that the levels of EIF4G1 expression are much higher in NSCLC cell lines and tumor tissues than those in the normal lung cells and adjacent normal tissues from the same patients. Using shRNA to knock down EIF4G1 expression stably, we found EIF4G1 required for NSCLC cell proliferation, anchorage-independent growth, migration and invasion. Furthermore, silencing of EIF4G1 induces NSCLC cell apoptosis and causes G0/G1 cell cycle arrest. To identify the partner protein network of EIF4G1 in NSCLC cells, we found that Ubiquitin-specific protease 10 (USP10) can directly interacts with EIF4G1, while acting as a negative regulator for EIF4G1-mediated functions. Together, our results indicate that EIF4G1 functions as an oncoprotein during NSCLC development, which may represent a novel and promising therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Yueyu Cao
- Department of Oncology, Shanghai East Hospital, Dalian Medical University, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Mengdan Wei
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bing Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yali Liu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Ying Lu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhipeng Tang
- Department of Oncology, Shanghai East Hospital, Dalian Medical University, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Tianbao Lu
- Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yujiao Yin
- Department of Oncology, Shanghai East Hospital, Dalian Medical University, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhiqiang Qin
- Department of Oncology, Shanghai East Hospital, Dalian Medical University, Shanghai 200120, China.,Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Departments of Microbiology/Immunology/Parasitology, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Zengguang Xu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Research Center for Translational Medicine and Key Laboratory of Arrhythmias, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|
23
|
Hamilton G, Rath B. Pharmacogenetics of platinum-based chemotherapy in non-small cell lung cancer: predictive validity of polymorphisms of ERCC1. Expert Opin Drug Metab Toxicol 2017; 14:17-24. [PMID: 29226731 DOI: 10.1080/17425255.2018.1416095] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The efficacy of platinum-based chemotherapy for patients with non-small cell lung cancer (NSCLC) is limited by chemoresistance. Platinum drugs damage DNA by introducing intrastrand and interstrand crosslinks which result in cell death. Excision repair cross-complementing 1 (ERCC1) is a member of the nucleotide excision repair (NER) pathway which erases such defects. Single nucleotide polymorphisms (SNPs) in ERCC1 impair this activity and have been suggested to predict the response to chemotherapy. Area covered: Among the polymorphisms of proteins involved in uptake, metabolism, cytotoxicity and efflux of platinum drugs, codon 118 C/T and C8092A in ERCC1 are the best characterized SNPs studied for their predictive power. Here, the divergent results for studies of these markers in NSCLC are summarized and the reasons for this contradictory data discussed. Expert opinion: Cytotoxicity of platinum compounds comprise complex cellular processes for which DNA repair may not constitute the rate limiting step. These drugs are administered as doublets to histologically diverse patients and, furthermore, the NER pathway in ERCC1 wildtype cohorts may be still impaired by the chemotherapeutics applied. At present, assessment of a limited number of polymorphism in DNA repair proteins is not reliably associated with response to treatment in NSCLC patients.
Collapse
Affiliation(s)
- Gerhard Hamilton
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| | - Barbara Rath
- a Department of Surgery , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
24
|
Van Der Steen N, Rolfo CD, Pauwels P, Peters GJ, Giovannetti E. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors and Chemotherapy: A Glimmer of Hope? J Clin Oncol 2017; 35:692-693. [PMID: 27918717 DOI: 10.1200/jco.2016.70.1987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Nele Van Der Steen
- Nele Van Der Steen, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; and VU Medical Center, Amsterdam, the Netherlands; Christian Diego Rolfo and Patrick Pauwels, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; Godefridus J. Peters, VU Medical Center, Amsterdam, the Netherlands; and Elisa Giovannetti, VU Medical Center, Amsterdam, The Netherlands; and University of Pisa, Pisa, Italy
| | - Christian Diego Rolfo
- Nele Van Der Steen, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; and VU Medical Center, Amsterdam, the Netherlands; Christian Diego Rolfo and Patrick Pauwels, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; Godefridus J. Peters, VU Medical Center, Amsterdam, the Netherlands; and Elisa Giovannetti, VU Medical Center, Amsterdam, The Netherlands; and University of Pisa, Pisa, Italy
| | - Patrick Pauwels
- Nele Van Der Steen, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; and VU Medical Center, Amsterdam, the Netherlands; Christian Diego Rolfo and Patrick Pauwels, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; Godefridus J. Peters, VU Medical Center, Amsterdam, the Netherlands; and Elisa Giovannetti, VU Medical Center, Amsterdam, The Netherlands; and University of Pisa, Pisa, Italy
| | - Godefridus J Peters
- Nele Van Der Steen, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; and VU Medical Center, Amsterdam, the Netherlands; Christian Diego Rolfo and Patrick Pauwels, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; Godefridus J. Peters, VU Medical Center, Amsterdam, the Netherlands; and Elisa Giovannetti, VU Medical Center, Amsterdam, The Netherlands; and University of Pisa, Pisa, Italy
| | - Elisa Giovannetti
- Nele Van Der Steen, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; and VU Medical Center, Amsterdam, the Netherlands; Christian Diego Rolfo and Patrick Pauwels, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; Godefridus J. Peters, VU Medical Center, Amsterdam, the Netherlands; and Elisa Giovannetti, VU Medical Center, Amsterdam, The Netherlands; and University of Pisa, Pisa, Italy
| |
Collapse
|
25
|
Meng M, Liao H, Zhang B, Pan Y, Kong Y, Liu W, Yang P, Huo Z, Cao Z, Zhou Q. Cigarette smoke extracts induce overexpression of the proto-oncogenic gene interleukin-13 receptor α2 through activation of the PKA-CREB signaling pathway to trigger malignant transformation of lung vascular endothelial cells and angiogenesis. Cell Signal 2017; 31:15-25. [PMID: 27986643 DOI: 10.1016/j.cellsig.2016.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/03/2016] [Accepted: 12/12/2016] [Indexed: 01/23/2023]
Abstract
Cigarette smoking is a major cause of lung cancer. Tumor-associated endothelial cells (TAECs) play important roles in tumor angiogenesis and metastasis. However, whether cigarette smoking can trigger genesis of lung TAECs has not been reported yet. In the current study, we used lung endothelial cell (EC) lines as a model to study the pathological effect of cigarette smoke extracts (CSEs) on human lung ECs, and found that a lower dose of 4% CSEs obviously caused abnormal morphological changes in ECs, increased the permeability of endothelial monolayer, while a higher concentration of 8% CSEs caused EC apoptosis. Strikingly, CSEs induced a 117-fold overexpression of a pro-tumorigenic interleukin-13 receptor α2 gene (IL-13Rα2, also named as CT-19) through activation of the protein kinase A (PKA) and cAMP response element-binding protein (CREB) signaling pathway. A PKA specific inhibitor H89 completely abolished CSEs-induced IL-13Rα2 overexpression. The overexpression of IL-13Rα2 in lung ECs significantly increased the tumorigenic, migratory, and angiogenic capabilities of the cells, suggesting that IL-13Rα2 promotes genesis of lung TAECs. Together, our data show that CSEs activate the PKA, CREB, and IL-13Rα2 axis in lung ECs, and IL-13Rα2 promotes the malignant transformation of lung ECs and genesis of TAECs with robust angiogenic and oncogenic capabilities. Our study provides new insight into the mechanism of CSEs-triggered lung cancer angiogenesis and tumorigenesis, suggesting that the PKA-CREB-IL-13Rα2 axis is a potential target for novel anti-lung tumor angiogenesis and anti-lung cancer drug discovery.
Collapse
Affiliation(s)
- Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Huaidong Liao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yanyan Pan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ying Kong
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wenming Liu
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ping Yang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zihe Huo
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
26
|
Lazzari C, Bulotta A, Ducceschi M, Viganò MG, Brioschi E, Corti F, Gianni L, Gregorc V. Historical Evolution of Second-Line Therapy in Non-Small Cell Lung Cancer. Front Med (Lausanne) 2017; 4:4. [PMID: 28168189 PMCID: PMC5253463 DOI: 10.3389/fmed.2017.00004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 01/06/2017] [Indexed: 12/26/2022] Open
Abstract
Innovative therapeutic agents have significantly improved outcome with an acceptable safety profile in a substantial proportion of non-small cell lung cancer (NSCLC) patients, who depend on oncogenic molecular alterations for their malignant phenotype. Despite the survival improvement achieved with first-line chemotherapy, about 30% of patients do not obtain a tumor response. Moreover, those patients, initially sensitive to treatment, acquire resistance and develop tumor progression after a median of about 5 months. Approximately 60% of the patients progressing from first-line chemotherapy receive further systemic treatment in the second-line setting. Moreover, new options have emerged in the second-line armamentarium for the treatment of patients with NSCLC, including immune checkpoint inhibitors and antiangiogenic agents. The current review provides an overview on the clinical studies that gained the approval of chemotherapy agents (docetaxel and pemetrexed) and epidermal growth factor receptor gene–tyrosine kinase inhibitors as second-line treatment options for NSCLC patients, not carrying molecular alterations.
Collapse
Affiliation(s)
- Chiara Lazzari
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Alessandra Bulotta
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Monika Ducceschi
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Maria Grazia Viganò
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Elena Brioschi
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Francesca Corti
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Luca Gianni
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| | - Vanesa Gregorc
- Department of Oncology, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute , Milan , Italy
| |
Collapse
|
27
|
Zheng N, Huo Z, Zhang B, Meng M, Cao Z, Wang Z, Zhou Q. Thrombomodulin reduces tumorigenic and metastatic potential of lung cancer cells by up-regulation of E-cadherin and down-regulation of N-cadherin expression. Biochem Biophys Res Commun 2016; 476:252-259. [PMID: 27223053 DOI: 10.1016/j.bbrc.2016.05.105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 05/21/2016] [Indexed: 11/26/2022]
Abstract
Thrombomodulin (TM) is an endothelial cell membrane protein and plays critical roles in anti-thrombosis, anti-inflammation, vascular endothelial protection, and is traditionally regarded as a "vascular protection god". In recent years, although TM has been reported to be down-regulated in a variety of malignant tumors including lung cancer, the role and mechanism of TM in lung cancer are enigmatic. In this study, we found that induction of TM overexpression by cholesterol-reducing drug atorvastatin significantly diminished the tumorigenic capability of the lung cancer cells. Moreover, we demonstrated that TM overexpression caused G0/G1 phase arrest and markedly reduced the colony forming capability of the cells. Furthermore, overexpression of TM inhibited cell migration and invasion. Consistently, depletion of TM promoted cell growth, reduced the cell population at the G0/G1 phase, and enhanced cell migratory ability. Mechanistic study revealed that TM up-regulated E-cadherin but down-regulated N-cadherin expression, resulting in reversal of epithelial-mesenchymal transition (EMT) in the lung cancer cells. Moreover, silencing TM expression led to decreased E-cadherin and increased N-cadherin. Taken together, our study suggests that TM functions as a tumor suppressive protein, providing a conceptual framework for inducing TM overexpression as a sensible strategy and approach for novel anti-lung cancer drug discovery.
Collapse
Affiliation(s)
- Nana Zheng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zihe Huo
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhiwei Wang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|