1
|
Huang Z, Chen K, Xiao X, Fan Z, Zhang Y, Deng L. DeepHeteroCDA: circRNA-drug sensitivity associations prediction via multi-scale heterogeneous network and graph attention mechanism. Brief Bioinform 2025; 26:bbaf159. [PMID: 40223811 PMCID: PMC11995009 DOI: 10.1093/bib/bbaf159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
Drug sensitivity is essential for identifying effective treatments. Meanwhile, circular RNA (circRNA) has potential in disease research and therapy. Uncovering the associations between circRNAs and cellular drug sensitivity is crucial for understanding drug response and resistance mechanisms. In this study, we proposed DeepHeteroCDA, a novel circRNA-drug sensitivity association prediction method based on multi-scale heterogeneous network and graph attention mechanism. We first constructed a heterogeneous graph based on drug-drug similarity, circRNA-circRNA similarity, and known circRNA-drug sensitivity associations. Then, we embedded the 2D structure of drugs into the circRNA-drug sensitivity heterogeneous graph and use graph convolutional networks (GCN) to extract fine-grained embeddings of drug. Finally, by simultaneously updating graph attention network for processing heterogeneous networks and GCN for processing drug structures, we constructed a multi-scale heterogeneous network and use a fully connected layer to predict the circRNA-drug sensitivity associations. Extensive experimental results highlight the superior of DeepHeteroCDA. The visualization experiment shows that DeepHeteroCDA can effectively extract the association information. The case studies demonstrated the effectiveness of our model in identifying potential circRNA-drug sensitivity associations. The source code and dataset are available at https://github.com/Hhhzj-7/DeepHeteroCDA.
Collapse
Affiliation(s)
- Zhijian Huang
- School of Computer Science and Engineering, Central South University, No. 932, South Lushan Road, Changsha 410083, Hunan, China
| | - Kai Chen
- School of Computer Science and Engineering, Central South University, No. 932, South Lushan Road, Changsha 410083, Hunan, China
| | - Xiaojun Xiao
- School of Software, Xinjiang University, No. 666, Shengli Road, Urumqi 830046, Xinjiang, China
| | - Ziyu Fan
- School of Computer Science and Engineering, Central South University, No. 932, South Lushan Road, Changsha 410083, Hunan, China
| | - Yuanpeng Zhang
- School of Software, Xinjiang University, No. 666, Shengli Road, Urumqi 830046, Xinjiang, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, No. 932, South Lushan Road, Changsha 410083, Hunan, China
| |
Collapse
|
2
|
Duan Y, Guo Z, Zhong W, Chen J, Xu S, Liu J, Xu J. An updated review of small-molecule HPK1 kinase inhibitors (2016-present). Future Med Chem 2024; 16:2431-2450. [PMID: 39582317 PMCID: PMC11622775 DOI: 10.1080/17568919.2024.2420630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a serine-threonine kinase specific to hematopoiesis and a member of the MAP4K family of Ste20-related protein kinases. Targeting HPK1 to ameliorate T cell exhaustion and enhance T cell functions is a promising strategy for clinical immunotherapies. Numerous studies have reported the progress in developing effective HPK1 inhibitors and elucidating their mechanisms of action. However, most inhibitors affect multiple signaling pathways, resulting in unintended side effects that limit their clinical development and application. Herein, we reviewed HPK1-related signaling pathways, clinical candidates and recent advances in small-molecule inhibitors targeting HPK1. Additionally, we present our perspectives on current challenges and potential future research field, hoping to provide inspiration for the development of novel HPK1 inhibitors.
Collapse
Affiliation(s)
- Yiping Duan
- Department of Medicinal Chemistry, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu, 211198, Peoples Republic China
| | - Zhichao Guo
- Department of Medicinal Chemistry, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu, 211198, Peoples Republic China
| | - Wenyi Zhong
- Department of Organic Chemistry, China Pharmaceutical University, School of Science, Nanjing, Jiangsu, 211198, Peoples Republic China
| | - Jichao Chen
- Nanjing University Chinese Medicine, School of Pharmacy, Nanjing, Jiangsu, 210023, Peoples Republic China
| | - Shengtao Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu, 211198, Peoples Republic China
| | - Jie Liu
- Department of Organic Chemistry, China Pharmaceutical University, School of Science, Nanjing, Jiangsu, 211198, Peoples Republic China
| | - Jinyi Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu, 211198, Peoples Republic China
| |
Collapse
|
3
|
Xia H, Dong C, Chen X, Wei Z, Gu L, Zhu X. SGTCDA: Prediction of circRNA-drug sensitivity associations with interpretable graph transformers and effective assessment. BMC Genomics 2024; 25:1113. [PMID: 39567908 PMCID: PMC11577602 DOI: 10.1186/s12864-024-11022-6] [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: 06/07/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
CircRNAs are a type of circular non-coding RNA whose associations with drug sensitivities have been demonstrated in recent studies. Due to the high cost of biomedical experiments for detecting the associations between circRNAs and drug sensitivities, several computational methods have been developed. However, these methods were evaluated mainly based on 5- or tenfold cross-validation, which are often over-optimistic. Furthermore, there are technique issues with these models, such as over-smoothing and over-squashing. To address these issues, we propose a strategy to evaluate models based on independent test sets for association prediction-related studies. In the light of this effective assessment, we constructed a model, SGTCDA, by integrating structural deep network embedding (SDNE) and a graph transformer to predict the potential associations of circRNA-drug sensitivity, which can efficiently capture long-range dependencies and local structural information of nodes. Our results on the training sets and the independent test sets indicate that SGTCDA outperforms the other state-of-the-art models, demonstrating its capacity for accurate prediction of circRNA-drug sensitivity. Moreover, we leveraged EdgeSHAPer to explain the performance of the proposed SGTCDA model, which illustrates that the edges between drugs are more important than other edges for the performance of the model. The source code and dataset of SGTCDA are available at: https://github.com/hwxia/SGTCDA .
Collapse
Affiliation(s)
- Hongwei Xia
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Province Key Laboratory of Smart Agricultural Technology and Equipment, Hefei, Anhui, 230036, China
- Research Center for Agricultural Information Perception and Intelligent Computing Engineering of Anhui Province, Hefei, Anhui, 230036, China
| | - Caiyue Dong
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Province Key Laboratory of Smart Agricultural Technology and Equipment, Hefei, Anhui, 230036, China
- Research Center for Agricultural Information Perception and Intelligent Computing Engineering of Anhui Province, Hefei, Anhui, 230036, China
| | - Xinxing Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Zhuoyu Wei
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Province Key Laboratory of Smart Agricultural Technology and Equipment, Hefei, Anhui, 230036, China
- Research Center for Agricultural Information Perception and Intelligent Computing Engineering of Anhui Province, Hefei, Anhui, 230036, China
| | - Lichuan Gu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China.
- Anhui Province Key Laboratory of Smart Agricultural Technology and Equipment, Hefei, Anhui, 230036, China.
- Research Center for Agricultural Information Perception and Intelligent Computing Engineering of Anhui Province, Hefei, Anhui, 230036, China.
| | - Xiaolei Zhu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China.
- Anhui Province Key Laboratory of Smart Agricultural Technology and Equipment, Hefei, Anhui, 230036, China.
- Research Center for Agricultural Information Perception and Intelligent Computing Engineering of Anhui Province, Hefei, Anhui, 230036, China.
| |
Collapse
|
4
|
Al-Sanea MM, Hafez HM, Mohamed AAB, El-Shafey HW, Elgazar AA, Tawfik SS, Ewes WA, Hussein S, Alsahli TG, Hamdi A. Design, Synthesis, Pharmacological Evaluation of Quinazolin-4(3 H)-Ones Bearing Urea Functionality as Potential VEGFR-2 Inhibitors. Drug Des Devel Ther 2024; 18:5109-5127. [PMID: 39554760 PMCID: PMC11568772 DOI: 10.2147/dddt.s490930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Background In response to the urgent need for continuous discovery of new anti-proliferative agents, a new series of quinazoline compounds 5a-r was prepared. Methods As a reference, four cancer cell lines-HCT116, HePG2, Hela, and MCF-7-and sorafenib (SOR) were used to assess the novel motifs' in vitro anticancer efficacy. The most cytotoxic compounds were tested in a VEGFR-2 suppressive test and flow cytometric test. Docking analysis was done to the three novel motifs. Results Compound 5d showed the best anti-tumor activity of the tested compounds with IC50 6.09, 2.39, 8.94 and 4.81 μM in succession. In addition, compound 5h revealed a potent anticancer effect against HCT116 and HePG2 with IC50 5.89 and 6.74 μM, respectively. Also, compound 5p exhibited very strong activity against HCT116, HePG2 & MCF7 with IC50 8.32, 9.72 and 7.99, respectively. Compound 5p had the highest inhibition against VEGFR-2 with an IC50 of 0.117 μM, in contrast to 0.069 μM for SOR. According to flow cytometric testing, the most effective VEGFR-2 inhibitory agent, 5p, was shown to suppress the G1/S cell population in MCF-7 cells. Docking analysis confirmed that the three novel motifs could bind to the VEGFR-2 enzyme's binding region like the co-crystallized ligand SOR did. Conclusion The enzyme inhibitory test of compound 5p showed that it is the most potent hybrid that caused MCF-7 cells to undergo apoptosis and generated a G1/S cell cycle arrest. Confirmation of the obtained results was done with the aid of the docking study, which showed that the three motifs might adhere to the enzyme's major active sites, and the results were in good accordance with the experimental VEGFR-2 inhibitory results. We can conclude that the new quinazoline compounds 5a-r could be used as candidates for development of more efficient anticancer inhibitors.
Collapse
Affiliation(s)
- Mohammad M Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia
| | - Hani M Hafez
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Esraa University, Baghdad, Iraq
| | - Ahmed A B Mohamed
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Hamed W El-Shafey
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Abdullah A Elgazar
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafr El Sheikh, Egypt
| | - Samar S Tawfik
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Shaimaa Hussein
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia
| | - Tariq G Alsahli
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia
| | - Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
5
|
Abed H, Radha R, Anjum S, Paul V, AlSawaftah N, Pitt WG, Ashammakhi N, Husseini GA. Targeted Cancer Therapy-on-A-Chip. Adv Healthc Mater 2024; 13:e2400833. [PMID: 39101627 PMCID: PMC11582519 DOI: 10.1002/adhm.202400833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/15/2024] [Indexed: 08/06/2024]
Abstract
Targeted cancer therapy (TCT) is gaining increased interest because it reduces the risks of adverse side effects by specifically treating tumor cells. TCT testing has traditionally been performed using two-dimensional (2D) cell culture and animal studies. Organ-on-a-chip (OoC) platforms have been developed to recapitulate cancer in vitro, as cancer-on-a-chip (CoC), and used for chemotherapeutics development and testing. This review explores the use of CoCs to both develop and test TCTs, with a focus on three main aspects, the use of CoCs to identify target biomarkers for TCT development, the use of CoCs to test free, un-encapsulated TCTs, and the use of CoCs to test encapsulated TCTs. Despite current challenges such as system scaling, and testing externally triggered TCTs, TCToC shows a promising future to serve as a supportive, pre-clinical platform to expedite TCT development and bench-to-bedside translation.
Collapse
Affiliation(s)
- Heba Abed
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Remya Radha
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Shabana Anjum
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
| | - Vinod Paul
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| | - William G. Pitt
- Department of Chemical EngineeringBrigham Young UniversityProvoUT84602USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME)Michigan State UniversityEast LansingMI48824USA
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095‐1600USA
| | - Ghaleb A. Husseini
- Department of Chemical and Biological EngineeringAmerican University of SharjahSharjahUAE
- Materials Science and Engineering PhD programCollege of Arts and SciencesAmerican University of SharjahSharjahUAE
| |
Collapse
|
6
|
Zheng S, Su Z, He Y, You L, Zhang G, Chen J, Lu L, Liu Z. Novel prognostic signature for hepatocellular carcinoma using a comprehensive machine learning framework to predict prognosis and guide treatment. Front Immunol 2024; 15:1454977. [PMID: 39380994 PMCID: PMC11458406 DOI: 10.3389/fimmu.2024.1454977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is highly aggressive, with delayed diagnosis, poor prognosis, and a lack of comprehensive and accurate prognostic models to assist clinicians. This study aimed to construct an HCC prognosis-related gene signature (HPRGS) and explore its clinical application value. Methods TCGA-LIHC cohort was used for training, and the LIRI-JP cohort and HCC cDNA microarray were used for validation. Machine learning algorithms constructed a prognostic gene label for HCC. Kaplan-Meier (K-M), ROC curve, multiple analyses, algorithms, and online databases were used to analyze differences between high- and low-risk populations. A nomogram was constructed to facilitate clinical application. Results We identified 119 differential genes based on transcriptome sequencing data from five independent HCC cohorts, and 53 of these genes were associated with overall survival (OS). Using 101 machine learning algorithms, the 10 most prognostic genes were selected. We constructed an HCC HPRGS with four genes (SOCS2, LCAT, ECT2, and TMEM106C). Good predictive performance of the HPRGS was confirmed by ROC, C-index, and K-M curves. Mutation analysis showed significant differences between the low- and high-risk patients. The low-risk group had a higher response to transcatheter arterial chemoembolization (TACE) and immunotherapy. Treatment response of high- and low-risk groups to small-molecule drugs was predicted. Linifanib was a potential drug for high-risk populations. Multivariate analysis confirmed that HPRGS were independent prognostic factors in TCGA-LIHC. A nomogram provided a clinical practice reference. Conclusion We constructed an HPRGS for HCC, which can accurately predict OS and guide the treatment decisions for patients with HCC.
Collapse
Affiliation(s)
- Shengzhou Zheng
- Department of Emergency, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Zhixiong Su
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yufang He
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Lijie You
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Guifeng Zhang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Jingbo Chen
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Lihu Lu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhenhua Liu
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Wang G, Shi C, He L, Li Y, Song W, Chen Z, Liu Z, Wang Y, He X, Yu Y, Tian Y, Wang X. Identification of the tumor metastasis-related tumor subgroups overexpressed NENF in triple-negative breast cancer by single-cell transcriptomics. Cancer Cell Int 2024; 24:319. [PMID: 39294690 PMCID: PMC11409682 DOI: 10.1186/s12935-024-03505-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024] Open
Abstract
Tumor metastasis is a continuous and dynamic process and is a major cause of tumor-related death in triple-negative breast cancer. However, this biological process remains largely unknown in triple-negative breast cancer. The emergence of single-cell sequencing enables a deeper understanding of the tumor microenvironment and provides a new strategy for discovering the potential mechanism of tumor metastasis. Herein, we integrated the single-cell expression profiling of primary and metastatic triple-negative breast cancer by Seurat package. Nine tumor cell subgroups were identified. Enrichment analysis suggested tumor subgroups (C0, C4) were associated with tumor metastasis with poor prognosis in TNBC. Weighted gene co-expression network was constructed and identified NENF was a metastasis-related gene. Subsequently, RT-qPCR, Immunohistochemistry, and western blot confirmed NENF is highly expressed in TNBC tissues. And cell function assays indicated NENF promote cell invasion and migration through regulating EMT in TNBC. Finally, TIDE and Connectivity Map database suggest the candidate drugs for targeting NENF. In conclusion, our findings provide a new insight into the progression and metastasis of TNBC and uncover NENF may be a prognostic biomarker and potential therapy targets.
Collapse
Affiliation(s)
- Guixin Wang
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Cangchang Shi
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Long He
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Yingxi Li
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Wenbin Song
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Zhaohui Chen
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Zhaoyi Liu
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Yizeng Wang
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Xianghui He
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China
| | - Yue Yu
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China
| | - Yao Tian
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China.
- Department of General Surgery, Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin Medical University General Hospital, Tianjin General Surgery Institute, 154 An-Shan Road, He-Ping District, Tianjin, 300052, P. R. China.
| | - Xin Wang
- the First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huan-Hu-Xi Road, He-Xi District, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University, Tianjin, 300060, China.
| |
Collapse
|
8
|
Matus MF, Häkkinen H. Rational Design of Targeted Gold Nanoclusters with High Affinity to Integrin αvβ3 for Combination Cancer Therapy. Bioconjug Chem 2024. [PMID: 39008847 DOI: 10.1021/acs.bioconjchem.4c00248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
The unique attributes of targeted nano-drug delivery systems (TNDDSs) over conventional cancer therapies in suppressing off-target effects make them one of the most promising options for cancer treatment. There is evidence that the density of surface-conjugated ligands is a crucial factor in achieving the desired therapeutic efficacy of TNDDSs, but this is hardly manageable in conventional nanomaterials. In this context, ligand-protected gold nanoclusters (AuNCs) are excellent candidates for developing new TNDDSs with a unique control on their surface functionalities, thus helping to achieve enhanced delivery performance. Here, we study the interactions and binding free energies between ten different functionalized Au144(SR)60 (SR = thiolate ligand) nanoclusters and integrin αvβ3 using molecular dynamics simulations and the umbrella sampling method to obtain the optimal formulations. The AuNCs were functionalized with anticancer drugs (5-fluorouracil or signaling pathways inhibitors, such as capivasertib, linifanib, tanespimycin, and taselisib) and integrin-targeting peptides (RGD4C or QS13), and we identified the optimal mixed ligand layer to enhance their binding affinity to the cancer cell receptor. The results showed that changing the proportions of the same type of ligands on the surface of AuNCs led to differences of up to 38 kcal/mol in computed binding free energies. RGD4C as the targeting peptide resulted in greater affinity for αvβ3, and in most formulations studied, a higher amount of drug than peptide was needed. Polar and charged residues, such as Ser123, Asp150, Tyr178, Arg214, and Asp251 were found to play a significant role in AuNC binding. Our simulations also revealed that Mn2+ cations are crucial for stabilizing the αvβ3-AuNC complex. These findings demonstrate the potential of carefully designing the surface composition of TNDDSs to optimize their target affinity and specificity.
Collapse
Affiliation(s)
| | - Hannu Häkkinen
- Department of Physics, University of Jyväskylä, FI-40014 Jyväskylä, Finland
- Department of Chemistry, Nanoscience Center, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| |
Collapse
|
9
|
Sarker B, Matiur Rahaman M, Alamin MH, Ariful Islam M, Nurul Haque Mollah M. Boosting edgeR (Robust) by dealing with missing observations and gene-specific outliers in RNA-Seq profiles and its application to explore biomarker genes for diagnosis and therapies of ovarian cancer. Genomics 2024; 116:110834. [PMID: 38527595 DOI: 10.1016/j.ygeno.2024.110834] [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: 08/21/2023] [Revised: 02/09/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
The edgeR (Robust) is a popular approach for identifying differentially expressed genes (DEGs) from RNA-Seq profiles. However, it shows weak performance against gene-specific outliers and is unable to handle missing observations. To address these issues, we proposed a pre-processing approach of RNA-Seq count data by combining the iLOO-based outlier detection and random forest-based missing imputation approach for boosting the performance of edgeR (Robust). Both simulation and real RNA-Seq count data analysis results showed that the proposed edgeR (Robust) outperformed than the conventional edgeR (Robust). To investigate the effectiveness of identified DEGs for diagnosis, and therapies of ovarian cancer (OC), we selected top-ranked 12 DEGs (IL6, XCL1, CXCL8, C1QC, C1QB, SNAI2, TYROBP, COL1A2, SNAP25, NTS, CXCL2, and AGT) and suggested hub-DEGs guided top-ranked 10 candidate drug-molecules for the treatment against OC. Hence, our proposed procedure might be an effective computational tool for exploring potential DEGs from RNA-Seq profiles for diagnosis and therapies of any disease.
Collapse
Affiliation(s)
- Bandhan Sarker
- Department of Statistics, Faculty of Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md Matiur Rahaman
- Department of Statistics, Faculty of Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Haining 314400, China.
| | - Muhammad Habibulla Alamin
- Department of Statistics, Faculty of Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Md Ariful Islam
- Bioinformatics Laboratory (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Laboratory (Dry), Department of Statistics, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
10
|
Aydin B, Beklen H, Arga KY, Bayrakli F, Turanli B. Epigenomic and transcriptomic landscaping unraveled candidate repositioned therapeutics for non-functioning pituitary neuroendocrine tumors. J Endocrinol Invest 2023; 46:727-747. [PMID: 36306107 DOI: 10.1007/s40618-022-01923-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 09/12/2022] [Indexed: 10/31/2022]
Abstract
PURPOSE Non-functioning pituitary neuroendocrine tumors are challengingly diagnosed tumors in the clinic. Transsphenoidal surgery remains the first-line treatment. Despite the development of state-of-the-art techniques, no drug therapy is currently approved for the treatment. There are also no randomized controlled trials comparing therapeutic strategies or drug therapy for the management after surgery. Therefore, novel therapeutic interventions for the therapeutically challenging NF-PitNETs are urgently needed. METHODS We integrated epigenome and transcriptome data (both coding and non-coding) that elucidate disease-specific signatures, in addition to biological and pharmacological data, to utilize rational pathway and drug prioritization in NF-PitNETs. We constructed an epigenome- and transcriptome-based PPI network and proposed hub genes. The signature-based drug repositioning based on the integration of multi-omics data was performed. RESULTS The construction of a disease-specific network based on three different biological levels revealed DCC, DLG5, ETS2, FOXO1, HBP1, HMGA2, PCGF3, PSME4, RBPMS, RREB1, SMAD1, SOCS1, SOX2, YAP1, ZFHX3 as hub proteins. Signature-based drug repositioning using hub proteins yielded repositioned drug candidates that were confirmed in silico via molecular docking. As a result of molecular docking simulations, palbociclib, linifanib, trametinib, eplerenone, niguldipine, and zuclopenthixol showed higher binding affinities with hub genes compared to their inhibitors and were proposed as potential repositioned therapeutics for the management of NF-PitNETs. CONCLUSION The proposed systems' biomedicine-oriented multi-omics data integration for drug repurposing to provide promising results for the construction of effective clinical therapeutics. To the best of our knowledge, this is the first study reporting epigenome- and transcriptome-based drug repositioning for NF-PitNETs using in silico confirmations.
Collapse
Affiliation(s)
- B Aydin
- Department of Bioengineering, Faculty of Engineering and Architecture, Konya Food and Agriculture University, Konya, Turkey
| | - H Beklen
- Department of Bioengineering, Faculty of Engineering, Marmara University, RTE Basibuyuk Campus, 34720, Istanbul, Turkey
| | - K Y Arga
- Department of Bioengineering, Faculty of Engineering, Marmara University, RTE Basibuyuk Campus, 34720, Istanbul, Turkey
- Genetic and Metabolic Diseases Research and Investigation Center (GEMHAM), Marmara University, Istanbul, Turkey
| | - F Bayrakli
- Department of Neurosurgery, Faculty of Medicine, Marmara University, Istanbul, Turkey
- Institute of Neurological Sciences, Marmara University, Istanbul, Turkey
| | - B Turanli
- Department of Bioengineering, Faculty of Engineering, Marmara University, RTE Basibuyuk Campus, 34720, Istanbul, Turkey.
| |
Collapse
|
11
|
Recent updates on thienopyrimidine derivatives as anticancer agents. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03040-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
AbstractThienopyrimidine derivatives hold a unique place between fused pyrimidine compounds. They are important and widely represented in medicinal chemistry as they are structural analogs of purines. Thienopyrimidine derivatives have various biological activities. The current review discusses different synthetic methods for the preparation of heterocyclic thienopyrimidine derivatives. It also highlights the most recent research on the anticancer effects of thienopyrimidines through the inhibition of various enzymes and pathways, which was published within the last 9 years.
Graphical Abstract
Collapse
|
12
|
Lee CM, Lee J, Kang MA, Kim HT, Lee J, Park K, Yang YH, Jang KY, Park SH. Linifanib induces apoptosis in human ovarian cancer cells via activation of FOXO3 and reactive oxygen species. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
13
|
Alamshany ZM, Tashkandi NY, Othman IMM, Anwar MM, Nossier ES. New thiophene, thienopyridine and thiazoline-based derivatives: Design, synthesis and biological evaluation as antiproliferative agents and multitargeting kinase inhibitors. Bioorg Chem 2022; 127:105964. [PMID: 35759881 DOI: 10.1016/j.bioorg.2022.105964] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/05/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022]
Abstract
Multitargeting kinase inhibitors recently proved to be a profitable approach for conquering cancer proliferation. The current study represents the design and synthesis of new thiophene, thienopyridine, and thiazoline-based derivatives 4-14a,b. All the target compounds were examined in vitro against three cancer cell lines; the liver (HepG-2), breast (MCF-7), and colon (HCT-116) where the thiophene-based compounds 5a-c, demonstrated the most potent activity. Furthermore, the latter derivatives revealed a safety profile against WI-38 normal cell line of selectivity indices ranging from 4.43 to 17.44. In vitro enzyme assay of 5a-c revealed that the carbohydrazide analog 5c has the most promising multitargeting inhibiting activity against Pim-1, VEGFR-2, and EGFRWT enzymes of IC50 values; 0.037 ± 0.02, 0.95 ± 0.24, and 0.16 ± 0.05 µM, respectively. As it was the most potent analog, 5c was further subjected to cell cycle and apoptosis analysis. The results indicated that it induced preG1 arrest and an apoptotic effect in the early and late stages. Moreover, further apoptosis studies were carried out for 5c to evaluate its proapoptotic potential. Interestingly, 5c enhanced the levels of Bax/Bcl-2 ratio, p53, and active caspase 3 by 18, 6.4, and 24 folds, respectively compared to the untreated cells. The antimicrobial evaluation showed that only compounds 3 and 5a produced broad-spectrum potency, while 5b and 5c exhibited outstanding antifungal effects. Finally, a molecular docking study was carried out to discover the probable interactions of compound 5c with the active sites of Pim-1, VEGFR-2, and EGFRWT kinases.
Collapse
Affiliation(s)
- Zahra M Alamshany
- Department of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah 21551, P.O. Box 42805, Saudi Arabia
| | - Nada Y Tashkandi
- Department of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah 21551, P.O. Box 42805, Saudi Arabia
| | - Ismail M M Othman
- Department of Chemistry, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Manal M Anwar
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Cairo 12622, Egypt.
| | - Eman S Nossier
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| |
Collapse
|
14
|
Zhu WF, Krämer A, Knapp S, Proschak E, Hernandez-Olmos V. Cascade Synthesis of Kinase-Privileged 3-Aminoindazoles via Intramolecular N-N Bond Formation. J Org Chem 2022; 87:3856-3862. [PMID: 35179025 DOI: 10.1021/acs.joc.1c03057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
3-Aminoindazoles are privileged scaffolds for bioactive drug-like molecules. In this study, a microwave-assisted cascade reaction for the synthesis of N-1 substituted 3-aminoindazoles with yields up to 81% has been developed. Starting from 3-(2-bromoaryl)-1,2,4-oxadiazol-5(4H)-ones, the reaction exhibits a broad substrate scope including anilines, aliphatic amines, and sulfonamides and bypasses selectivity issues between N-1 and 3-amino group. Furthermore, the Differential Scanning Fluorimetry screen of a kinase panel demonstrated the value of targeting N-1 substituted 3-aminoindazoles as kinase-biased fragments.
Collapse
Affiliation(s)
- W Felix Zhu
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Andreas Krämer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.,Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences (BMLS), Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Victor Hernandez-Olmos
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
15
|
Matus MF, Malola S, Häkkinen H. Ligand Ratio Plays a Critical Role in the Design of Optimal Multifunctional Gold Nanoclusters for Targeted Gastric Cancer Therapy. ACS NANOSCIENCE AU 2021; 1:47-60. [PMID: 37102116 PMCID: PMC10125177 DOI: 10.1021/acsnanoscienceau.1c00008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nanodrug delivery systems (NDDSs) based on water-soluble and atomically precise gold nanoclusters (AuNCs) are under the spotlight due to their great potential in cancer theranostics. Gastric cancer (GC) is one of the most aggressive cancers with a low early diagnosis rate, with drug therapy being the primary means to overcome its increasing incidence. In this work, we designed and characterized a set of 28 targeted nanosystems based on Au144(p-MBA)60 (p-MBA = para-mercaptobenzoic acid) nanocluster to be potentially employed as combination therapy in GC treatment. The proposed multifunctional AuNCs are functionalized with cytotoxic drugs (5-fluorouracil and epirubicin) or inhibitors of different signaling pathways (phosphatidylinositol 3-kinases (PI3K)/protein kinase B (Akt)/mammalian target of the rapamycin (mTOR), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor (HIF)) and RGD peptides as targeting ligands, and we studied the role of ligand ratio in their optimal structural conformation using peptide-protein docking and all-atom molecular dynamics (MD) simulations. The results reveal that the peptide/drug ratio is a crucial factor influencing the potential targeting ability of the nanosystem. The most convenient features were observed when the peptide amount was favored over the drug in most cases; however, we demonstrated that the system composition and the intermolecular interactions on the ligand shell are crucial for achieving the desired effect. This approach helps guide the experimental stage, providing essential information on the size and composition of the nanosystem at the atomic level for ligand tuning in order to increase the desired properties.
Collapse
|
16
|
El-Metwally SA, Abou-El-Regal MM, Eissa IH, Mehany ABM, Mahdy HA, Elkady H, Elwan A, Elkaeed EB. Discovery of thieno[2,3-d]pyrimidine-based derivatives as potent VEGFR-2 kinase inhibitors and anti-cancer agents. Bioorg Chem 2021; 112:104947. [PMID: 33964580 DOI: 10.1016/j.bioorg.2021.104947] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/05/2021] [Accepted: 04/22/2021] [Indexed: 11/26/2022]
Abstract
Vascular endothelial growth factor-2 (VEGFR-2) is considered one of the most important factors in tumor angiogenesis, and consequently a number of anticancer therapeutics have been developed to inhibit VEGFR-2 signaling. Accordingly, eighteen derivatives of thieno[2,3-d]pyrimidines having structural characteristics similar to VEGFR-2 inhibitors were designed and synthesized. Anticancer activities of the new derivatives were assessed against three human cancer cell lines (HCT-116, HepG2, and MCF-7) using MTT. Sorafenib was used as positive control. Compounds 17c-i, and 20b showed excellent anticancer activities against HCT-116 and HepG2 cell lines, while compounds 17i and 17g was found to be active against MCF-7 cell line. Compound 17f exhibited the highest cytotoxic activities against the examined cell lines, HCT-116 and HepG2, with IC50 values of 2.80 ± 0.16 and 4.10 ± 0.45 µM, respectively. Aiming at exploring the mechanism of action of these compounds, the most active cytotoxic derivatives were in vitro tested for their VEGFR-2 inhibitory activity. Compound 17f showed high activity against VEGFR-2 with an IC50 value of 0.23 ± 0.03 µM, that is equal to that of reference, sorafenib (IC50 = 0.23 ± 0.04 µM). Molecular docking studies also were performed to investigate the possible binding interactions of the target compounds with the active sites of VEGFR-2. The synthesized compounds were analyzed for their ADMET and toxicity properties. Results showed that most of the compounds have low to very low BBB penetration levels and they have non-inhibitory effect against CYP2D6. All compounds were predicted to be non-toxic against developmental toxicity potential model except compounds 17b and 20b.
Collapse
Affiliation(s)
- Souad A El-Metwally
- Department of Basic Science, Higher Technological Institute, 10th of Ramadan City 228, Egypt
| | - Mohsen M Abou-El-Regal
- Department of Chemistry, Faculty of Science, Ain Shams University, Abassia, Cairo 11566, Egypt
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Ahmed B M Mehany
- Department of Zoology, Faculty of Science, Al-Azhar University, Cairo 11884, Egypt
| | - Hazem A Mahdy
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Hazem Elkady
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Alaa Elwan
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Riyadh, Saudi Arabia; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| |
Collapse
|
17
|
Abstract
Supplemental Digital Content is available in the text. Derivatives of bis-aryl urea have been widely investigated for their various biological activities, such as antiviral, anti-inflammatory and antiproliferative. We evaluated a new chemical entity consisting of bis-aryl urea moiety, N69B, for its anticancer activities and explored their underlying molecular mechanism. The compound inhibited proliferation of multiple types of murine and human cancer cells in vitro, and reduced tumor growth in mouse 4T1 breast tumor model in vivo. Protein microarray analysis revealed and western blot confirmed that the compound significantly increased protein levels of cathepsins, especially cathepsin D, a lysosomal aspartyl protease known to have various pathophysiological functions. Further studies showed that the compound induced tumor cell apoptosis through the Bid/Bax/Cytochrome C/caspase 9/caspase 3 pathway, in which cathepsin D appeared to be a main mediator. Unlike kinase inhibition commonly seen with many other anticancer bis-aryl urea derivatives, this unique mechanism of N69B may suggest potential of the compound as a novel anticancer drug.
Collapse
|
18
|
Shamanth S, Nagarakere SC, Sagar KS, Narayana Y, Mamatha M, Rangappa KS, Kempegowda M. T3P mediated intramolecular rearrangement of o-aminobenzamide to o-ureidobenzonitrile using isothiocyanates. SYNTHETIC COMMUN 2021. [DOI: 10.1080/00397911.2021.1873384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
| | - Sandhya C. Nagarakere
- PG Department of Studies in Chemistry and Research Centre, St. Philomena’s College, Mysuru, India
| | - Kunigal S. Sagar
- DOS in Chemistry, Manasagangotri, University of Mysore, Mysuru, India
| | - Yatheesh Narayana
- DOS in Chemistry, Manasagangotri, University of Mysore, Mysuru, India
| | | | | | | |
Collapse
|
19
|
Abstract
The diarylurea is a scaffold of great importance in medicinal chemistry as it is present in numerous heterocyclic compounds with antithrombotic, antimalarial, antibacterial, and anti-inflammatory properties. Some diarylureas, serine-threonine kinase or tyrosine kinase inhibitors, were recently reported in literature. The first to come into the market as an anticancer agent was sorafenib, followed by some others. In this review, we survey progress over the past 10 years in the development of new diarylureas as anticancer agents.
Collapse
|
20
|
Elseginy SA, Hamdy R, Menon V, Almehdi AM, El-Awady R, Soliman SSM. Design, synthesis, and computational validation of novel compounds selectively targeting HER2-expressing breast cancer. Bioorg Med Chem Lett 2020; 30:127658. [PMID: 33130288 DOI: 10.1016/j.bmcl.2020.127658] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022]
Abstract
Human epidermal growth factor receptor (HER) is a family of multidomain proteins that plays important role in the regulation of several biological functions. HER2 is a member of HER that is highly presented in breast cancer cells. Here, we designed and synthesized a series of diaryl urea/thiourea compounds. The compounds were tested on HER2+ breast cancer cells including MCF-7 and SkBr3, compared to HER2- breast cancer cells including MDA-MB-231 and BT-549. Only compounds 12-14 at 10 µM showed selective anti-proliferative activity against MCF-7 and SkBr3 by 65-79%. Compounds 12-14 showed >80% inhibition of the intracellular kinase domain of HER2. The results obtained indicated that compounds 12-14 are selectively targeting HER2+ cells. The IC50 of compound 13 against MCF-7 and SkBR3 were 1.3 ± 0.009 and 0.73 ± 0.03 µM, respectively. Molecular docking and MD simulations (50 ns) were carried out, and their binding free energies were calculated. Compounds 12-14 formed strong hydrogen bond and pi-pi stacking interactions with the key residues Thr862 and Phe864. 3DQSAR model confirmed the role of 3-bromo substituent of pyridine ring and 4-chloro substituent of phenyl ring in the activity of the compounds. In conclusion, novel compounds, particularly 13 were developed selectively against HER2-expressing/overexpressing breast cancer cells including MCF7 and SkBr3.
Collapse
Affiliation(s)
- Samia A Elseginy
- Green Chemistry Department, Chemical Industries Research Division, National Research Center P.O. Box 12622, Egypt; Molecular Modelling Lab., Biochemistry School, Bristol University, Bristol, UK
| | - Rania Hamdy
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Varsha Menon
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Ahmed M Almehdi
- College of Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates; College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
21
|
Design, synthesis, molecular docking and cytotoxic activity of novel urea derivatives of 2-amino-3-carbomethoxythiophene. J CHEM SCI 2020. [DOI: 10.1007/s12039-020-01834-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
22
|
Theoretical Investigations on Interactions of Arylsulphonyl Indazole Derivatives as Potential Ligands of VEGFR2 Kinase. Int J Mol Sci 2020; 21:ijms21134793. [PMID: 32645858 PMCID: PMC7369845 DOI: 10.3390/ijms21134793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/28/2020] [Accepted: 07/03/2020] [Indexed: 01/21/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) is a key receptor in the angiogenesis process. The VEGFR2 expression is upregulated in many cancers so this receptor is an important target for anticancer agents. In the present paper, we analyse interactions of several dimeric indazoles, previously investigated for anticancer activity, with the amino acids present in the VEGFR2 binding pocket. Using the docking method and MD simulations as well as theoretical computations (SAPT0, PIEDA, semi-empirical PM7), we confirmed that these azoles can efficiently bind into the kinase pocket and their poses can be stabilised by the formation of hydrogen bonds, π–π stacking, π–cation, and hybrid interactions with some amino acids of the kinase cavity like Ala866, Lys868, Glu885, Thr916, Glu917, and Phe918.
Collapse
|
23
|
Exploration of carbamide derived pyrimidine-thioindole conjugates as potential VEGFR-2 inhibitors with anti-angiogenesis effect. Eur J Med Chem 2020; 200:112457. [PMID: 32422489 DOI: 10.1016/j.ejmech.2020.112457] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/28/2020] [Accepted: 05/10/2020] [Indexed: 12/12/2022]
Abstract
The development of new small molecules from known structural motifs through molecular hybridization is one of the trends in drug discovery. In this connection, we have combined the two pharmacophoric units (pyrimidine and thioindole) in a single entity via molecular hybridization strategy along with introduction of urea functionality at C2 position of pyrimidine to increase the efficiency of H-bonding interactions. Among the synthesized conjugates 12a-aa, compound 12k was found to exhibit significant IC50 values 5.85, 7.87, 6.41 and 10.43 μM against MDA-MB-231 (breast), HepG2 (liver), A549 (lung) and PC-3 (prostate) cancer cell lines, respectively. All these compounds were further evaluated for their inhibitory activities against VEGFR-2 protein. The results specified that among the tested compounds, 12d, 12e, 12k, 12l, 12p, 12q, 12t and 12u prominently suppressed VEGFR-2, with IC50 values of 310-920 nM in association to the positive control (210 nM). Angiogenesis inhibition was evident by tube formation assay in HUVECs and cell-invasion by transwell assay. The mechanism of cellular toxicity on MDA-MB-231 was found through depolarisation of mitochondrial membrane potential, increased ROS production and subsequent DNA damage resulting in apoptosis induction. Moreover, clonogenic and wound healing assays designated the inhibition of colony formation and cell migration by 12k in a dose-dependent manner. Molecular docking studies also shown that compound 12k capably intermingled with catalytically active residues GLU-885, ASP-1046 of the VEGFR-2 through hydrogen-bonding interactions.
Collapse
|
24
|
Abstract
The urea functionality is inherent to numerous bioactive compounds, including a variety of clinically approved therapies. Urea containing compounds are increasingly used in medicinal chemistry and drug design in order to establish key drug-target interactions and fine-tune crucial drug-like properties. In this perspective, we highlight physicochemical and conformational properties of urea derivatives. We provide outlines of traditional reagents and chemical procedures for the preparation of ureas. Also, we discuss newly developed methodologies mainly aimed at overcoming safety issues associated with traditional synthesis. Finally, we provide a broad overview of urea-based medicinally relevant compounds, ranging from approved drugs to recent medicinal chemistry developments.
Collapse
Affiliation(s)
- Arun K Ghosh
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Margherita Brindisi
- Department of Chemistry and Department of Medicinal Chemistry, Purdue University, West Lafayette, Indiana 47907, United States.,Department of Excellence of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
25
|
Wen N, Guo B, Zheng H, Xu L, Liang H, Wang Q, Wang D, Chen X, Zhang S, Li Y, Zhang L. Bromodomain inhibitor jq1 induces cell cycle arrest and apoptosis of glioma stem cells through the VEGF/PI3K/AKT signaling pathway. Int J Oncol 2019; 55:879-895. [PMID: 31485609 PMCID: PMC6741838 DOI: 10.3892/ijo.2019.4863] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Bromodomain and extraterminal domain proteins, especially bromodomain-containing protein 4 (Brd4), have recently emerged as therapeutic targets for several cancers, although the role and mechanism of Brd4 in glioblastoma multiforme (GBM) are unclear. In this study, we aimed to explore the underlying mechanisms of the anti-tumor effects of Brd4 and the bromodomain inhibitor JQ1 on glioma stem cells (GSCs). In vitro, JQ1 and small interfering RNAs targeting Brd4 (siBrd4) inhibited the proliferation and self-renewal of GSCs. In vivo, JQ1 significantly inhibited the growth of xenograft GSCs tumors. The RNA-seq analysis revealed that the PI3K-AKT pathway played an important role in GBM. Vascular endothelial growth factor (VEGF) and VEGF receptor 2 phosphorylation was downregulated by exposure to JQ1 in GSCs, thereby reducing PI3K and AKT activity. In addition, treatment with JQ1 inhibited MMP expression, thereby inhibiting degradation of the extracellular matrix by MMP and angiogenesis in GBM tumors. Suppression of AKT phosphorylation inhibited the expression of the retinoblastoma/E2F1 complex, resulting in cell cycle arrest. In addition, treatment with siBrd4 or JQ1 induced apoptosis by activating AKT downstream target genes involved in apoptosis. In conclusion, these results suggest that Brd4 has great potential as a therapeutic target, and JQ1 has notable anti-tumor effects against GBM which may be mediated via the VEGF/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Naiyan Wen
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Baofeng Guo
- Department of Plastic Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hongwu Zheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Libo Xu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hang Liang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qian Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ding Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuyang Chen
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shengnan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ling Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
26
|
Abstract
Recent breakthroughs in our understanding of the molecular pathophysiology of retinal vascular disease have allowed us to specifically target pathological angiogenesis while minimizing damage to the neurosensory retina. This is perhaps best exemplified by the development of therapies targeting the potent angiogenic growth factor and vascular permeability mediator, vascular endothelial growth factor (VEGF). Anti-VEGF therapies, initially introduced for the treatment of choroidal neovascularization in patients with age-related macular degeneration, have also had a dramatic impact on the management of retinal vascular disease and are currently an indispensable component for the treatment of macular edema in patients with diabetic eye disease and retinal vein occlusions. Emerging evidence supports expanding the use of therapies targeting VEGF for the treatment of retinal neovascularization in patients with diabetic retinopathy and retinopathy of prematurity. However, VEGF is among a growing list of angiogenic and vascular hyperpermeability factors that promote retinal vascular disease. Many of these mediators are expressed in response to stabilization of a single family of transcription factors, the hypoxia-inducible factors (HIFs), that regulate the expression of these angiogenic stimulators. Here we review the basic principles driving pathological angiogenesis and discuss the current state of retinal anti-angiogenic pharmacotherapy as well as future directions.
Collapse
Affiliation(s)
- Yannis M Paulus
- Kellogg Eye Center, University of Michigan School of Medicine, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 N. Broadway St., Smith Building, 4039, Baltimore, MD, 21287, USA.
| |
Collapse
|
27
|
Zhao S, Chu Y, Zhang Y, Zhou Y, Jiang Z, Wang Z, Mao L, Li K, Sun W, Li P, Jia S, Wang C, Xu A, Loomes K, Tang S, Wu D, Hui X, Nie T. Linifanib exerts dual anti-obesity effect by regulating adipocyte browning and formation. Life Sci 2019; 222:117-124. [PMID: 30708100 DOI: 10.1016/j.lfs.2019.01.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/21/2019] [Accepted: 01/28/2019] [Indexed: 12/31/2022]
Abstract
Obesity is caused by energy imbalance and accompanied by adipocyte hypertrophy and hyperplasia. Therefore, both enhancement of adipocyte energy expenditure and inhibition of adipogenesis are viable ways to combat obesity. Using the Ucp1-2A-luciferase reporter animal model previously reported by us as a screening platform, a chemical compound Linifanib was identified as a potent inducer of UCP1 expression in primary inguinal adipocytes in vitro and in vivo. Signal pathway analyses showed that Linifanib promoted adipocyte browning by attenuating STAT3 phosphorylation. The effects of Linifanib on adipocyte browning were blocked by the compound, SD19, which activates the STAT3 signaling cascade. Linifanib also inhibited adipocyte differentiation, by blocking mitotic clonal expansion, which could be rescued by STAT3 activator. Taken together, our results indicate that Linifanib might serve as a potential drug for the treatment of obesity.
Collapse
Affiliation(s)
- Shiting Zhao
- Central Laboratory of the First Affiliated Hospital, Jinan University, Guangzhou 510630, China; Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Yi Chu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Yuwei Zhang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China; Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Yulai Zhou
- School of Pharmaceutical Sciences, Jilin University, Changchun 130012, Jilin, China
| | - Zhiwu Jiang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Zhengqi Wang
- Central Laboratory of the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Liufeng Mao
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Kuai Li
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Wei Sun
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Peng Li
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Shiqi Jia
- Central Laboratory of the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Cunchuan Wang
- Central Laboratory of the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Kerry Loomes
- School of Biological Sciences & Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Shibing Tang
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, University of Hong Kong, Hong Kong, China.
| | - Tao Nie
- Central Laboratory of the First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
28
|
Spectrophotometric and molecular modelling studies on in vitro interaction of tyrosine kinase inhibitor linifanib with bovine serum albumin. PLoS One 2017; 12:e0176015. [PMID: 28419132 PMCID: PMC5395234 DOI: 10.1371/journal.pone.0176015] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/04/2017] [Indexed: 12/15/2022] Open
Abstract
Linifanib (LNF) possess antitumor activity and acts by inhibiting receptor tyrosine kinase VEGF and PDGF. The interaction of BSA with the drug can provide valuable information regarding the pharmacokinetic and pharmacodynamics behavior of drug. In our study the spectrophotometric methods and molecular docking studies were executed to understand the interaction behavior of BSA and LNF. BSA has an intrinsic fluorescence and that fluorescence was quenched by LNF. This quenching process was studied at three different temperatures of 288, 300and 308 K. The interaction between LNF and BSA was due to static quenching because the Ksv (Stern-Volmer constant) at 288 K was higher than at 300 and 308 K. Kq (quenching rate constant) behaved in a similar fashion as the Ksv. Several other parameters like binding constants, number of binding sites and binding energy in addition to molecular docking studies were also used to evaluate the interaction process. A decrease in the binding constants was observed with increasing temperatures and the binding site number approximated unity. The decreasing binding constant indicates LNF–BSA complex stability. The site mark competition experiment confirmed the binding site for LNF was located on site II of BSA. UV–visible studies along with synchronous fluorescence confirm a small change in the conformation of BSA upon interaction with LNF. The thermodynamic analysis provided the values for free energy ΔG0, ΔH0 and ΔS0. The ΔG0 at the 288, 300 and 308 K ranged in between -21.5 to -23.3 kJ mol-1, whereas the calculated values of ΔH (-55.91 kJ mol-1) and ΔS0 (-111.74 J mol-1·K-1). The experimental and molecular docking results suggest that the interaction between LNF and BSA was spontaneous and they exhibited hydrogen bonding and van der Waals force between them.
Collapse
|
29
|
Design, Synthesis and Structure-Activity Relationships of Novel Diaryl Urea Derivatives as Potential EGFR Inhibitors. Molecules 2016; 21:molecules21111572. [PMID: 27869742 PMCID: PMC6273962 DOI: 10.3390/molecules21111572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 01/09/2023] Open
Abstract
Two novel series of diaryl urea derivatives 5a–i and 13a–l were synthesized and evaluated for their cytotoxicity against H-460, HT-29, A549, and MDA-MB-231 cancer cell lines in vitro. Therein, 4-aminoquinazolinyl-diaryl urea derivatives 5a–i demonstrated significant activity, and seven of them are more active than sorafenib, with IC50 values ranging from 0.089 to 5.46 μM. Especially, compound 5a exhibited the most active potency both in cellular (IC50 = 0.15, 0.089, 0.36, and 0.75 μM, respectively) and enzymatic assay (IC50 = 56 nM against EGFR), representing a promising lead for further optimization.
Collapse
|
30
|
Chen J, Guo J, Chen Z, Wang J, Liu M, Pang X. Linifanib (ABT-869) Potentiates the Efficacy of Chemotherapeutic Agents through the Suppression of Receptor Tyrosine Kinase-Mediated AKT/mTOR Signaling Pathways in Gastric Cancer. Sci Rep 2016; 6:29382. [PMID: 27387652 PMCID: PMC4937412 DOI: 10.1038/srep29382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/17/2016] [Indexed: 12/25/2022] Open
Abstract
Gastric cancer, highly dependent on tumor angiogenesis, causes uncontrolled lethality, in part due to chemoresistance. Here, we demonstrate that linifanib (ABT-869), a novel multi-targeted receptor tyrosine kinase inhibitor, markedly augments cytotoxicity of chemotherapies in human gastric cancer. ABT-869 and chemotherapeutic agents exhibited a strong synergy to inhibit the viability of several gastric cancer cell lines, with combination index values ranging from 0.017 to 0.589. Additionally, the combination of ABT-869 and chemotherapeutic agents led to remarkable suppression of vascular endothelial growth factor (VEGF)-induced angiogenesis in vitro and in vivo. Importantly, in a preclinical gastric cancer xenograft mouse model, drug co-treatments led to increased mouse survival as well as a synergistic reduction in tumor size and the inhibition of tumor angiogenesis. Mechanistic studies further revealed that all of the co-treatments containing ABT-869 resulted in decreased activation of the VEGF receptor, the epidermal growth factor receptor and the insulin growth factor receptor. Inhibition of these receptor tyrosine kinases consequently attenuated the activation of the downstream AKT/mTOR signaling pathway both in cultured gastric cancer cells and in gastric cancer xenografts. Collectively, our findings suggest that the addition of ABT-869 to traditional chemotherapies may be a promising strategy for the treatment of human gastric cancer.
Collapse
Affiliation(s)
- Jing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.,Key Laboratory of Reproduction and Genetics in Ningxia, Ningxia Medical University, Yinchuan 750004, China
| | - Jiawei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhi Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jieqiong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.,Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China.,Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas 77030, USA
| | - Xiufeng Pang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|