1
|
Kahk NM, Mohamed FEA, Abdelhakeem MM, Bakr RB. Optimization of pyrazole/1,2,4-triazole as dual EGFR/COX-2 inhibitors: Design, synthesis, anticancer potential, apoptosis induction and cell cycle analysis. Eur J Med Chem 2025; 287:117340. [PMID: 39914141 DOI: 10.1016/j.ejmech.2025.117340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
A novel series of pyrazol-4-yl-1,2,4-triazole-3-thiol derivatives 14a-l was designed, prepared and characterized by many spectroscopic techniques. All the novel compounds were screened for their anti-proliferative activity towards breast cancer cell line (MCF-7), colon cancer cell line (HT-29) and lung cancer cell line (A-549) utilizing celecoxib, erlotinib and osimertinib as standards. Compounds 14b, 14g and 14k were the most active towards HT-29, MCF-7 and A-549 cell lines, sequentially with IC50 = 1.20-2.93 μM compared with celecoxib (IC50 = 16.47-41.45 μM), erlotinib (IC50 = 1.95-33.57 μM) and osimertinib (IC50 = 0.75-3.45 μM). These most active derivatives 14b, 14g and 14k were further investigated for their inhibitory potential against COX and EGFR enzymes. These compounds 14b, 14g and 14k suppressed COX-2 (IC50 = 0.560-4.692 μM), EGFRWT (IC50 = 0.121-0.423 μM) and EGFRT790M (IC50 = 0.076-0.764 μM) enzymes. Compounds 14b, 14g and 14k displayed apoptosis induction by up-regulating Bax and down-regulating Bcl-2 protein levels. Cell cycle analysis recorded that exposure of MFC-7 cells to compound 14g resulted in a significant increase in the percentage of cells at the G2/M to 39.15 % compared to the standard erlotinib (9.87 %). Docking study of the most potent candidates 14b, 14g and 14k within COX-2, EGFRWT and EGFRT790M active regions was conducted to suggest the binding mode of these compounds inside these target enzymes.
Collapse
Affiliation(s)
- Nesma M Kahk
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Fatma E A Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Marwa M Abdelhakeem
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt
| | - Rania B Bakr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Beni-Suef University, Beni- Suef, 62514, Egypt.
| |
Collapse
|
2
|
Ma Y, Chu Y, Xu Z, Xie C, Ma X, Zhang L, Hu J, Zou B, Wu H, Zhou G. Ultrafast and Highly Specific Detection of One-Base Mutated Cell-Free DNA at a Very Low Abundance. Anal Chem 2024; 96:117-126. [PMID: 38114445 DOI: 10.1021/acs.analchem.3c03326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Liquid biopsy as well as genotyping plays important roles in guiding the use of tumor-targeted drugs and monitoring the generation of drug resistance. However, current methods, such as next-generation sequencing (NGS) and pyrosequencing, require long analysis time and complicated steps. To achieve ultrafast and highly specific detection of cell-free DNA (cfDNA) from blood, we improved our recently developed FEN1-aided RPA (FARPA), which combined flap endonuclease 1 (FEN1)-catalyzed invasive reactions with recombinase polymerase amplification (RPA) by inactivating the RPA enzymes before invasive reactions, designing short RPA primers, and changing invasive reaction conditions. Using the L858R and T790M mutations as examples, FARPA was sensitive to detect 5 copies of targeted mutants, specific to sense the mutants with an abundance as low as 0.01% from blood, and ultrafast to get results within 40 min. The method was readily expended to genotyping, and 15 min was enough to report the allele species directly from oral swab samples by coupling quick DNA extraction reagents. Validation was carried out by detecting clinical samples, including 20 cfDNA from patients with non-small cell lung cancer (NSCLC) for liquid biopsy and 43 human genomic DNA (gDNA) purified from blood (33) or lysed from oral swabs (10) for genotyping, giving 100% agreement with NGS and pyrosequencing, respectively. Furthermore, a portable battery-driven device with dual-channel fluorescence detection was successfully constructed to facilitate point-of-care testing (POCT) of liquid biopsy and genotyping, providing doctors with a potential tool to achieve genotyping- or mutant-guided personalized medicine at emergency or source-limited regions.
Collapse
Affiliation(s)
- Yi Ma
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Yanan Chu
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zhaoluo Xu
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Chunmei Xie
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Xueping Ma
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Likun Zhang
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Jingjing Hu
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Bingjie Zou
- Key Laboratory of Drug Quality Control and Pharmacovigilance of Ministry of Education, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Wu
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
- Department of Clinical Pharmacy, Nanjing Jinling Hospital, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Guohua Zhou
- Department of Clinical Pharmacy, State Key Laboratory of Analytical Chemistry for Life Science and Jiangsu Key Laboratory of Molecular Medicine, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| |
Collapse
|
3
|
Cong D, Zhao Y, Zhang W, Li J, Bai Y. Applying machine learning algorithms to develop a survival prediction model for lung adenocarcinoma based on genes related to fatty acid metabolism. Front Pharmacol 2023; 14:1260742. [PMID: 37920207 PMCID: PMC10619909 DOI: 10.3389/fphar.2023.1260742] [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: 07/18/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023] Open
Abstract
Background: The progression of lung adenocarcinoma (LUAD) may be related to abnormal fatty acid metabolism (FAM). The present study investigated the relationship between FAM-related genes and LUAD prognosis. Methods: LUAD samples from The Cancer Genome Atlas were collected. The scores of FAM-associated pathways from the Kyoto Encyclopedia of Genes and Genomes website were calculated using the single sample gene set enrichment analysis. ConsensusClusterPlus and cumulative distribution function were used to classify molecular subtypes for LUAD. Key genes were obtained using limma package, Cox regression analysis, and six machine learning algorithms (GBM, LASSO, XGBoost, SVM, random forest, and decision trees), and a RiskScore model was established. According to the RiskScore model and clinical features, a nomogram was developed and evaluated for its prediction performance using a calibration curve. Differences in immune abnormalities among patients with different subtypes and RiskScores were analyzed by the Estimation of STromal and Immune cells in MAlignant Tumours using Expression data, CIBERSORT, and single sample gene set enrichment analysis. Patients' drug sensitivity was predicted by the pRRophetic package in R language. Results: LUAD samples had lower scores of FAM-related pathways. Three molecular subtypes (C1, C2, and C3) were defined. Analysis on differential prognosis showed that the C1 subtype had the most favorable prognosis, followed by the C2 subtype, and the C3 subtype had the worst prognosis. The C3 subtype had lower immune infiltration. A total of 12 key genes (SLC2A1, PKP2, FAM83A, TCN1, MS4A1, CLIC6, UBE2S, RRM2, CDC45, IGF2BP1, ANGPTL4, and CD109) were screened and used to develop a RiskScore model. Survival chance of patients in the high-RiskScore group was significantly lower. The low-RiskScore group showed higher immune score and higher expression of most immune checkpoint genes. Patients with a high RiskScore were more likely to benefit from the six anticancer drugs we screened in this study. Conclusion: We developed a RiskScore model using FAM-related genes to help predict LUAD prognosis and develop new targeted drugs.
Collapse
Affiliation(s)
- Dan Cong
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yanan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wenlong Zhang
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jun Li
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuansong Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Zheng Y, Zhao Q, Lin J, Dai X, Zhu C, Wang Y, Fu H. Xijiao Dihuang decoction relieves the erlotinib-induced dermatitis. Exp Cell Res 2023; 423:113437. [PMID: 36435221 DOI: 10.1016/j.yexcr.2022.113437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Erlotinib treatment can lead to skin diseases that drastically affected the quality of life of patients. Quercetin (Que), the active component in Xijiao Dihuang Decoction (XDD), was identified to improve inflammatory skin diseases. However, the mechanism of XDD treating erlotinib-induced cutaneous toxicity was not clear at the molecular level. METHODS Keratinocytes were treated with erlotinib, and the expression of inflammatory cytokines and chemokines was revealed by ELISA and qRT-PCR. The macrophage polarization was determined by flow cytometry. The key component of XDD, Que, and the target genes of dermatitis were selected via network pharmacology analysis. The binding effects of Que and target genes were verified using molecular docking and cellular thermal shift assay (CETSA)-western blot assay. Animal experiments were performed in vivo to verify the therapeutic effect of XDD on erlotinib-induced skin toxicity. RESULTS Erlotinib induced M1 polarization of macrophages after stimulating epidermal keratinocytes. While this effect was associated with increased production of inflammatory cytokines and chemokines, such production was prominently decreased by XDD treatment. By combining network pharmacological analysis, molecular docking, and CETSA, it was confirmed that Que had a binding relationship with IL-2 and CXCL8. In vivo results implied that erlotinib abated tumor growth and stimulated dermatitis in HR-1 nude mice, while Que alleviated erlotinib-induced skin damage without affecting this tumor repression effect. CONCLUSION The results indicated that XDD could relieve the dermatitis induced by erlotinib and provide a favorable theoretical basis for the clinical relief by using this method.
Collapse
Affiliation(s)
- Yun Zheng
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China
| | - Qiong Zhao
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China.
| | - Jing Lin
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China
| | - Xiaoyang Dai
- College of Pharmacy, Zhejiang University, Hangzhou, 310030, China
| | - Chenyu Zhu
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China
| | - Yujie Wang
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China
| | - Hongye Fu
- Department of Thoracic Oncology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310022, China
| |
Collapse
|
5
|
Nasretdinov AF, Menshikov KV, Sultanbaev AV, Musin SI, Sultanbaeva NI, Men'shikova IA. The mechanism of action of different generations of EGFR-inhibitors in malignant lung tumors. Literature review and data synthesis. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Knowledge of the molecular characteristics of the progression of non-small cell lung tumors currently provides the patient with the best treatment options. One of the most well-known and studied genetic abnormalities is a mutation in the EGFR gene, which makes the tumor sensitive to therapy with tyrosine kinase inhibitors. In the conditions of the existence of several therapeutic options at once, it is required to know not only the results of clinical trials, but also the fundamental features of the mechanisms of action of a particular drug. The article contains a literature review, presenting the features of the functioning of EGFR (epidermal growth factor receptor), the mechanisms of action of EGFR inhibitors of different generations (erlotinib, gefitinib, afatinib, osimertinib), generalization and analysis of the main differences between them.
Collapse
|
6
|
Rupp T, Debasly S, Genest L, Froget G, Castagné V. Therapeutic Potential of Fingolimod and Dimethyl Fumarate in Non-Small Cell Lung Cancer Preclinical Models. Int J Mol Sci 2022; 23:ijms23158192. [PMID: 35897763 PMCID: PMC9330228 DOI: 10.3390/ijms23158192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 01/27/2023] Open
Abstract
New therapies are required for patients with non-small cell lung cancer (NSCLC) for which the current standards of care poorly affect the patient prognosis of this aggressive cancer subtype. In this preclinical study, we aim to investigate the efficacy of Fingolimod, a described inhibitor of sphingosine-1-phosphate (S1P)/S1P receptors axis, and Dimethyl Fumarate (DMF), a methyl ester of fumaric acid, both already approved as immunomodulators in auto-immune diseases with additional expected anti-cancer effects. The impact of both drugs was analyzed with in vitro cell survival analysis and in vivo graft models using mouse and human NSCLC cells implanted in immunocompetent or immunodeficient mice, respectively. We demonstrated that Fingolimod and DMF repressed tumor progression without apparent adverse effects in vivo in three preclinical mouse NSCLC models. In vitro, Fingolimod did not affect either the tumor proliferation or the cytotoxicity, although DMF reduced tumor cell proliferation. These results suggest that Fingolimod and DMF affected tumor progression through different cellular mechanisms within the tumor microenvironment. Fingolimod and DMF might uncover potential therapeutic opportunities in NSCLC.
Collapse
Affiliation(s)
- Tristan Rupp
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
- Correspondence: or ; Tel.: +33-(0)2-43-69-36-07
| | - Solène Debasly
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
- CNRS UMR 7369 (Matrice Extracellulaire et Dynamique Cellulaire, MEDyC), Université de Reims-Champagne-Ardenne, Campus Moulin de la Housse, 51687 Reims, France
| | - Laurie Genest
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| | - Guillaume Froget
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| | - Vincent Castagné
- Porsolt SAS, ZA de Glatigné, 53940 Le Genest-Saint-Isle, France; (S.D.); (L.G.); (G.F.); (V.C.)
| |
Collapse
|
7
|
Park Y. Personalized Risk-Based Screening Design for Comparative Two-Arm Group Sequential Clinical Trials. J Pers Med 2022; 12:448. [PMID: 35330448 PMCID: PMC8953575 DOI: 10.3390/jpm12030448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Personalized medicine has been emerging to take into account individual variability in genes and environment. In the era of personalized medicine, it is critical to incorporate the patients' characteristics and improve the clinical benefit for patients. The patients' characteristics are incorporated in adaptive randomization to identify patients who are expected to get more benefit from the treatment and optimize the treatment allocation. However, it is challenging to control potential selection bias from using observed efficacy data and the effect of prognostic covariates in adaptive randomization. This paper proposes a personalized risk-based screening design using Bayesian covariate-adjusted response-adaptive randomization that compares the experimental screening method to a standard screening method based on indicators of having a disease. Personalized risk-based allocation probability is built for adaptive randomization, and Bayesian adaptive decision rules are calibrated to preserve error rates. A simulation study shows that the proposed design controls error rates and yields a much smaller number of failures and a larger number of patients allocated to a better intervention compared to existing randomized controlled trial designs. Therefore, the proposed design performs well for randomized controlled clinical trials under personalized medicine.
Collapse
Affiliation(s)
- Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
8
|
Kesri R, Goyal H, Gupta G, Bharti D, Sharma R. Prevalence and Clinicopathologic Risk Factors for Epidermal Growth Factor Receptor, Anaplastic Lymphoma Kinase, and ROS-1 Fusion in Metastatic Non-small Cell Lung Carcinoma. JOURNAL OF RADIATION AND CANCER RESEARCH 2021. [DOI: 10.4103/jrcr.jrcr_43_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
9
|
D'Souza LC, Mishra S, Chakraborty A, Shekher A, Sharma A, Gupta SC. Oxidative Stress and Cancer Development: Are Noncoding RNAs the Missing Links? Antioxid Redox Signal 2020; 33:1209-1229. [PMID: 31891666 DOI: 10.1089/ars.2019.7987] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Significance: It is now clear that genetic changes underlie the basis of cancer, and alterations in functions of multiple genes are responsible for the process of tumorigenesis. Besides the classical genes that are usually implicated in cancer, the role of noncoding RNAs (ncRNAs) and reactive oxygen species (ROS) as independent entitites has also been investigated. Recent Advances: The microRNAs and long noncoding RNAs (lncRNAs), two main classes of ncRNAs, are known to regulate many aspects of tumor development. ROS, generated during oxidative stress and pathological conditions, are known to regulate every step of tumor development. Conversely, oxidative stress and ROS producing agents can suppress tumor development. The malignant cells normally produce high levels of ROS compared with normal cells. The interaction between ROS and ncRNAs regulates the expression of multiple genes and pathways implicated in cancer, suggesting a unique mechanistic relationship among ncRNA-ROS-cancer. The mechanistic relationship has been reported in hepatocellular carcinoma, glioma, and malignancies of blood, breast, colorectum, esophagus, kidney, lung, mouth, ovary, pancreas, prostate, and stomach. The ncRNA-ROS regulate several cancer-related cell signaling pathways, namely, protein kinase B (AKT), epidermal growth factor receptor (EGFR), forkhead box O3 (FOXO3), kelch-like ECH-associated protein 1 (Keap1), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), p53, phosphatase and tensin homologue (PTEN), and wingless-related integration site (Wnt)/glycogen synthase kinase-3 beta (GSK3β). Critical Issues: To date, most of the reports about ncRNA-oxidative stress-carcinogenesis relationships are based on cell lines. The mechanistic basis for this relationship has not been completely elucidated. Future Directions: Attempts should be made to explore the association of lncRNAs with ROS. The significance of the ncRNA-oxidative stress-carcinogenesis interplay should also be explored through studies in animal models.
Collapse
Affiliation(s)
- Leonard Clinton D'Souza
- Division of Environmental Health and Toxicology, Nitte University Centre for Science Education and Research (NUCSER), Mangaluru, India
| | - Shruti Mishra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anirban Chakraborty
- Division of Molecular Genetics and Cancer, Nitte University Centre for Science Education and Research (NUCSER), Mangaluru, India
| | - Anusmita Shekher
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anurag Sharma
- Division of Environmental Health and Toxicology, Nitte University Centre for Science Education and Research (NUCSER), Mangaluru, India
| | - Subash Chandra Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
10
|
Gefitinib sensitization of cisplatin-resistant wild-type EGFR non-small cell lung cancer cells. J Cancer Res Clin Oncol 2020; 146:1737-1749. [PMID: 32342201 PMCID: PMC7185832 DOI: 10.1007/s00432-020-03228-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 04/18/2020] [Indexed: 01/23/2023]
Abstract
Purpose The usual first-line strategy of wild-type EGFR (wtEGFR) non-small cell lung cancer (NSCLC) remains cisplatin-based chemotherapy. However, cisplatin often loses effectiveness because most tumors acquire drug resistance over time. As EGFR is the most important pro-survival/proliferation signal receptor in NSCLC cells, we aimed at investigating whether cisplatin resistance is related to EGFR activation and further evaluating the combined effects of cisplatin/gefitinib (EGFR-tyrosine kinase inhibitor, EGFR-TKI) on cisplatin-resistant wtEGFR NSCLC cells. Materials and methods EGFR activation was analysed in parental and cisplatin-resistant wtEGFR NSCLC cell lines (H358 and H358R, A549 and A549R). Cellular proliferation and apoptosis of H358R/A549R cells treated with cisplatin or gefitinib, alone or in combination were investigated, and the related effector protein was detected by western blot analysis. Anti-tumor effect of two drugs combined was evaluated in animal models of H358R xenografts in vivo. Results EGFR was significantly phosphorylated in cisplatin-resistant wtEGFR NSCLC cells H358R and A549R than their parental cells. In H358R and A549R cells, anti-proliferative ability of gefitinib was further improved, and gefitinib combined with cisplatin enhanced inhibition of cellular survive/proliferation, and promotion of apoptosis in vitro. The combined effects were also associated with the inhibition of EGFR downstream effector proteins. Similarly, in vivo, gefitinib and cisplatin in combination significantly inhibited tumor growth of H358R xenografts. Conclusion
Abnormal activation of EGFR may induce wtEGFR NSCLC cell resistance to cisplatin. The combined effects of cisplatin/gefitinib suggest that gefitinib, as a combination therapy for patients with cisplatin-resistant wtEGFR NSCLC should be considered.
Collapse
|
11
|
Liu J, Yang Z, Kong Y, He Y, Xu Y, Cao X. Antitumor activity of alantolactone in lung cancer cell lines NCI-H1299 and Anip973. J Food Biochem 2019; 43:e12972. [PMID: 31489665 DOI: 10.1111/jfbc.12972] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 05/13/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022]
Abstract
Alantolactone is a sesquiterpene lactone extracted from Inula helenium L. plants possessing many biological activities, including anti-inflammatory, antiproliferation, and antimicrobial. The inhibitory effects and the underlying mechanisms of alantolactone on lung cancer cells NCI-H1299 and Anip973 were investigated in this study. The results showed that alantolactone could decrease cell viability and induce cell apoptosis of NCI-H1299 and Anip973. After the cells were treated with alantolactone, the expression of Bcl-2 decreased, while the expression of Bax increased, the expression of MMP-9, MMP-7, and MMP-2 gradually decreased after alantolactone treatment. Furthermore, results showed that alantolactone could activate p38 MAPK pathway and suppress NF-κB pathway, which are involving in lung cancer development. These results indicated that alantolactone was a potential agent for lung cancer treatment. PRACTICAL APPLICATIONS: Lung cancer is one of the most common contributors of cancer death in the world. Chemoprevention and chemotherapy with natural substances are prospective methods for lung cancer treatment. In recent years, the anti-cancer activity of various sesquiterpene lactones has attracted a great deal of interest. Alantolactone is the major active sesquiterpene lactones isolated from Inula helenium L, which is used as a medicine in ancient Romans due to wide range of pharmacological activities. The results obtained from this study revealed the inhibitory effects of alantolactone on lung cancer cells and might provide some experimental basis for prevention and treatment of lung cancer with alantolactone.
Collapse
Affiliation(s)
- Jianli Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| | - Zhijun Yang
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| | - Yuchi Kong
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| | - Yin He
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| | - Yongliang Xu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| | - Xiangyu Cao
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, P.R. China
| |
Collapse
|
12
|
New phosphate derivatives of betulin as anticancer agents: Synthesis, crystal structure, and molecular docking study. Bioorg Chem 2019; 87:613-628. [PMID: 30947097 DOI: 10.1016/j.bioorg.2019.03.060] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 02/27/2019] [Accepted: 03/19/2019] [Indexed: 12/21/2022]
Abstract
Betulin derivatives exhibit an antiproliferative activity and have been tested for many cancer cell lines. This paper describes a new series of 3-phosphate derivatives of betulin bearing different substituents at C28 position. The synthesized compounds were tested in vitro for their antiproliferative effect against human leukemia (MV-4-11 and CCRF/CEM), lung carcinoma (A549), prostate cancer (DU 145), melanoma (Hs 294T) cell lines, and murine leukemia P388. To explore the possible mechanism of anticancer activity for the most in vitro active compounds (4, 5, 7 and 8) and betulin, molecular docking was performed to the binding sites of potential anticancer targets, described for the various triterpene derivatives, including topoisomerase I and II, epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGFR), transcription factor NF-κB, anti-apoptotic protein Bcl-2 and peroxisome proliferator-activated receptor (PPARγ). According to the results of the docking, the best fit to the binding pocket of PPARγ was shown by compound 4.
Collapse
|
13
|
Le BT, Raguraman P, Kosbar TR, Fletcher S, Wilton SD, Veedu RN. Antisense Oligonucleotides Targeting Angiogenic Factors as Potential Cancer Therapeutics. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 14:142-157. [PMID: 30594893 PMCID: PMC6307321 DOI: 10.1016/j.omtn.2018.11.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
Abstract
Cancer is one of the leading causes of death worldwide, and conventional cancer therapies such as surgery, chemotherapy, and radiotherapy do not address the underlying molecular pathologies, leading to inadequate treatment and tumor recurrence. Angiogenic factors, such as EGF, PDGF, bFGF, TGF-β, TGF-α, VEGF, endoglin, and angiopoietins, play important roles in regulating tumor development and metastasis, and they serve as potential targets for developing cancer therapeutics. Nucleic acid-based therapeutic strategies have received significant attention in the last two decades, and antisense oligonucleotide-mediated intervention is a prominent therapeutic approach for targeted manipulation of gene expression. Clinical benefits of antisense oligonucleotides have been recognized by the U.S. Food and Drug Administration, with full or conditional approval of Vitravene, Kynamro, Exondys51, and Spinraza. Herein we review the scope of antisense oligonucleotides that target angiogenic factors toward tackling solid cancers.
Collapse
Affiliation(s)
- Bao T Le
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Prithi Raguraman
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Tamer R Kosbar
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Susan Fletcher
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Steve D Wilton
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Rakesh N Veedu
- Centre for Comparative Genomics, Murdoch University, Murdoch, WA 6150, Australia; Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia.
| |
Collapse
|
14
|
Receptor Tyrosine Kinase-Targeted Cancer Therapy. Int J Mol Sci 2018; 19:ijms19113491. [PMID: 30404198 PMCID: PMC6274851 DOI: 10.3390/ijms19113491] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022] Open
Abstract
In the past two decades, several molecular targeted inhibitors have been developed and evaluated clinically to improve the survival of patients with cancer. Molecular targeted inhibitors inhibit the activities of pathogenic tyrosine kinases. Particularly, aberrant receptor tyrosine kinase (RTK) activation is a potential therapeutic target. An increased understanding of genetics, cellular biology and structural biology has led to the development of numerous important therapeutics. Pathogenic RTK mutations, deletions, translocations and amplification/over-expressions have been identified and are currently being examined for their roles in cancers. Therapies targeting RTKs are categorized as small-molecule inhibitors and monoclonal antibodies. Studies are underway to explore abnormalities in 20 types of RTK subfamilies in patients with cancer or other diseases. In this review, we describe representative RTKs important for developing cancer therapeutics and predicting or evaluated resistance mechanisms.
Collapse
|
15
|
Swami U, Ma D, Zhang J. Response to Erlotinib in a Patient with Compound EGFR L747S and Exon 19 Deletion. J Thorac Oncol 2018; 13:e129-e130. [PMID: 29935854 DOI: 10.1016/j.jtho.2018.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Umang Swami
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Deqin Ma
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jun Zhang
- Division of Hematology, Oncology and Blood and Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa.
| |
Collapse
|
16
|
Wang Y, Wang M, Wang Q, Geng Z, Sun M. Incidence and risk of infections associated with EGFR-TKIs in advanced non-small-cell lung cancer: a systematic review and meta-analysis of randomized controlled trials. Oncotarget 2018; 8:29406-29415. [PMID: 28107192 PMCID: PMC5438740 DOI: 10.18632/oncotarget.14707] [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: 08/15/2016] [Accepted: 10/27/2016] [Indexed: 12/17/2022] Open
Abstract
Currently, the overall incidence and risk of infections with epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) in non-small-cell lung cancer (NSCLC) patients remained undetermined. We searched Pubmed for related articles published from 1 January 1990 to 31 November 2015. Eligible studies included prospective randomized controlled trials (RCTs) evaluating therapy with or without EGFR-TKIs in patients with NSCLC. Data on infections were extracted. Pooled incidence, Peto odds ratio (Peto OR), and 95% confidence intervals (CIs) were calculated. A total of 17,420 patients from 25 RCTs were included. The use of EGFR-TKIs significantly increased the risk of developing all-grade infections (Peto OR 1.48, 95%CI: 1.12-1.96, p = 0.006) in NSCLC patients, but not for severe (Peto OR 1.26, 95%CI: 0.96-1.67, p = 0.098) and fatal infections (Peto OR 0.81, 95%CI: 0.43-1.53, p = 0.52). Meta-regression indicated the risk of infections tended to increase with the treatment duration of EGFR-TKIs. No publication of bias was detected. In conclusion, the use of EGFR-TKIs significantly increased the risk of developing all-grade infectious events in NSCLC patients, but not for severe and fatal infections. Clinicians should be aware of the risks of infections with the administration of these drugs in these patients.
Collapse
Affiliation(s)
- Yingtian Wang
- Department of Respiratory Medicine, Beijing Airport Hospital, Shunyi district, Beijing, China
| | - Mingzhen Wang
- Department of Respiratory Medicine, Beijing Airport Hospital, Shunyi district, Beijing, China.,Department of Respiratory Medicine, Dongying People's Hospital, Dongying, Shandong, Shandong, China
| | - Qiaoxia Wang
- Department of Respiratory Medicine, Beijing Airport Hospital, Shunyi district, Beijing, China.,Department of Respiratory Medicine, Dongying People's Hospital, Dongying, Shandong, Shandong, China
| | - Zhiying Geng
- Department of Respiratory Medicine, Beijing Airport Hospital, Shunyi district, Beijing, China.,Department of Respiratory Medicine, Dongying People's Hospital, Dongying, Shandong, Shandong, China
| | - Mingxiang Sun
- Department of Respiratory Medicine, Beijing Airport Hospital, Shunyi district, Beijing, China.,Department of Respiratory Medicine, Dongying People's Hospital, Dongying, Shandong, Shandong, China
| |
Collapse
|
17
|
Licochalcone A Inhibits the Proliferation of Human Lung Cancer Cell Lines A549 and H460 by Inducing G2/M Cell Cycle Arrest and ER Stress. Int J Mol Sci 2017; 18:ijms18081761. [PMID: 28805696 PMCID: PMC5578150 DOI: 10.3390/ijms18081761] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 08/06/2017] [Accepted: 08/08/2017] [Indexed: 01/13/2023] Open
Abstract
Licochalcone A (LicA), a flavonoid isolated from the famous Chinese medicinal herb Glycyrrhiza uralensis Fisch, has wide spectrum of pharmacological activities. In this study, the anti-cancer effects and potential mechanisms of LicA in non-small cell lung cancer (NSCLC) cells were studied. LicA decreased cell viability and induced apoptosis in a dose-dependent manner in NSCLC cells. LicA inhibited lung cancer cells growth by blocking cell cycle progression at the G2/M transition and inducing apoptosis. LicA treatment decreased the expression of MDM2, Cyclin B1, Cdc2 and Cdc25C in H460 and A549 cancer cell lines. In addition, LicA induced caspase-3 activation and poly-ADP-ribose polymerase (PARP) cleavage, which displayed features of apoptotic signals. Furthermore, LicA increased the expression of endoplasmic reticulum (ER) stress related proteins, such as p-EIF2α and ATF4. These data provide evidence that LicA has the potential to be used in the treatment of lung cancer.
Collapse
|
18
|
Qin X, Lu J, Wang P, Xu P, Liu M, Wang X. Cytochrome P450 3A selectively affects the pharmacokinetic interaction between erlotinib and docetaxel in rats. Biochem Pharmacol 2017; 143:129-139. [PMID: 28716728 DOI: 10.1016/j.bcp.2017.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/12/2017] [Indexed: 10/19/2022]
Abstract
Erlotinib as a first-line drug is used in non-small cell lung cancer (NSCLC) patients with sensitive EGFR mutations, while resistance to this drug will occur after several years of treatment. Therefore, the microtubule disturber docetaxel is introduced as combined regimen in clinical trials. This report investigated the potentials and mechanisms of drug-drug interaction (DDI) between erlotinib and docetaxel using wild type (WT) and Cyp3a1/2 knockout (KO) rats. The erlotinib O-demethylation and docetaxel hydroxylation reactions in the absence or the presence of another drug were analyzed in vitro via the assay of rat liver microsomes. In whole animal studies, erlotinib and docetaxel were given to WT and KO rats individually or jointly, and the pharmacokinetic profiles of these two drugs were analyzed and compared among different groups. The results showed that docetaxel not only inhibited the CYP3A-mediated biotransformation of erlotinib in vitro, but also significantly increased the maximum concentration and systemic exposure of erlotinib in vivo in WT rats. In contrast, the DDI was significantly attenuated in KO rats. On the other hand, erlotinib did not influence docetaxel either in vitro biotransformation or in vivo pharmacokinetic behaviors. These results exhibited the potentials of erlotinib-docetaxel interaction and indicated that the CYP3A played the perpetrating role of docetaxel on erlotinib in rats. A better understanding of this DDI with CYP3A may help the regulation of the use of these two drugs, avoid potential problems, and adjust dose carefully and early in clinic.
Collapse
Affiliation(s)
- Xuan Qin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jian Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Peili Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Peipei Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xin Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
19
|
Zhou L, Yuan C, Zhang C, Zhang L, Gao Z, Wang C, Liu H, Wu Y, Guo H. Enantioselective Synthesis of Quinazoline-Based Heterocycles through Phosphine-Catalyzed Asymmetric [3+3] Annulation of Morita−Baylis−Hillman Carbonates with Azomethine Imines. Adv Synth Catal 2017. [DOI: 10.1002/adsc.201601434] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Leijie Zhou
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Chunhao Yuan
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Cheng Zhang
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Lei Zhang
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Zhenzhen Gao
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Chang Wang
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Honglei Liu
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Yang Wu
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
| | - Hongchao Guo
- Department of Applied Chemistry; China Agricultural University; 2 West Yuanmingyuan Road Beijing 100193 People's Republic of China
- Key Laboratory of Green Pesticide and Agricultural Bioengineering; Ministry of Education; Guizhou University; Guiyang 550025 People's Republic of China
| |
Collapse
|
20
|
Wu T, Geng J, Guo W, Gao J, Zhu X. Asiatic acid inhibits lung cancer cell growth in vitro and in vivo by destroying mitochondria. Acta Pharm Sin B 2017; 7:65-72. [PMID: 28119810 PMCID: PMC5237705 DOI: 10.1016/j.apsb.2016.04.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/18/2016] [Accepted: 03/09/2016] [Indexed: 12/27/2022] Open
Abstract
Asiatic acid (AA), a pentacyclic triterpene found in Centella asiatica, displays significant anti-proliferative effects on cancer cells in vitro although the underlying mechanism of this effect remains unknown. This study investigated the efficacy and mechanism of action of AA against lung cancer both in vivo and in vitro. Using the MTT assay, AA was found to induce apoptosis in a dose- and time-dependent manner, an effect enhanced by pretreatment with an autophagy inhibitor. It also elevated expression of microtubule-associated protein 1 light chain 3 (LC3) and decreased the expression of p62. Furthermore, exposure to AA resulted in collapse of the mitochondrial membrane potential and generation of reactive oxygen species (ROS), suggesting mitochondria are the target of AA. In the mouse lung cancer xenograft model, oral administration of AA significantly inhibited tumor volume and weight accompanied by significant apoptosis of lung cancer cells. In addition, it led to a significant decrease in the expression of proliferating cell nuclear antigen (PCNA). In summary, the results show that AA significantly reduces lung cancer cell growth both in vitro and in vivo and that the associated apoptosis is mediated through mitochondrial damage.
Collapse
|
21
|
Santag S, Siegel F, Wengner AM, Lange C, Bömer U, Eis K, Pühler F, Lienau P, Bergemann L, Michels M, von Nussbaum F, Mumberg D, Petersen K. BAY 1143269, a novel MNK1 inhibitor, targets oncogenic protein expression and shows potent anti-tumor activity. Cancer Lett 2016; 390:21-29. [PMID: 28043914 DOI: 10.1016/j.canlet.2016.12.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 12/29/2022]
Abstract
The initiation of mRNA translation has received increasing attention as an attractive target for cancer treatment in the recent years. The oncogenic eukaryotic translation initiation factor 4E (eIF4E) is the major substrate of MAP kinase-interacting kinase 1 (MNK1), and it is located at the junction of the cancer-associated PI3K and MAPK pathways. The fact that MNK1 is linked to cell transformation and tumorigenesis renders the kinase a promising target for cancer therapy. We identified a novel small molecule MNK1 inhibitor, BAY 1143269, by high-throughput screening and lead optimization. In kinase assays, BAY 1143269 showed potent and selective inhibition of MNK1. By targeting MNK1 activity, BAY 1143269 strongly regulated downstream factors involved in cell cycle regulation, apoptosis, immune response and epithelial-mesenchymal transition in vitro or in vivo. In addition, BAY 1143269 demonstrated strong efficacy in monotherapy in cell line and patient-derived non-small cell lung cancer xenograft models as well as delayed tumor regrowth in combination treatment with standard of care chemotherapeutics. In summary, the inhibition of MNK1 activity with a highly potent and selective inhibitor BAY 1143269 may provide an innovative approach for anti-cancer therapy.
Collapse
Affiliation(s)
- Susann Santag
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Franziska Siegel
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Antje M Wengner
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Claudia Lange
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Ulf Bömer
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Knut Eis
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Florian Pühler
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Philip Lienau
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Linda Bergemann
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Martin Michels
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Franz von Nussbaum
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Dominik Mumberg
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany
| | - Kirstin Petersen
- Bayer AG, Drug Discovery, Pharmaceuticals, Müllerstr. 178, 13353 Berlin, Germany.
| |
Collapse
|
22
|
Kennedy SP, Hastings JF, Han JZR, Croucher DR. The Under-Appreciated Promiscuity of the Epidermal Growth Factor Receptor Family. Front Cell Dev Biol 2016; 4:88. [PMID: 27597943 PMCID: PMC4992703 DOI: 10.3389/fcell.2016.00088] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 08/08/2016] [Indexed: 12/26/2022] Open
Abstract
Each member of the epidermal growth factor receptor (EGFR) family plays a key role in normal development, homeostasis, and a variety of pathophysiological conditions, most notably in cancer. According to the prevailing dogma, these four receptor tyrosine kinases (RTKs; EGFR, ERBB2, ERBB3, and ERBB4) function exclusively through the formation of homodimers and heterodimers within the EGFR family. These combinatorial receptor interactions are known to generate increased interactome diversity and therefore influence signaling output, subcellular localization and function of the heterodimer. This molecular plasticity is also thought to play a role in the development of resistance toward targeted cancer therapies aimed at these known oncogenes. Interestingly, many studies now challenge this dogma and suggest that the potential for EGFR family receptors to interact with more distantly related RTKs is much greater than currently appreciated. Here we discuss how the promiscuity of these oncogenic receptors may lead to the formation of many unexpected receptor pairings and the significant implications for the efficiency of many targeted cancer therapies.
Collapse
Affiliation(s)
- Sean P Kennedy
- Systems Biology Ireland, University College DublinDublin, Ireland; Kinghorn Cancer Centre, Garvan Institute of Medical ResearchSydney, NSW, Australia
| | - Jordan F Hastings
- Kinghorn Cancer Centre, Garvan Institute of Medical Research Sydney, NSW, Australia
| | - Jeremy Z R Han
- Kinghorn Cancer Centre, Garvan Institute of Medical Research Sydney, NSW, Australia
| | - David R Croucher
- Kinghorn Cancer Centre, Garvan Institute of Medical ResearchSydney, NSW, Australia; School of Medicine, University College DublinDublin, Ireland; St Vincent's Hospital Clinical School, University of New South WalesSydney, NSW, Australia
| |
Collapse
|
23
|
Sorensen SF, Demuth C, Weber B, Sorensen BS, Meldgaard P. Increase in soluble PD-1 is associated with prolonged survival in patients with advanced EGFR-mutated non-small cell lung cancer treated with erlotinib. Lung Cancer 2016; 100:77-84. [PMID: 27597284 DOI: 10.1016/j.lungcan.2016.08.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/23/2016] [Accepted: 08/06/2016] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The central immune co-inhibitory programmed cell death receptor/ligand 1 (PD-1/PD-L1) pathway plays a key role in tumor immune evasion in non-small cell lung cancer (NSCLC). Soluble PD-1 (sPD-1) can be detected in the blood, and preclinical evidence suggests that sPD-1 blocks PD-1/-L1 interaction and improves anti-tumor immunity. The present study compares the concentration of sPD-1 in the serum of advanced NSCLC patients with Epidermal Growth Factor Receptor (EGFR) mutation prior to erlotinib treatment and at the time of progression and correlates these results to patient outcome. MATERIALS AND METHODS Blood samples from 38 patients with EGFR-mutated advanced NSCLC treated with erlotinib were analyzed for sPD-1 by sandwich ELISA. EGFR mutational status was assessed in circulating tumor DNA (ctDNA) and tumor biopsies. RESULTS sPD-1 could be detected in 21% of patients prior to erlotinib treatment, and at disease progression in 37% (p=0.015). An increase in sPD-1 during erlotinib therapy was found in 34%, a decrease in 8% and no change in 58% of patients. An increase in sPD-1 during treatment was associated with prolonged progression-free (adjusted HR 0.32, p=0.013) and overall survival (adjusted HR 0.33, p=0.006), but not associated with the emergence of EGFR T790M mutation in ctDNA at progression or any clinicopathological factors. CONCLUSION Patients with an increase in sPD-1 during erlotinib treatment have a more favorable outcome. Our results emphasize the vast clinical impact of the PD-1/PD-L1 axis, and support the existing preclinical evidence in the bioactive function of sPD-1.
Collapse
Affiliation(s)
| | - Christina Demuth
- Department of Clinical Biochemistry, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Britta Weber
- Department of Oncology, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Noerrebrogade 44, 8000 Aarhus C, Denmark
| |
Collapse
|
24
|
Wu T, Liu W, Guo W, Zhu X. Silymarin suppressed lung cancer growth in mice via inhibiting myeloid-derived suppressor cells. Biomed Pharmacother 2016; 81:460-467. [PMID: 27261626 DOI: 10.1016/j.biopha.2016.04.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/09/2016] [Accepted: 04/11/2016] [Indexed: 12/26/2022] Open
Abstract
In this study, we investigated the antitumor activity of Silymarin in a mouse model of colon cancer xenograft of Lewis lung cancer (LLC) cells. Silymarin significantly suppressed tumor growth and induced apoptosis of cells in tumor tissues at a dose of 25 and 50mg/kg. Silymarin treatment enhanced the infiltration and function of CD8(+) T cells. In the meantime, Silymarin decreased the level of IL-10 while elevated the level of IL-2 and IFN-γ in the serum of tumor-bearing mice. Finally, Silymarin reduced the proportion of myeloid-derived suppressor cells (MDSC) in the tumor tissue and also the mRNA expressions of inducible nitric oxide synthases-2 (iNOS2), arginase-1 (Arg-1) and MMP9, which indicated that the function of MDSC in tumor tissues were suppressed. Altogether, our data here showed that Silymarin inhibited the MDSC and promoted the infiltration and function of CD8(+) T cells thus suppressed the growth of LLC xenografts, which provides evidence for the possible use of Silymarin against lung cancer.
Collapse
Affiliation(s)
- Tiancong Wu
- Department of Radiation Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Jiangsu Province, Nanjing, 210093, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Jiangsu Province, Nanjing, 210093, China.
| | - Xixu Zhu
- Department of Radiation Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu Province, China.
| |
Collapse
|
25
|
Abdel-Rahman O, Ahmed H, ElHalawani H. Risk of elevated transaminases in non-small cell lung cancer (NSCLC) patients treated with erlotinib, gefitinib and afatinib: a meta-analysis. Expert Rev Respir Med 2015; 10:223-34. [DOI: 10.1586/17476348.2016.1115723] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|