1
|
Su Y, Zhu K, Wang J, Liu B, Chang Y, Chang D, You Y. Advancing Src kinase inhibition: From structural design to therapeutic innovation - A comprehensive review. Eur J Med Chem 2025; 287:117369. [PMID: 39952096 DOI: 10.1016/j.ejmech.2025.117369] [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: 12/15/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Src kinase, a non-receptor tyrosine kinase implicated in cellular signaling networks, plays a pivotal role in tumor progression and therapeutic resistance. Despite intensive research efforts spanning decades, no Src-selective kinase inhibitors have yet entered clinical use, highlighting the challenges in developing targeted therapeutics. Here we review recent advances in small-molecule Src inhibitor development, focusing on structural design strategies, binding mechanisms, and therapeutic applications. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and substrate-competitive inhibition that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly through exploitation of non-ATP binding pockets and covalent inhibition strategies. Integration of artificial intelligence, living organoid platforms, and targeted protein degradation technologies is accelerating inhibitor optimization. We discuss key challenges in Src inhibitor development, including the need for enhanced selectivity, reduced off-target effects, and improved resistance profiles. Our analysis reveals promising directions for future therapeutic development, emphasizing the importance of rational design principles guided by structural insights and emerging technologies. These findings provide a framework for developing next-generation Src inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Yifeng Su
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jiahao Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Boyan Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yue Chang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Degui Chang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, 610072, China.
| | - Yaodong You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, 610072, China.
| |
Collapse
|
2
|
Agir N, Georgakopoulos-Soares I, Zaravinos A. A Multi-Omics Analysis of a Mitophagy-Related Signature in Pan-Cancer. Int J Mol Sci 2025; 26:448. [PMID: 39859167 PMCID: PMC11765132 DOI: 10.3390/ijms26020448] [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: 12/13/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Mitophagy, an essential process within cellular autophagy, has a critical role in regulating key cellular functions such as reproduction, metabolism, and apoptosis. Its involvement in tumor development is complex and influenced by the cellular environment. Here, we conduct a comprehensive analysis of a mitophagy-related gene signature, composed of PRKN, PINK1, MAP1LC3A, SRC, BNIP3L, BECN1, and OPTN, across various cancer types, revealing significant differential expression patterns associated with molecular subtypes, stages, and patient outcomes. Pathway analysis revealed a complex interplay between the expression of the signature and potential effects on the activity of various cancer-related pathways in pan-cancer. Immune infiltration analysis linked the mitophagy signature with certain immune cell types, particularly OPTN with immune infiltration in melanoma. Methylation patterns correlated with gene expression and immune infiltration. Mutation analysis also showed frequent alterations in PRKN (34%), OPTN (21%), PINK1 (28%), and SRC (15%), with implications for the tumor microenvironment. We also found various correlations between the expression of the mitophagy-related genes and sensitivity in different drugs, suggesting that targeting this signature could improve therapy efficacy. Overall, our findings underscore the importance of mitophagy in cancer biology and drug resistance, as well as its potential for informing treatment strategies.
Collapse
Affiliation(s)
- Nora Agir
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus;
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| |
Collapse
|
3
|
Lin C, Sniezek CM, McGann CD, Karki R, Giglio RM, Garcia BA, McFaline-Figeroa JL, Schweppe DK. Defining the heterogeneous molecular landscape of lung cancer cell responses to epigenetic inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.592075. [PMID: 38853901 PMCID: PMC11160595 DOI: 10.1101/2024.05.23.592075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Epigenetic inhibitors exhibit powerful antiproliferative and anticancer activities. However, cellular responses to small-molecule epigenetic inhibition are heterogenous and dependent on factors such as the genetic background, metabolic state, and on-/off-target engagement of individual small-molecule compounds. The molecular study of the extent of this heterogeneity often measures changes in a single cell line or using a small number of compounds. To more comprehensively profile the effects of small-molecule perturbations and their influence on these heterogeneous cellular responses, we present a molecular resource based on the quantification of chromatin, proteome, and transcriptome remodeling due to histone deacetylase inhibitors (HDACi) in non-isogenic cell lines. Through quantitative molecular profiling of 10,621 proteins, these data reveal coordinated molecular remodeling of HDACi treated cancer cells. HDACi-regulated proteins differ greatly across cell lines with consistent (JUN, MAP2K3, CDKN1A) and divergent (CCND3, ASF1B, BRD7) cell-state effectors. Together these data provide valuable insight into cell-type driven and heterogeneous responses that must be taken into consideration when monitoring molecular perturbations in culture models.
Collapse
Affiliation(s)
- Chuwei Lin
- Genome Sciences, University of Washington, Seattle, WA 98105, USA
| | | | | | - Rashmi Karki
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ross M. Giglio
- Biomedical Engineer, Columbia University, New York, NY 10027, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Devin K. Schweppe
- Genome Sciences, University of Washington, Seattle, WA 98105, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
4
|
Kook E, Lee J, Kim DH. YES1 as a potential target to overcome drug resistance in EGFR-deregulated non-small cell lung cancer. Arch Toxicol 2024; 98:1437-1455. [PMID: 38443724 DOI: 10.1007/s00204-024-03693-7] [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: 11/24/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024]
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) such as gefitinib and osimertinib have primarily been used as first-line treatments for patients with EGFR-activating mutations in non-small cell lung cancer (NSCLC). Novel biomarkers are required to distinguish patients with lung cancer who are resistant to EGFR-TKIs. The aim of the study is to investigate the expression and functional role of YES1, one of the Src-family kinases, in EGFR-TKI-resistant NSCLC. YES1 expression was elevated in gefitinib-resistant HCC827 (HCC827/GR) cells, harboring EGFR mutations. Moreover, HCC827/GR cells exhibited increased reactive oxygen species (ROS) levels compared to those of the parent cells, resulting in the phosphorylation/activation of YES1 due to oxidation of the cysteine residue. HCC827/GR cells showed elevated expression levels of YES1-associated protein 1 (YAP1), NF-E2-related factor 2 (Nrf2), cancer stemness-related markers, and antioxidant proteins compared to those of the parent cells. Knockdown of YES1 in HCC827/GR cells suppressed YAP1 phosphorylation, leading to the inhibition of Bcl-2, Bcl-xL, and Cyclin D1 expression. Silencing YES1 markedly attenuated the proliferation, migration, and tumorigenicity of HCC827/GR cells. Dasatinib inhibited the proliferation of HCC827/GR cells by targeting YES1-mediated signaling pathways. Furthermore, the combination of gefitinib and dasatinib demonstrated a synergistic effect in suppressing the proliferation of HCC827/GR cells. Notably, YES1- and Nrf2-regulated genes showed a positive regulatory relationship in patients with lung cancer and in TKI-resistant NSCLC cell lines. Taken together, these findings suggest that modulation of YES1 expression and activity may be an attractive therapeutic strategy for the treatment of drug-resistant NSCLC.
Collapse
Affiliation(s)
- Eunjin Kook
- Department of Chemistry, Kyonggi University, Suwon, Gyeonggi-do, 16227, Republic of Korea
| | - JungYeol Lee
- New Drug Discovery Center, DGMIF, Daegu, 41061, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, Gyeonggi-do, 16227, Republic of Korea.
| |
Collapse
|
5
|
Liao Y, Remsing Rix LL, Li X, Fang B, Izumi V, Welsh EA, Monastyrskyi A, Haura EB, Koomen JM, Doebele RC, Rix U. Differential network analysis of ROS1 inhibitors reveals lorlatinib polypharmacology through co-targeting PYK2. Cell Chem Biol 2024; 31:284-297.e10. [PMID: 37848034 PMCID: PMC10922442 DOI: 10.1016/j.chembiol.2023.09.011] [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: 01/30/2023] [Revised: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
Multiple tyrosine kinase inhibitors (TKIs) are often developed for the same indication. However, their relative overall efficacy is frequently incompletely understood and they may harbor unrecognized targets that cooperate with the intended target. We compared several ROS1 TKIs for inhibition of ROS1-fusion-positive lung cancer cell viability, ROS1 autophosphorylation and kinase activity, which indicated disproportionately higher cellular potency of one TKI, lorlatinib. Quantitative chemical and phosphoproteomics across four ROS1 TKIs and differential network analysis revealed that lorlatinib uniquely impacted focal adhesion signaling. Functional validation using pharmacological probes, RNA interference, and CRISPR-Cas9 knockout uncovered a polypharmacology mechanism of lorlatinib by dual targeting ROS1 and PYK2, which form a multiprotein complex with SRC. Rational multi-targeting of this complex by combining lorlatinib with SRC inhibitors exhibited pronounced synergy. Taken together, we show that systems pharmacology-based differential network analysis can dissect mixed canonical/non-canonical polypharmacology mechanisms across multiple TKIs enabling the design of rational drug combinations.
Collapse
Affiliation(s)
- Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xueli Li
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Victoria Izumi
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Eric A Welsh
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Andrii Monastyrskyi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA; Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
6
|
Liang KH, Luo YH, Wang ML, Chiou SH, Chen YM, Hsu HS. A multiomic investigation of lung adenocarcinoma molecular subtypes. J Chin Med Assoc 2024; 87:33-39. [PMID: 37991388 DOI: 10.1097/jcma.0000000000001029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Lung adenocarcinoma-an aggressive and life-threatening malignancy-is a type of non-small-cell lung cancer. Despite medical advancements, the prognosis of lung adenocarcinoma remains unfavorable, likely because of its heterogeneous nature. Furthermore, few subtype-specific treatments are available for lung adenocarcinoma. This study was conducted to explore the molecular subtypes of lung adenocarcinoma. METHODS We performed a joint analysis of transcriptome and proteome data from East Asian patients with lung adenocarcinoma (nonsmokers, 86.5%). RESULTS Four novel subtypes were identified based on distinct molecular characteristics: subtypes I, II, III, and IV. In patients with subtype I lung adenocarcinoma, eukaryotic translation initiation factor 4 gamma 1 activates cell proliferation; inhibiting this factor suppresses tumor growth, and reducing its level induces autophagy. Subtype II is characterized by Kristen rat sarcoma viral oncogene homolog-activating oncogenesis; the onset age of this subtype is the lowest among all subtypes. Subtype III manifests as an advanced disease at diagnosis; it is characterized by a core serum response-related oncogenic signature, which indicates poor overall survival in Western patients with lung cancer. Subtype IV is more common in men than in women; it has astroglial characteristics. A Connectivity Map analysis revealed that the oncogenic expression patterns corresponding to subtypes I, II, III, and IV can be reversed by the inhibitors of Inhibitor of κB (IκB) kinase (eg, withaferin A), mammalian target of rapamycin (eg, everolimus), Src proto-oncogene (Src) (eg, saracatinib), and Transforming Growth Factor (TGF)-β/Smad (eg, LY-364947), respectively. CONCLUSION This study introduced an innovative multiomics data analysis pipeline. Using this approach, we successfully identified four molecular subtypes of lung adenocarcinoma and their candidate therapeutic agents. The newly identified subtypes can be combined with the current biomarkers to generate a comprehensive roadmap for treatment decision-making.
Collapse
Affiliation(s)
- Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Food Safety and Health Risk Assessment, College of Phmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Institute of Biomedical Informatics, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yung-Hung Luo
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Food Safety and Health Risk Assessment, College of Phmaceutical Science, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General, Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
7
|
Deng RR, Yuan YP. Ropivacaine inhibits the proliferation and metastasis of gastric cancer cells via the SNX10/SRC/STAT3 pathway. Chem Biol Drug Des 2024; 103:e14405. [PMID: 37989501 DOI: 10.1111/cbdd.14405] [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: 07/24/2023] [Revised: 10/13/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Gastric cancer currently has no effective treatment due to its high metastasis and heterogeneity. It has been reported that ropivacaine (Rop) can inhibit the growth, migration, and invasion of gastric cancer. However, the therapeutic mechanism of Rop still needs to be further explored to provide insights for its clinical application. This study aimed to explore the effects of Rop on the growth, migration, and invasion of gastric cancer cells and the underlying mechanisms. The expression levels of SNX10 were assessed in gastric cancer tissues and cell line AGS by qRT-PCR. Cell Counting Kit-8 (CCK8) assay, wound-healing assay, and transwell assay were then used to examine the effects of Rop on the AGS cell viability, migration, invasion, and proliferation, respectively. Additionally, colony formation assay was used to measure cell proliferation ability, and flow cytometry was used to detect apoptosis level. Protein levels of SNX10, SRC, and STAT3 were detected by western blot. According to the experimental results, the decreased SNX10 mRNA expression was observed in gastric cancer tissue and cell line AGS. Rop inhibited the proliferation, migration, and invasion of AGS cells, but promoted apoptosis and upregulated SNX10 expression. Moreover, Rop inhibited the expression of MMP-2 and MMP-9, phosphorylation of SRC and STAT3. SNX10 knockdown could reverse Rop-induced anticancer effects. Collectively, Rop showed a potential role in preventing proliferation and metastasis of gastric cancer. The action mechanism of Rop may be related to the upregulation of SNX10 expression and further inhibition of SRC/STAT3 signaling pathway. Our findings provide new insights into the anticancer properties of Rop.
Collapse
Affiliation(s)
- Rong-Rong Deng
- Department of Anaesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of Anaesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| | - You-Ping Yuan
- Department of Anaesthesiology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Department of Anaesthesiology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, China
| |
Collapse
|
8
|
Garg A, Karhana S, Bano A, Khan IA, Reeta, Nidhi, Khan MA. Network pharmacology and molecular docking study-based approach to explore mechanism of benzimidazole-based anthelmintics for the treatment of lung cancer. J Biomol Struct Dyn 2023; 42:10739-10760. [PMID: 37740654 DOI: 10.1080/07391102.2023.2258419] [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: 11/07/2022] [Accepted: 09/07/2023] [Indexed: 09/24/2023]
Abstract
Emerging studies have reported the potential anticancer activity of benzimidazole-based anthelmintics (BBA) against lung cancer (LC). However, mechanism underlying the anticancer activity of BBA is unclear. Therefore, in the current study, network pharmacology and molecular docking-based approach were used to explore the potential molecular mechanism for the treatment of LC. The potential targets for BBA were obtained from multiple databases including SwissTargetPrediction, Drug Bank, Therapeutic Target Database, and Comparative Toxicogenomics Database while LC targets were collected from DisGeNet gene discovery platform, Integrated Genomic Database of NSCLC, Catalogue of Somatic Mutations in Cancer and Online Mendelian Inheritance in Man database. Protein-protein interaction (PPI) diagram of common targets was constructed using STRING online platform. Topological analysis was performed using Cytoscape and gene enrichment analysis was conducted using FunRich software. Highest degree targets were then confirmed using molecular docking and molecular dynamics simulations. The BBA were prioritized according to their S scores, with ricobendazole ranking highest followed by flubendazole, fenbendazole, mebendazole, triclabendazole, albendazole, oxibendazole, parbendazole, thiabendazole and oxfendazole. The potential targets of BBA identified using topological analysis and molecular docking were found to be CCND1 (cyclin D1), EGFR (Epidermal Growth Factor Receptor), ERBB2 (Erb-B2 Receptor Tyrosine Kinase 2/CD340), PTGS2 (Prostaglandin-endoperoxide synthase 2), and SRC (Proto-oncogene tyrosine-protein kinase). Furthermore, molecular dynamics confirmed that CCND1 and EGFR are the potential targets of ricobendazole for the treatment of LC. BBA can be further explored as a therapeutic strategy for the treatment of lung cancer under in vitro and in vivo studies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aakriti Garg
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sonali Karhana
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Aysha Bano
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Imran A Khan
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Reeta
- Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, New Delhi, India
| | - Nidhi
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Ashif Khan
- Centre for Translational & Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| |
Collapse
|
9
|
Yang Z, Guo H, Zhang K, Jiang Z, Jin R, Guo D, Li Z, Wang Y, Meng L. The Design, Synthesis, and Evaluation of Evodiamine Derivatives with
Hydroxy Groups. LETT DRUG DES DISCOV 2023; 20:1135-1146. [DOI: 10.2174/1570180819666220903150621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
Background:
Most of the structural modifications to evodiamine (EVO) have focused on the
3- and 10-positions, while structural modifications to the EVO 2-position have not yet been reported. In
this study, we investigated the scaffold diversity and bioactivity of EVO from position 2 to gain more
insight into the influence of the chemical space around EVO on bioactivity.
Objective:
The study aims to synthesize two derivatives of EVO with hydroxy groups, 8a and 8b, and to
investigate the antitumor activity of EVO derivatives with hydroxy groups compared to EVO.
Methods:
The synthesized compounds were structurally characterized by 1H NMR, 13C NMR, and mass
spectrometry. The effects of compounds 8a, 8b, and EVO on the proliferation of H460, A549, and Eca109
cells in vitro were determined by MTT. The effect of EVO, 8a and 8b on apoptosis of H460 cells was
investigated by the annexed V-FITC/propidium iodide (PI) combination assay. The expression of EVO,
8a and 8b on apoptosis-related proteins was examined by Western blot analysis. To simulate the binding
ability between small molecules and proteins, molecular docking calculations of EGFRWT and EGFRT790M
with 8a and 8b, respectively, were performed using Schrödinger software.
Results:
In the cytotoxicity assay, compound 8b showed lower IC50 values for the three tumor cell lines
(6.69 μM for H460 cells, 20.02 μM for A549 cells, and 16.47 μM for Eca109 cells) compared to compound
8a and EVO, and 8b induced apoptosis by affecting apoptosis-related proteins CRAF, AKT, and
ERK in a late apoptotic manner. The molecular docking results showed that 8b has a good binding ability
to EGFR upstream of apoptosis-related proteins.
Conclusion:
These findings suggest that 8b has significantly higher antitumor biological activity than
EVO and 8a. This antitumor effect has important implications for the study of EVO derivatives in antitumor
models.
Collapse
Affiliation(s)
- Zheng Yang
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Hui Guo
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
- Instrumental Analysis Center, Xi'an Jiao Tong University, Xi'an, 710049,
P.R. China
| | - Keyao Zhang
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering
Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong
Province, 519000, China
| | - Ruyi Jin
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Dongyan Guo
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Zhi Li
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Yuwei Wang
- College of Pharmacy, Shaanxi Key Lab Basic & New Herbal Medicament Res, Shaanxi University of Chinese Medicine,
Xian Yang, Shaanxi 712046, China
| | - Lingjie Meng
- Instrumental Analysis Center, Xi'an Jiao Tong University, Xi'an, 710049,
P.R. China
| |
Collapse
|
10
|
Maldonado H, Leyton L. CSK-mediated signalling by integrins in cancer. Front Cell Dev Biol 2023; 11:1214787. [PMID: 37519303 PMCID: PMC10382208 DOI: 10.3389/fcell.2023.1214787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Cancer progression and metastasis are processes heavily controlled by the integrin receptor family. Integrins are cell adhesion molecules that constitute the central components of mechanosensing complexes called focal adhesions, which connect the extracellular environment with the cell interior. Focal adhesions act as key players in cancer progression by regulating biological processes, such as cell migration, invasion, proliferation, and survival. Src family kinases (SFKs) can interplay with integrins and their downstream effectors. SFKs also integrate extracellular cues sensed by integrins and growth factor receptors (GFR), transducing them to coordinate metastasis and cell survival in cancer. The non-receptor tyrosine kinase CSK is a well-known SFK member that suppresses SFK activity by phosphorylating its specific negative regulatory loop (C-terminal Y527 residue). Consequently, CSK may play a pivotal role in tumour progression and suppression by inhibiting SFK oncogenic effects in several cancer types. Remarkably, CSK can localise near focal adhesions when SFKs are activated and even interact with focal adhesion components, such as phosphorylated FAK and Paxillin, among others, suggesting that CSK may regulate focal adhesion dynamics and structure. Even though SFK oncogenic signalling has been extensively described before, the specific role of CSK and its crosstalk with integrins in cancer progression, for example, in mechanosensing, remain veiled. Here, we review how CSK, by regulating SFKs, can regulate integrin signalling, and focus on recent discoveries of mechanotransduction. We additionally examine the cross talk of integrins and GFR as well as the membrane availability of these receptors in cancer. We also explore new pharmaceutical approaches to these signalling pathways and analyse them as future therapeutic targets.
Collapse
Affiliation(s)
- Horacio Maldonado
- Receptor Dynamics in Cancer Laboratory, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
11
|
Ehm P, Bettin B, Jücker M. Activated Src kinases downstream of BCR-ABL and Flt3 induces proteasomal degradation of SHIP1 by phosphorylation of tyrosine 1021. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119467. [PMID: 36958526 DOI: 10.1016/j.bbamcr.2023.119467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/03/2023] [Accepted: 03/12/2023] [Indexed: 03/25/2023]
Abstract
Within the various subtypes of ALL, patients with a BCR-ABL-positive background as well as with a genetic change in the KMT2A gene have by far the worst survival probabilities. Interestingly, both subtypes are characterized by highly activated tyrosine kinases. SHIP1 serves as an important negative regulator of the PI3K/AKT signaling pathway, which is often constitutively activated in ALL. The protein expression of SHIP1 is decreased in most T-ALL and in some subgroups of B-ALL. In this study, we analyzed the expression of SHIP1 protein in detail in the context of groups with aberrant activated tyrosine kinases, namely BCR-ABL (Ph+) and Flt3 (KMT2A translocations). We demonstrate that constitutively activated Src kinases downstream of BCR-ABL and receptor tyrosine kinases reduce the SHIP1 expression in a SHIP1-Y1021 phosphorylated-dependent manner with subsequent ubiquitin marked proteasomal degradation. Inhibition of BCR-ABL (Imatinib), Flt3 (Quizartinib) or Src-Kinase-Family (Saracatinib) leads to significant reconstitution of SHIP1 protein expression. These results further support a functional role of SHIP1 as tumor suppressor protein and could be the basis for the establishment of a targeted therapy form.
Collapse
Affiliation(s)
- Patrick Ehm
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; Research Institute Children's Cancer Center Hamburg, Hamburg and Dept. of Pediatric Oncology and Hematology, University Medical Center, Hamburg, Germany.
| | - Bettina Bettin
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
12
|
Princiotto S, Musso L, Manetti F, Marcellini V, Maga G, Crespan E, Perini C, Zaffaroni N, Beretta GL, Dallavalle S. Synthesis and biological activity evaluation of 3-(hetero) arylideneindolin-2-ones as potential c-Src inhibitors. J Enzyme Inhib Med Chem 2022; 37:2382-2394. [PMID: 36050846 PMCID: PMC9448371 DOI: 10.1080/14756366.2022.2117317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Inhibition of c-Src is considered one of the most studied approaches to cancer treatment, with several heterocyclic compounds approved during the last 15 years as chemotherapeutic agents. Starting from the biological evaluation of an in-house collection of small molecules, indolinone was selected as the most promising scaffold. In this work, several functionalised indolinones were synthesised and their inhibitory potency and cytotoxic activity were assayed. The pharmacological profile of the most active compounds, supported by molecular modelling studies, revealed that the presence of an amino group increased the affinity towards the ATP-binding site of c-Src. At the same time, bulkier derivatizations seemed to improve the interactions within the enzymatic pocket. Overall, these data represent an early stage towards the optimisation of new, easy-to-be functionalised indolinones as potential c-Src inhibitors.
Collapse
Affiliation(s)
- Salvatore Princiotto
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Loana Musso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| | - Fabrizio Manetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Valentina Marcellini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Siena, Italy
| | - Giovanni Maga
- Institute of Molecular Genetics IGM, CNR "Luigi Luca Cavalli-Sforza", Pavia, Italy
| | - Emmanuele Crespan
- Institute of Molecular Genetics IGM, CNR "Luigi Luca Cavalli-Sforza", Pavia, Italy
| | - Cecilia Perini
- Institute of Molecular Genetics IGM, CNR "Luigi Luca Cavalli-Sforza", Pavia, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giovanni Luca Beretta
- Molecular Pharmacology Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Sabrina Dallavalle
- Department of Food, Environmental and Nutritional Sciences (DeFENS), University of Milan, Milan, Italy
| |
Collapse
|
13
|
Zhang M, Wang Y, Amin A, Khan MA, Yu Z, Liang C. Network Pharmacology Analysis of Bioactive Components and Mechanisms of Action of Qi Wei Wan Formula for Treating Non-Small Cell Lung Carcinoma. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221120215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Astragali Radix (AR) and Schisandrae chinensis Fructus (SCF) have been used individually and in traditional Chinese medicine (TCM) formulas for treating non-small cell lung carcinoma (NSCLC). Qi Wei Wan (QWW), a 2-herb TCM formula composed of AR and SCF, is used to treat blood deficiency, fatigue, and metabolic abnormalities. We speculate that QWW may be more effective in treating NSCLC than AR or SCF alone. We identified 28 bioactive compounds in QWW and 322 targets of these compounds from databases. Network pharmacology analysis was used to identify 248 putative NSCLC-related gene targets of the bioactive compounds in QWW. Common target genes were analyzed to build protein–protein interaction networks. Implicated biological functions and pathways (p53, PI3K-Akt, etc) were identified by Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses. Molecular docking of core target proteins with the key active compounds was also performed. This study identified the potential gene targets and mechanisms involved in the anti-NSCLC effects of QWW.
Collapse
Affiliation(s)
- Minghe Zhang
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Ye Wang
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Aftab Amin
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Muhammad Ajmal Khan
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhiling Yu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Chun Liang
- Division of Life Science and State Key Lab of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
- EnKang Pharmaceuticals (Guangzhou), Ltd., Guangzhou, China
| |
Collapse
|
14
|
Bello-Alvarez C, Zamora-Sánchez CJ, Camacho-Arroyo I. Rapid Actions of the Nuclear Progesterone Receptor through cSrc in Cancer. Cells 2022; 11:cells11121964. [PMID: 35741094 PMCID: PMC9221966 DOI: 10.3390/cells11121964] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 12/30/2022] Open
Abstract
The nuclear progesterone receptor (PR) is mainly known for its role as a ligand-regulated transcription factor. However, in the last ten years, this receptor’s extranuclear or rapid actions have gained importance in the context of physiological and pathophysiological conditions such as cancer. The PR’s polyproline (PXPP) motif allows protein–protein interaction through SH3 domains of several cytoplasmatic proteins, including the Src family kinases (SFKs). Among members of this family, cSrc is the most well-characterized protein in the scenario of rapid actions of the PR in cancer. Studies in breast cancer have provided the most detailed information on the signaling and effects triggered by the cSrc–PR interaction. Nevertheless, the study of this phenomenon and its consequences has been underestimated in other types of malignancies, especially those not associated with the reproductive system, such as glioblastomas (GBs). This review will provide a detailed analysis of the impact of the PR–cSrc interplay in the progression of some non-reproductive cancers, particularly, in GBs.
Collapse
Affiliation(s)
- Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| | - Carmen J Zamora-Sánchez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México C.P. 0451, Mexico
| |
Collapse
|
15
|
Lai Y, Chang H, Chen H, Chang G, Chen JJW. Peruvoside is a novel Src inhibitor that suppresses NSCLC cell growth and motility by downregulating multiple Src-EGFR-related pathways. Am J Cancer Res 2022; 12:2576-2593. [PMID: 35812056 PMCID: PMC9251685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/11/2022] [Indexed: 06/15/2023] Open
Abstract
The tyrosine kinase Src plays an essential role in the progression of many cancers and is involved in several epidermal growth factor receptor (EGFR)-mediated signalling pathways. To improve the efficacy of lung cancer treatments, this study aimed to identify novel compounds that can disrupt the Src-EGFR interaction and that are less dependent on EGFR status with wild-type and mutations than other compounds. We used the Src pY419 ELISA as the platform to screen a compound library of more than 400 plant-derived active ingredients and identified peruvoside as a candidate Src-EGFR crosstalk inhibitor. The effects of peruvoside were evaluated by western blotting, cell function assays, combination Index (CI)-isobologram analyses and in vivo experiments. Peruvoside significantly suppressed the phosphorylation of Src, EGFR, and signal transducer and activator of transcription 3 (STAT3) in a dose- and time-dependent manner and somewhat suppressed their protein expression. Cell function assays revealed that peruvoside inhibited the proliferation, invasion, migration, and colony formation of lung cancer cells in vitro and tumour growth in vivo. Furthermore, peruvoside sensitized gefitinib-resistant tumour cells (A549, PC9/gef and H1975) to gefitinib treatment, indicating that peruvoside may exert synergistic effects when used in combination with established therapeutic agents. Our data also demonstrated that the inhibitory effects of peruvoside on lung cancer progression might be attributed to its ability to regulate Src, phosphoinositide 3-kinase (PI3K), c-Jun N-terminal kinase (JNK), Paxillin, p130cas, and EGFR. Our findings suggest that peruvoside suppresses non-small-cell lung carcinoma (NSCLC) malignancy by downregulating multiple Src-related pathways and could serve as a potential base molecule for developing new anticancer drugs and therapeutic strategies for lung cancer.
Collapse
Affiliation(s)
- Yihua Lai
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Rheumatic Diseases Research Center, China Medical University HospitalTaichung, Taiwan
- College of Medicine, China Medical UniversityTaichung, Taiwan
- Rheumatology and Immunology Center, China Medical University HospitalTaichung, Taiwan
| | - Hsiuhui Chang
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Hueiwen Chen
- Graduate Institute of Toxicology, National Taiwan University College of MedicineTaipei, Taiwan
| | - Geechen Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Chung Shan Medical University HospitalTaichung, Taiwan
- School of Medicine, Chung Shan Medical UniversityTaichung, Taiwan
| | - Jeremy JW Chen
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Biotechnology Center, National Chung Hsing UniversityTaichung, Taiwan
- Institute of Molecular Biology, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
16
|
Glück M, Dally L, Jücker M, Ehm P. JAK2-V617F is a negative regulation factor of SHIP1 protein and thus influences the AKT signaling pathway in patients with Myeloproliferative Neoplasm (MPN). Int J Biochem Cell Biol 2022; 149:106229. [PMID: 35609769 DOI: 10.1016/j.biocel.2022.106229] [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: 12/16/2021] [Revised: 04/19/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Myeloproliferative neoplasms (MPN) are a group of chronic haematological disorders. At the molecular level of MPN cells, the gain-of-function mutation V617F of the Janus kinase 2 (JAK2) leads to a constitutive activation of the downstream signaling cascade and is a conventional criteria for diagnosis. Here, the functional role of the tumor suppressor SHIP1 (SH2 domain containing inositol-5 phosphatase 1) in the pathogenesis of MPNs was investigated. METHODS Primary blood samples of MPN-patients were analysed using Western Blot technique regarding the level of SHIP1 expression. Moreover, SHIP1 and SHIP1-mutations were lentivirally transduced in the JAK2-V617F-positive UKE-1 cell line and expression was monitored over time. In addition, we examined SHIP1 reconstitution by inhibition of JAK2-V617F. Furthermore, we transfected SHIP1-expressing cells with a JAK2-V617F respectively a BCR-ABL construct and investigated changes in SHIP1 expression. RESULTS Four out of five MPN-patient samples showed a loss or a reduction in SHIP1 expression. We identified JAK2 as a negative regulator of SHIP1 expression in MPN cells and inhibition of JAK2-V617F implicates a reconstituted SHIP1 expression. This is significant because SHIP1 negatively regulates the AKT signaling pathway and in consequence the reconstitution of SHIP1 expression leads to a decreased cell growth. Moreover, we examined the impact of SHIP1 and patient-derived SHIP1-mutations on AKT phosphorylation and show the benefit of a combined therapy in MPN cells with inhibitors of the AKT/mTOR pathway. CONCLUSION In summary, the data suggest that SHIP1 may play a role during the development of MPNs and could be the basis for establishing a targeted therapy.
Collapse
Affiliation(s)
- Madeleine Glück
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Lina Dally
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Patrick Ehm
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
17
|
The Pyrazolo[3,4-d]Pyrimidine Derivative Si306 Encapsulated into Anti-GD2-Immunoliposomes as Therapeutic Treatment of Neuroblastoma. Biomedicines 2022; 10:biomedicines10030659. [PMID: 35327462 PMCID: PMC8945814 DOI: 10.3390/biomedicines10030659] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 12/03/2022] Open
Abstract
Si306, a pyrazolo[3,4-d]pyrimidine derivative recently identified as promising anticancer agent, has shown favorable in vitro and in vivo activity profile against neuroblastoma (NB) models by acting as a competitive inhibitor of c-Src tyrosine kinase. Nevertheless, Si306 antitumor activity is associated with sub-optimal aqueous solubility, which might hinder its further development. Drug delivery systems were here developed with the aim to overcome this limitation, obtaining suitable formulations for more efficacious in vivo use. Si306 was encapsulated in pegylated stealth liposomes, undecorated or decorated with a monoclonal antibody able to specifically recognize and bind to the disialoganglioside GD2 expressed by NB cells (LP[Si306] and GD2-LP[Si306], respectively). Both liposomes possessed excellent morphological and physio-chemical properties, maintained over a period of two weeks. Compared to LP[Si306], GD2-LP[Si306] showed in vitro specific cellular targeting and increased cytotoxic activity against NB cell lines. After intravenous injection in healthy mice, pharmacokinetic profiles showed increased plasma exposure of Si306 when delivered by both liposomal formulations, compared to that obtained when Si306 was administered as free form. In vivo tumor homing and cytotoxic effectiveness of both liposomal formulations were finally tested in an orthotopic animal model of NB. Si306 tumor uptake resulted significantly higher when encapsulated in GD2-LP, compared to Si306, either free or encapsulated into untargeted LP. This, in turn, led to a significant increase in survival of mice treated with GD2-LP[Si306]. These results demonstrate a promising antitumor efficacy of Si306 encapsulated into GD2-targeted liposomes, supporting further therapeutic developments in pre-clinical trials and in the clinic for NB.
Collapse
|
18
|
Martin-Fernandez ML. Fluorescence Imaging of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Resistance in Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14030686. [PMID: 35158954 PMCID: PMC8833717 DOI: 10.3390/cancers14030686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Lung cancer is the leading cause of cancer-related deaths, with a low (<21%) 5-year survival rate. Lung cancer is often driven by the misfunction of molecules on the surface of cells of the epithelium, which orchestrate mechanisms by which these cells grow and proliferate. Beyond common non-specific treatments, such as chemotherapy or radiotherapy, among molecular-specific treatments, a number of small-molecule drugs that block cancer-driven molecular activity have been developed. These drugs initially have significant success in a subset of patients, but these patients systematically develop resistance within approximately one year of therapy. Substantial efforts towards understanding the mechanisms of resistance have focused on the genomics of cancer progression, the response of cells to the drugs, and the cellular changes that allow resistance to develop. Fluorescence microscopy of many flavours has significantly contributed to the last two areas, and is the subject of this review. Abstract Non-small cell lung cancer (NSCLC) is a complex disease often driven by activating mutations or amplification of the epidermal growth factor receptor (EGFR) gene, which expresses a transmembrane receptor tyrosine kinase. Targeted anti-EGFR treatments include small-molecule tyrosine kinase inhibitors (TKIs), among which gefitinib and erlotinib are the best studied, and their function more often imaged. TKIs block EGFR activation, inducing apoptosis in cancer cells addicted to EGFR signals. It is not understood why TKIs do not work in tumours driven by EGFR overexpression but do so in tumours bearing classical activating EGFR mutations, although the latter develop resistance in about one year. Fluorescence imaging played a crucial part in research efforts to understand pro-survival mechanisms, including the dysregulation of autophagy and endocytosis, by which cells overcome the intendedly lethal TKI-induced EGFR signalling block. At their core, pro-survival mechanisms are facilitated by TKI-induced changes in the function and conformation of EGFR and its interactors. This review brings together some of the main advances from fluorescence imaging in investigating TKI function and places them in the broader context of the TKI resistance field, highlighting some paradoxes and suggesting some areas where super-resolution and other emerging methods could make a further contribution.
Collapse
Affiliation(s)
- Marisa L Martin-Fernandez
- Central Laser Facility, Science & Technology Facilities Council, Rutherford Appleton Laboratory, Didcot OX11 0FA, UK
| |
Collapse
|
19
|
Kaewmeesri P, Pocasap P, Kukongviriyapan V, Prawan A, Kongpetch S, Senggunprai L. Anti-metastatic Potential of Natural Triterpenoid Cucurbitacin B Against Cholangiocarcinoma Cells by Targeting Src Protein. Integr Cancer Ther 2022; 21:15347354221124861. [PMID: 36154723 PMCID: PMC9520142 DOI: 10.1177/15347354221124861] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/30/2022] [Accepted: 08/23/2022] [Indexed: 11/28/2022] Open
Abstract
Owing to the crucial role of Src in cancer metastasis, interruption of Src and its signaling has been considered a promising strategy for cancer metastasis treatment. Cucurbitacin B, a dietary triterpenoid, has been shown to possess anti-proliferative and apoptosis-inducing activities in cholangiocarcinoma (CCA) cells via suppressing the activation of FAK which is a main downstream Src effector. We hypothesized that cucurbitacin B might act as a Src suppressant which conferring anti-metastasis effect against CCA cells. To investigate this, the role of Src in regulating metastasis behavior of CCA cells and the effect of cucurbitacin B on Src-mediated metastatic phenotype of these cells were determined. The results showed that activation of Src significantly enhanced the migratory and invasive abilities of CCA cells. Molecular analysis revealed that Src-facilitated metastasis behavior of CCA cells occurred by modifying expression of a wide range of metastasis-related genes in the cells. Consistent with gene expression results, activation of Src significantly induced the protein expression of 2 important metastasis-associated molecules, MMP-9 and VEGF. Cucurbitacin B markedly suppressed activation of Src and its key effector, FAK. As a consequence, the alteration of expression profiles of metastasis-associated genes induced by Src activator in CCA cells was diminished by cucurbitacin B treatment. The compound also down-regulated Src-induced expression of MMP-9 and VEGF proteins in the cells. Moreover, molecular docking analysis revealed that cucurbitacin B could interact with Src kinase domain and possibly restrain the kinase from being activated by hindering the ATP binding. In conclusion, cucurbitacin B exhibited anti-metastatic property in CCA cells via negatively influencing Src and Src-related oncogenic signaling. This compound may therefore be a potential therapeutic drug for further development as an anti-Src agent for treatment of metastatic CCA.
Collapse
Affiliation(s)
- Putthaporn Kaewmeesri
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Piman Pocasap
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
20
|
Kalal BS, Modi PK, Najar MA, Behera SK, Upadhya D, Prasad TSK, Pai VR. Hyperphosphorylation of HDAC2 promotes drug resistance in a novel dual drug resistant mouse melanoma cell line model: an in vitro study. Am J Cancer Res 2021; 11:5881-5901. [PMID: 35018231 PMCID: PMC8727796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023] Open
Abstract
Drug-resistant melanoma is very difficult to treat, and a novel approach is needed to overcome resistance. The present study aims at identifying the alternate pathways utilized in the dual drug-resistant mouse melanoma cells (B16F10R) for their survival and proliferation. The dual drug-resistant mouse melanoma, B16F10R, was established by treating the cells with a combination of U0126 (MEK1/2 inhibitor) and LY294002 (PI3K-AKT kinase inhibitor) in a dose-escalating manner till they attained a resistance fold factor of ≥2. The altered phosphoproteome in the B16F10R, as compared to the parental B16F10C, was analyzed using a high-resolution Orbitrap Fusion Tribrid mass spectrometer. Histone deacetylases 2 (HDAC2) was validated for its role in drug resistance by using its inhibitor, valproic acid (VPA). In the B16F10R cells, 363 altered phosphoproteins were identified, among which 126 were hyperphosphorylated, and 137 were hypophosphorylated (1.5-fold change). Pathway analysis shows the altered phosphoproteins are from RNA metabolism and cell cycle proteins. Inhibition of HDAC2 by VPA induces apoptosis in B16F10C and B16F10R. The present study highlights the role of HDAC2, a cell cycle regulator, in the development of resistance to dual drugs in murine melanoma. Therefore, designing leads for targeting HDAC2 along with key signaling pathways may be explored in treatment strategies.
Collapse
Affiliation(s)
- Bhuvanesh Sukhlal Kalal
- Department of Biochemistry, Yenepoya Medical College, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Santosh Kumar Behera
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Department of Anatomy, Kasturba Medical College, Manipal Academy of Higher Education ManipalUdupi 576104, Karnataka, India
| | | | - Vinitha Ramanath Pai
- Department of Biochemistry, Yenepoya Medical College, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| |
Collapse
|
21
|
Mechelke T, Wittig F, Ramer R, Hinz B. Interleukin-1β Induces Tissue Factor Expression in A549 Cells via EGFR-Dependent and -Independent Mechanisms. Int J Mol Sci 2021; 22:ijms22126606. [PMID: 34205482 PMCID: PMC8235322 DOI: 10.3390/ijms22126606] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/03/2022] Open
Abstract
Tissue factor (TF) plays an important role in the progression and angiogenesis of tumor cells. The present study investigated the mechanism of interleukin-1β (IL-1β)-induced TF expression in A549 lung cancer cells. Based on mRNA and protein analyses, including appropriate inhibitor experiments, IL-1β was shown to induce TF expression in a time-dependent manner, mediated by IL-1 receptor-dependent phosphorylation of the mitogen-activated protein kinases (MAPK) p38, p42/44 and c-jun N-terminal kinase (JNK), as well as the Src kinase and the epidermal growth factor receptor (EGFR). Thereby, inhibition of EGFR transactivation by the Src inhibitor PP1 or direct EGFR inhibition by the EGFR tyrosine kinase inhibitor (TKI) erlotinib led to a reduction of IL-1β-induced TF expression and to a suppression of p42/44 MAPK and EGFR activation, while IL-1β-induced p38 MAPK and JNK activation remained unchanged. A knockdown of EGFR by siRNA was associated with decreased IL-1β-mediated p42/44 MAPK activation, which was no longer inhibitable by erlotinib. Concentration-dependent inhibition of IL-1β-induced TF expression was also observed in the presence of gefitinib and afatinib, two other EGFR TKIs. In summary, our results suggest that IL-1β leads to increased TF formation in lung cancer cells via both Src/EGFR/p42/44 MAPK-dependent and EGFR-independent signaling pathways, with the latter mediated via p38 MAPK and JNK.
Collapse
|
22
|
Chiu LY, Hsin IL, Tsai JN, Chen CJ, Ou CC, Wu WJ, Sheu GT, Ko JL. Combination treatment of Src inhibitor Saracatinib with GMI, a Ganoderma microsporum immunomodulatory protein, induce synthetic lethality via autophagy and apoptosis in lung cancer cells. J Cell Physiol 2020; 236:1148-1157. [PMID: 32686156 DOI: 10.1002/jcp.29924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022]
Abstract
Saracatinib is an oral Src-kinase inhibitor and has been studied in preclinical models and clinical trials of cancer therapy. GMI, a fungal immunomodulatory protein from Ganoderma microsporum, possesses antitumor capacity. The aim of this study is to evaluate the cytotoxic effect of combination treatment with saracatinib and GMI on parental and pemetrexed-resistant lung cancer cells. Cotreatment with saracatinib and GMI induced synergistic and additive cytotoxic effect in A549 and A400 cells by annexin V/propidium iodide assay and combination index. Using western blot assay, saracatinib, and GMI combined treatment synergistically induced caspase-7 activation in A549 cells. Different from A549 cells, saracatinib and GMI cotreatment markedly increased LC3B-II in A400 cells. ATG5 silencing abolished the caspase-7 activation and reduced cell death in A549 cells after cotreatment. This is the first study to provide a novel strategy of treating lung cancer with or without drug resistance via combination treatment with GMI and saracatinib.
Collapse
Affiliation(s)
- Ling-Yen Chiu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Exercise Health Science, National Taiwan University of Sport, Taichung, Taiwan
| | - I-Lun Hsin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jen-Ning Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Jung Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chu-Chyn Ou
- School of Nutrition, Chung Shan Medical University, Taichung, Taiwan
| | - Wen-Jun Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Gwo-Tarng Sheu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Internal Medicine, Division of Medical Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
23
|
Weng CW, Li JH, Tsai JY, Lin SH, Chang GC, Liu CC, Chen JJW. Pharmacophore-based virtual screening for the identification of the novel Src inhibitor SJG-136 against lung cancer cell growth and motility. Am J Cancer Res 2020; 10:1668-1690. [PMID: 32642283 PMCID: PMC7339285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 05/25/2020] [Indexed: 06/11/2023] Open
Abstract
Aberrant elevated Src activity is related to lung cancer growth and metastasis. Therefore, the development of potent small molecule inhibitors to target Src kinase is a potential therapeutic strategy for lung cancer. This study aimed to develop a computational model for the in silico screening of Src inhibitors and then assess the suppressive effect of candidate compounds on cellular functions. A 3D-quantitative structure-activity relationship (QSAR) pharmacophore model consisting of two hydrogen bond acceptors and two hydrophobic regions was constructed by using 28 structurally diverse compounds with IC50 values spanning four orders of magnitude. A National Cancer Institute (NCI) compound dataset was employed for virtual screening by applying the pharmacophore model and molecular docking. Candidate compounds were chosen from the top 20% of scored hits. Among these compounds, the suppressive effects of 30 compounds available in the NCI on Src phosphorylation were validated by using an enzyme-linked immunosorbent assay. Among these compounds, SJG-136, a pyrrolobenzodiazepine dimer, showed a significant inhibitory effect against Src activity in a dose-dependent manner. Further investigations showed that SJG-136 can inhibit lung cancer cell proliferation, clonogenicity, invasion and migration in vitro and tumour growth in vivo. Furthermore, SJG-136 also had an inhibitory effect on Src-related signaling pathways, including the FAK, paxillin, p130Cas, PI3K, AKT, and MEK pathways. In conclusion, we have established a pharmacophore-based virtual screening approach to identify novel Src inhibitors that can inhibit lung cancer cell growth and motility through suppressing Src-related pathways. These findings may contribute to the development of targeted drugs for lung cancer treatment, such as lead compounds.
Collapse
Affiliation(s)
- Chia-Wei Weng
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Jia-Hua Li
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Jeng-Yuan Tsai
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Shih-Hsuan Lin
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
| | - Gee-Chen Chang
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General HospitalTaichung, Taiwan
| | - Chun-Chi Liu
- Institute of Genomics and Bioinformatics, National Chung Hsing UniversityTaichung, Taiwan
| | - Jeremy JW Chen
- Institute of Biomedical Sciences, National Chung Hsing UniversityTaichung, Taiwan
- Biotechnology Center, National Chung Hsing UniversityTaichung, Taiwan
- Institute of Molecular Biology, National Chung Hsing UniversityTaichung, Taiwan
| |
Collapse
|
24
|
Lima NC, Atkinson E, Bunney TD, Katan M, Huang PH. Targeting the Src Pathway Enhances the Efficacy of Selective FGFR Inhibitors in Urothelial Cancers with FGFR3 Alterations. Int J Mol Sci 2020; 21:E3214. [PMID: 32370101 PMCID: PMC7246793 DOI: 10.3390/ijms21093214] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 01/08/2023] Open
Abstract
Selective FGFR inhibitors such as infigratinib (BGJ398) and erdafitinib (JNJ-42756493) have been evaluated in clinical trials for cancers with FGFR3 molecular alterations, particularly in urothelial carcinoma patients. However, a substantial proportion of these patients (up to 50%) display intrinsic resistance to these drugs and receive minimal clinical benefit. There is thus an unmet need for alternative therapeutic strategies to overcome primary resistance to selective FGFR inhibitors. In this study, we demonstrate that cells expressing cancer-associated activating FGFR3 mutants and the FGFR3-TACC3 fusion showed primary resistance to infigratinib in long-term colony formation assays in both NIH-3T3 and urothelial carcinoma models. We find that expression of these FGFR3 molecular alterations resulted in elevated constitutive Src activation compared to wildtype FGFR3 and that cells co-opted this pathway as a means to achieve intrinsic resistance to infigratinib. Targeting the Src pathway with low doses of the kinase inhibitor dasatinib synergistically sensitized multiple urothelial carcinoma lines harbouring endogenous FGFR3 alterations to infigratinib. Our data provide preclinical rationale that supports the use of dasatinib in combination with selective FGFR inhibitors as a means to overcome intrinsic drug resistance in the salvage therapy setting in urothelial cancer patients with FGFR3 molecular alterations.
Collapse
Affiliation(s)
- Nadia Carvalho Lima
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (N.C.L.); (E.A.)
| | - Eliza Atkinson
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (N.C.L.); (E.A.)
| | - Tom D. Bunney
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK; (T.D.B.); (M.K.)
| | - Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, UK; (T.D.B.); (M.K.)
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (N.C.L.); (E.A.)
| |
Collapse
|
25
|
Du G, Rao S, Gurbani D, Henning NJ, Jiang J, Che J, Yang A, Ficarro SB, Marto JA, Aguirre AJ, Sorger PK, Westover KD, Zhang T, Gray NS. Structure-Based Design of a Potent and Selective Covalent Inhibitor for SRC Kinase That Targets a P-Loop Cysteine. J Med Chem 2020; 63:1624-1641. [PMID: 31935084 PMCID: PMC7493195 DOI: 10.1021/acs.jmedchem.9b01502] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SRC is a major regulator of many signaling pathways and contributes to cancer development. However, development of a selective SRC inhibitor has been challenging, and FDA-approved SRC inhibitors, dasatinib and bosutinib, are multitargeted kinase inhibitors. Here, we describe our efforts to develop a selective SRC covalent inhibitor by targeting cysteine 277 on the P-loop of SRC. Using a promiscuous covalent kinase inhibitor (CKI) SM1-71 as a starting point, we developed covalent inhibitor 15a, which discriminates SRC from other covalent targets of SM1-71 including TAK1 and FGFR1. As an irreversible covalent inhibitor, compound 15a exhibited sustained inhibition of SRC signaling both in vitro and in vivo. Moreover, 15a exhibited potent antiproliferative effects in nonsmall cell lung cancer cell lines harboring SRC activation, thus providing evidence that this approach may be promising for further drug development efforts.
Collapse
Affiliation(s)
- Guangyan Du
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Suman Rao
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
- Laboratory of Systems Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Deepak Gurbani
- Departments of Biochemistry and Radiation Oncology , The University of Texas Southwestern Medical Center at Dallas , Dallas , Texas 75390 , United States
| | - Nathaniel J Henning
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Jie Jiang
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Jianwei Che
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Annan Yang
- Department of Medical Oncology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Scott B Ficarro
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jarrod A Marto
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Andrew J Aguirre
- Department of Medical Oncology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Peter K Sorger
- Laboratory of Systems Biology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Kenneth D Westover
- Departments of Biochemistry and Radiation Oncology , The University of Texas Southwestern Medical Center at Dallas , Dallas , Texas 75390 , United States
| | - Tinghu Zhang
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| | - Nathanael S Gray
- Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
- Department of Cancer Biology , Dana Farber Cancer Institute , 450 Brookline Avenue , Boston , Massachusetts 02215 , United States
| |
Collapse
|
26
|
Shen J, Li L, Yang T, Cohen PS, Sun G. Biphasic Mathematical Model of Cell-Drug Interaction That Separates Target-Specific and Off-Target Inhibition and Suggests Potent Targeted Drug Combinations for Multi-Driver Colorectal Cancer Cells. Cancers (Basel) 2020; 12:cancers12020436. [PMID: 32069833 PMCID: PMC7072552 DOI: 10.3390/cancers12020436] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/04/2020] [Accepted: 02/11/2020] [Indexed: 11/25/2022] Open
Abstract
Quantifying the response of cancer cells to a drug, and understanding the mechanistic basis of the response, are the cornerstones for anti-cancer drug discovery. Classical single target-based IC50 measurements are inadequate at describing cancer cell responses to targeted drugs. In this study, based on an analysis of targeted inhibition of colorectal cancer cell lines, we develop a new biphasic mathematical model that accurately describes the cell–drug response. The model describes the drug response using three kinetic parameters: ratio of target-specific inhibition, F1, potency of target-specific inhibition, Kd1, and potency of off-target toxicity, Kd2. Determination of these kinetic parameters also provides a mechanistic basis for predicting effective combination targeted therapy for multi-driver cancer cells. The experiments confirmed that a combination of inhibitors, each blocking a driver pathway and having a distinct target-specific effect, resulted in a potent and synergistic blockade of cell viability, improving potency over mono-agent treatment by one to two orders of magnitude. We further demonstrate that mono-driver cancer cells represent a special scenario in which F1 becomes nearly 100%, and the drug response becomes monophasic. Application of this model to the responses of >400 cell lines to kinase inhibitor dasatinib revealed that the ratio of biphasic versus monophasic responses is about 4:1. This study develops a new mathematical model of quantifying cancer cell response to targeted therapy, and suggests a new framework for developing rational combination targeted therapy for colorectal and other multi-driver cancers.
Collapse
Affiliation(s)
- Jinyan Shen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (J.S.); (L.L.); (T.Y.); (P.S.C.)
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Li Li
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (J.S.); (L.L.); (T.Y.); (P.S.C.)
- Department of Cell Biology and Medical Genetics, Shanxi Medical University, Taiyuan 030001, China
| | - Tao Yang
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (J.S.); (L.L.); (T.Y.); (P.S.C.)
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Paul S. Cohen
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (J.S.); (L.L.); (T.Y.); (P.S.C.)
| | - Gongqin Sun
- Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; (J.S.); (L.L.); (T.Y.); (P.S.C.)
- Correspondence: ; Tel.: +1-401-874-5937
| |
Collapse
|
27
|
Canonici A, Browne AL, Ibrahim MFK, Fanning KP, Roche S, Conlon NT, O’Neill F, Meiller J, Cremona M, Morgan C, Hennessy BT, Eustace AJ, Solca F, O’Donovan N, Crown J. Combined targeting EGFR and SRC as a potential novel therapeutic approach for the treatment of triple negative breast cancer. Ther Adv Med Oncol 2020; 12:1758835919897546. [PMID: 32064003 PMCID: PMC6987485 DOI: 10.1177/1758835919897546] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Background: Triple negative breast cancer (TNBC) is an aggressive subtype of breast
cancer with limited therapeutic options. Epidermal growth factor receptor
(EGFR) has been shown to be over-expressed in TNBC and represents a rational
treatment target. Methods: We examined single agent and combination effects for afatinib and dasatinib
in TNBC. We then determined IC50 and combination index values
using Calcusyn. Functional analysis of single and combination treatments was
performed using reverse phase protein array and cell cycle analysis.
Finally, we determined the anticancer effects of the combination in
vivo. Results: A total of 14 TNBC cell lines responded to afatinib with IC50
values ranging from 0.008 to 5.0 µM. Three cell lines, belonging to the
basal-like subtype of TNBC, were sensitive to afatinib. The addition of
afatinib enhanced response to the five other targeted therapies in HCC1937
and HDQP1 cells. The combination of afatinib with dasatinib caused the
greatest growth inhibition in both cell lines. The afatinib/dasatinib
combination was synergistic and/or additive in 13/14 TNBC cell lines.
Combined afatinib/dasatinib treatment induced G1 cell cycle arrest. Reverse
phase protein array results showed the afatinib/dasatinib combination
resulted in efficient inhibition of both pERK(T202/T204) and pAkt(S473)
signalling in BT20 cells, which was associated with the greatest
antiproliferative effects. High baseline levels of pSrc(Y416) and pMAPK(p38)
correlated with sensitivity to afatinib, whereas low levels of B-cell
lymphoma 2 (Bcl2) and mammalian target of rapamycin (mTOR) correlated with
synergistic growth inhibition by combined afatinib and dasatinib treatment.
In vivo, the combination treatment inhibited tumour
growth in a HCC1806 xenograft model. Conclusions: We demonstrate that afatinib combined with dasatinib has potential clinical
activity in TNBC but warrants further preclinical investigation.
Collapse
Affiliation(s)
- Alexandra Canonici
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Alacoque L. Browne
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Mohamed F. K. Ibrahim
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Kevin P. Fanning
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Sandra Roche
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Neil T. Conlon
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Fiona O’Neill
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Justine Meiller
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - Mattia Cremona
- Medical Oncology Group, Department of Molecular
Medicine, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin,
Ireland
| | - Clare Morgan
- Medical Oncology Group, Department of Molecular
Medicine, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin,
Ireland
| | - Bryan T. Hennessy
- Medical Oncology Group, Department of Molecular
Medicine, Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin,
Ireland
| | | | - Flavio Solca
- Boehringer Ingelheim RCV GmbH & Co KG,
Vienna, Austria
| | - Norma O’Donovan
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
| | - John Crown
- National Institute for Cellular Biotechnology,
Dublin City University, Dublin, Ireland
- Department of Medical Oncology, St Vincent’s
University Hospital, Dublin, Ireland
| |
Collapse
|
28
|
Bhummaphan N, Petpiroon N, Prakhongcheep O, Sritularak B, Chanvorachote P. Lusianthridin targeting of lung cancer stem cells via Src-STAT3 suppression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152932. [PMID: 31100681 DOI: 10.1016/j.phymed.2019.152932] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cancer stem cells (CSCs) are well-recognized as a majority cause of treatment failure and can give rise to relapse. The discovery of compounds attenuating CSCs' properties is crucial for enabling advances in novel therapeutics to limit recurrence. CSCs' features in lung cancer are regulated through a reduction in Src-STAT3-c-Myc, which drives cancer progression, drug resistance, and metastasis. METHODS The effect of lusianthridin suppresses CSC-like phenotypes was determined by 3D culture and anchorage independent growth. The expression of CSC markers and associated proteins were determined by Western blot analyses. Protein ubiquitination and degradation were assessed using immunoprecipitation. RESULTS Herein, we report that lusianthridin, a pure compound from Dendrobium venustum, dramatically suppressed CSCs in lung cancer cells as verified by several CSC phenotype assessments and CSC markers. The CSC phenotypes in lusianthridin-treated cells were suppressed through downregulation of Src-STAT3-c-Myc pathways. Ectopic Src introduced by the transfection augmented CSC phenotypes in lung cancer cells through STAT3 (increased active p-STAT3Tyr705) and c-Myc signals, while the ShRNA-Src transfection or Src inhibitor dasatinib exhibited opposite results. Treatment of the Src-overexpressing cells with lusianthridin resulted in the reversal of active STAT3 (p-STAT3Tyr705) and c-Myc as well as the CSC marker CD133. Importantly, we confirmed the CSC-targeted activity of lusianthridin in CSC-rich primary lung cancer cells. The compound dramatically inhibited the formation of tumor spheres of primary lung cancer cells. Finally, we demonstrated that after CSC-attenuation by lusianthridin, the lung cancer cells exhibited significantly higher susceptibility to chemotherapeutic drugs. Such a sensitizing effect caused by pro-survival suppression and pro-apoptotic induction together with the abolishment of stemness indicated by the decrease in CSC markers CD133, ABCG2, and ALDH1A1. CONCLUSION These findings revealed a novel pharmacological action and the underlying mechanism of lusianthridin in negatively regulating CSC-like phenotypes and sensitizing resistant cancer cells to cemetery.
Collapse
Affiliation(s)
- Narumol Bhummaphan
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Chulalongkorn University, Bangkok 10330, Thailand; Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nalinrat Petpiroon
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Ornjira Prakhongcheep
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand.
| |
Collapse
|
29
|
Li X, Li Z, Wu X, Xiong Z, Yang T, Fu Z, Liu X, Tan X, Zhong F, Wan X, Wang D, Ding X, Yang R, Hou H, Li C, Liu H, Chen K, Jiang H, Zheng M. Deep Learning Enhancing Kinome-Wide Polypharmacology Profiling: Model Construction and Experiment Validation. J Med Chem 2019; 63:8723-8737. [PMID: 31364850 DOI: 10.1021/acs.jmedchem.9b00855] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The kinome-wide virtual profiling of small molecules with high-dimensional structure-activity data is a challenging task in drug discovery. Here, we present a virtual profiling model against a panel of 391 kinases based on large-scale bioactivity data and the multitask deep neural network algorithm. The obtained model yields excellent internal prediction capability with an auROC of 0.90 and consistently outperforms conventional single-task models on external tests, especially for kinases with insufficient activity data. Moreover, more rigorous experimental validations including 1410 kinase-compound pairs showed a high-quality average auROC of 0.75 and confirmed many novel predicted "off-target" activities. Given the verified generalizability, the model was further applied to various scenarios for depicting the kinome-wide selectivity and the association with certain diseases. Overall, the computational model enables us to create a comprehensive kinome interaction network for designing novel chemical modulators or drug repositioning and is of practical value for exploring previously less studied kinases.
Collapse
Affiliation(s)
- Xutong Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zhaojun Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Information Management, Dezhou University, 566 West University Road, Dezhou 253023, China
| | - Xiaolong Wu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zhaoping Xiong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Tianbiao Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Zunyun Fu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaohong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Xiaoqin Tan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Feisheng Zhong
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaozhe Wan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Dingyan Wang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xiaoyu Ding
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ruirui Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Hui Hou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chunpu Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Hong Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Kaixian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.,School of Life Science and Technology, ShanghaiTech University, 393 Huaxiazhong Road, Shanghai 200031, China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| |
Collapse
|
30
|
Cheng F, Lu W, Liu C, Fang J, Hou Y, Handy DE, Wang R, Zhao Y, Yang Y, Huang J, Hill DE, Vidal M, Eng C, Loscalzo J. A genome-wide positioning systems network algorithm for in silico drug repurposing. Nat Commun 2019; 10:3476. [PMID: 31375661 PMCID: PMC6677722 DOI: 10.1038/s41467-019-10744-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 05/26/2019] [Indexed: 01/28/2023] Open
Abstract
Recent advances in DNA/RNA sequencing have made it possible to identify new targets rapidly and to repurpose approved drugs for treating heterogeneous diseases by the 'precise' targeting of individualized disease modules. In this study, we develop a Genome-wide Positioning Systems network (GPSnet) algorithm for drug repurposing by specifically targeting disease modules derived from individual patient's DNA and RNA sequencing profiles mapped to the human protein-protein interactome network. We investigate whole-exome sequencing and transcriptome profiles from ~5,000 patients across 15 cancer types from The Cancer Genome Atlas. We show that GPSnet-predicted disease modules can predict drug responses and prioritize new indications for 140 approved drugs. Importantly, we experimentally validate that an approved cardiac arrhythmia and heart failure drug, ouabain, shows potential antitumor activities in lung adenocarcinoma by uniquely targeting a HIF1α/LEO1-mediated cell metabolism pathway. In summary, GPSnet offers a network-based, in silico drug repurposing framework for more efficacious therapeutic selections.
Collapse
Affiliation(s)
- Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Chuang Liu
- Alibaba Research Center for Complexity Sciences, Hangzhou Normal University, 311121, Hangzhou, China
| | - Jiansong Fang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruisheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yuzheng Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, 200237, Shanghai, China
| | - Yi Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, 200237, Shanghai, China
| | - Jin Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
| | - David E Hill
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Xu D, Tian W, Jiang C, Huang Z, Zheng S. The anthelmintic agent oxfendazole inhibits cell growth in non‑small cell lung cancer by suppressing c‑Src activation. Mol Med Rep 2019; 19:2921-2926. [PMID: 30720086 DOI: 10.3892/mmr.2019.9897] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 12/04/2018] [Indexed: 11/05/2022] Open
Abstract
The c‑Src protein family of tyrosine kinases are important in the tumorigenesis of many types of tumors, and may be a potential target for antitumor drug discovery. In the present study, immunoblotting was performed to analyze protein expression, CCK‑8 assay was carried out to assess cell viability and cell cycle was analyzed using a flow cytometer. The anthelmintic agent oxfendazole was observed to be a novel c‑Src inhibitor that blocked the activation of c‑Src. Oxfendazole also suppressed the cell growth of non‑small cell lung cancer (NSCLC) cells, and overexpression of c‑Src decreased the cytotoxicity of oxfendazole against NSCLC cells. In addition, oxfendazole induced cell cycle arrest at the G0/G1 phase, and downregulated the protein levels of Cyclin‑dependent kinase (CDK)‑4, CDK6, retinoblastoma protein and E2 transcription factor 1, and upregulated the expression levels of p53 and p21 in NSCLC cells. Furthermore, oxfendazole enhanced the cytotoxicity of cisplatin against NSCLC cells. These results demonstrated that oxfendazole exerted its antitumor activity by suppressing c‑Src signaling, and it was also indicated that the anthelmintic agent oxfendazole may be effective for anti‑NSCLC therapy in the clinic as a single agent or in combination with other antitumor drugs.
Collapse
Affiliation(s)
- Dafu Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wenze Tian
- Department of Thoracic Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Chao Jiang
- Department of Oncology, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Ziming Huang
- Department of Emergency Surgery, Huai'an First People's Hospital, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Shiying Zheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
32
|
Ma X, Zhang L, Song J, Nguyen E, Lee RS, Rodgers SJ, Li F, Huang C, Schittenhelm RB, Chan H, Chheang C, Wu J, Brown KK, Mitchell CA, Simpson KJ, Daly RJ. Characterization of the Src-regulated kinome identifies SGK1 as a key mediator of Src-induced transformation. Nat Commun 2019; 10:296. [PMID: 30655532 PMCID: PMC6336867 DOI: 10.1038/s41467-018-08154-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 12/20/2018] [Indexed: 12/13/2022] Open
Abstract
Despite significant progress, our understanding of how specific oncogenes transform cells is still limited and likely underestimates the complexity of downstream signalling events. To address this gap, we use mass spectrometry-based chemical proteomics to characterize the global impact of an oncogene on the expressed kinome, and then functionally annotate the regulated kinases. As an example, we identify 63 protein kinases exhibiting altered expression and/or phosphorylation in Src-transformed mammary epithelial cells. An integrated siRNA screen identifies nine kinases, including SGK1, as being essential for Src-induced transformation. Accordingly, we find that Src positively regulates SGK1 expression in triple negative breast cancer cells, which exhibit a prominent signalling network governed by Src family kinases. Furthermore, combined inhibition of Src and SGK1 reduces colony formation and xenograft growth more effectively than either treatment alone. Therefore, this approach not only provides mechanistic insights into oncogenic transformation but also aids the design of improved therapeutic strategies. The systemic understanding of oncogenic kinase signalling is still limited. Here, the authors combine chemical proteomics with functional screens to assess the impact of oncogenic Src on the expressed kinome and identify SGK1 as a critical mediator of Src-induced cell transformation.
Collapse
Affiliation(s)
- Xiuquan Ma
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Luxi Zhang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jiangning Song
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Monash Centre for Data Science, Faculty of Information Technology, Monash University, Melbourne, VIC, 3800, Australia
| | - Elizabeth Nguyen
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Rachel S Lee
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Samuel J Rodgers
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Fuyi Li
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Cheng Huang
- Monash Biomedical Proteomics Facility and Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Ralf B Schittenhelm
- Monash Biomedical Proteomics Facility and Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Howard Chan
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Chanly Chheang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Jianmin Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Centre for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kristin K Brown
- Cancer Therapeutics Program and Cancer Metabolism Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Christina A Mitchell
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Kaylene J Simpson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.,Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, 3800, Australia. .,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
33
|
Bazzani L, Donnini S, Finetti F, Christofori G, Ziche M. PGE2/EP3/SRC signaling induces EGFR nuclear translocation and growth through EGFR ligands release in lung adenocarcinoma cells. Oncotarget 2018; 8:31270-31287. [PMID: 28415726 PMCID: PMC5458206 DOI: 10.18632/oncotarget.16116] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Prostaglandin E2 (PGE2) interacts with tyrosine kinases receptor signaling in both tumor and stromal cells supporting tumor progression. Here we demonstrate that in non-small cell lung carcinoma (NSCLC) cells, A549 and GLC82, PGE2 promotes nuclear translocation of epidermal growth factor receptor (nEGFR), affects gene expression and induces cell growth. Indeed, cyclin D1, COX-2, iNOS and c-Myc mRNA levels are upregulated following PGE2 treatment. The nuclear localization sequence (NLS) of EGFR as well as its tyrosine kinase activity are required for the effect of PGE2 on nEGFR and downstream signaling activities. PGE2 binds its bona fide receptor EP3 which by activating SRC family kinases, induces ADAMs activation which, in turn, releases EGFR-ligands from the cell membrane and promotes nEGFR. Amphiregulin (AREG) and Epiregulin (EREG) appear to be involved in nEGFR promoted by the PGE2/EP3-SRC axis. Pharmacological inhibition or silencing of the PGE2/EP3/SRC-ADAMs signaling axis or EGFR ligands i.e. AREG and EREG expression abolishes nEGFR induced by PGE2. In conclusion, PGE2 induces NSCLC cell proliferation by EP3 receptor, SRC-ADAMs activation, EGFR ligands shedding and finally, phosphorylation and nEGFR. Since nuclear EGFR is a hallmark of cancer aggressiveness, our findings reveal a novel mechanism for the contribution of PGE2 to tumor progression.
Collapse
Affiliation(s)
- Lorenzo Bazzani
- Department of Life Sciences, University of Siena, 53100, Siena, Italy.,Department of Biomedizin, University of Basel, 4058, Basel, Switzerland
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | - Federica Finetti
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| | | | - Marina Ziche
- Department of Life Sciences, University of Siena, 53100, Siena, Italy
| |
Collapse
|
34
|
Kim YJ, Hong S, Sung M, Park MJ, Jung K, Noh KW, Oh DY, Lee MS, Oh E, Shin YK, Choi YL. LYN expression predicts the response to dasatinib in a subpopulation of lung adenocarcinoma patients. Oncotarget 2018; 7:82876-82888. [PMID: 27756880 PMCID: PMC5347739 DOI: 10.18632/oncotarget.12657] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 10/01/2016] [Indexed: 01/07/2023] Open
Abstract
Therapies targeting SRC family kinases (SFKs) have shown efficacy in treating non-small cell lung cancer (NSCLC). However, recent clinical trials have found that the SFK inhibitor dasatinib is ineffective in some patient cohorts. Regardless, dasatinib treatment may benefit some NSCLC patient subgroups. Here, we investigated whether expression of LYN, a member of the SFK family, is associated with patient survival, the efficacy of dasatinib, and/or NSCLC cell viability. LYN expression was associated with poor overall survival in a multivariate analysis, and this association was strongest in non-smoker female patients with adenocarcinoma (ADC). In lung ADC cells, LYN expression enhanced cell proliferation, migration, and invasion. Dasatinib inhibited LYN activity and decreased cell viability in LYN-positive ADC cell lines and xenografts. Additionally, we identified the SFKs SRC and YES as candidate dasatinib targets in LYN-negative ADC cell lines. Our findings suggest that LYN is a useful prognostic marker and a selective target of dasatinib therapy in the lung ADC subpopulation especially in female non-smokers with lung ADC.
Collapse
Affiliation(s)
- Yu Jin Kim
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sungyoul Hong
- Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Minjung Sung
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min Jeong Park
- Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Kyungsoo Jung
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Ka-Won Noh
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Doo-Yi Oh
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Mi-Sook Lee
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Ensel Oh
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea.,The Center for Anti-cancer Companion Diagnostics, Bio-MAX/N-Bio, Seoul National University, Seoul, Korea
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea.,Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Chang N, Lee HW, Lim JE, Jeong DE, Song HJ, Kim S, Nam DH, Sung HH, Jeong BC, Seo SI, Jeon SS, Lee HM, Choi HY, Jeon HG. Establishment and antitumor effects of dasatinib and PKI-587 in BD-138T, a patient-derived muscle invasive bladder cancer preclinical platform with concomitant EGFR amplification and PTEN deletion. Oncotarget 2018; 7:51626-51639. [PMID: 27438149 PMCID: PMC5239502 DOI: 10.18632/oncotarget.10539] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/29/2016] [Indexed: 11/25/2022] Open
Abstract
Muscle-invasive bladder cancer (MIBC) consists of a heterogeneous group of tumors with a high rate of metastasis and mortality. To facilitate the in-depth investigation and validation of tailored strategies for MIBC treatment, we have developed an integrated approach using advanced high-throughput drug screening and a clinically relevant patient-derived preclinical platform. We isolated patient-derived tumor cells (PDCs) from a rare MIBC case (BD-138T) that harbors concomitant epidermal growth factor receptor (EGFR) amplification and phosphatase and tensin homolog (PTEN) deletion. High-throughput in vitro drug screening demonstrated that dasatinib, a SRC inhibitor, and PKI-587, a dual PI3K/mTOR inhibitor, exhibited targeted anti-proliferative and pro-apoptotic effects against BD-138T PDCs. Using established patient-derived xenograft models that successfully retain the genomic and molecular characteristics of the parental tumor, we confirmed that these anti-tumor responses occurred through the inhibition of SRC and PI3K/AKT/mTOR signaling pathways. Taken together, these experimental results demonstrate that dasatinib and PKI-587 might serve as promising anticancer drug candidates for treating MIBC with combined EGFR gene amplification and PTEN deletion.
Collapse
Affiliation(s)
- Nakho Chang
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea
| | - Hye Won Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea.,Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea
| | - Joung Eun Lim
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Da Eun Jeong
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Hye Jin Song
- Department of Anatomy and Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Sudong Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea.,Samsung Advanced Institute of Technology, Samsung Electronics Co., Ltd., Seoul 06351, Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.,Institute for Refractory Cancer Research, Samsung Medical Center, Seoul 06351, Korea
| | - Hyun Hwan Sung
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Byong Chang Jeong
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Seong Soo Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hyun Moo Lee
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Han-Yong Choi
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| |
Collapse
|
36
|
Nakanishi T, Menju T, Nishikawa S, Takahashi K, Miyata R, Shikuma K, Sowa T, Imamura N, Hamaji M, Motoyama H, Hijiya K, Aoyama A, Sato T, Chen‐Yoshikawa TF, Sonobe M, Date H. The synergistic role of ATP-dependent drug efflux pump and focal adhesion signaling pathways in vinorelbine resistance in lung cancer. Cancer Med 2018; 7:408-419. [PMID: 29318780 PMCID: PMC5806107 DOI: 10.1002/cam4.1282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 10/13/2017] [Accepted: 11/13/2017] [Indexed: 12/31/2022] Open
Abstract
The vinorelbine (VRB) plus cisplatin regimen is widely used to treat non-small cell lung cancer (NSCLC), but its cure rate is poor. Drug resistance is the primary driver of chemotherapeutic failure, and the causes of resistance remain unclear. By focusing on the focal adhesion (FA) pathway, we have highlighted a signaling pathway that promotes VRB resistance in lung cancer cells. First, we established VRB-resistant (VR) lung cancer cells (NCI-H1299 and A549) and examined its transcriptional changes, protein expressions, and activations. We treated VR cells by Src Family Kinase (SFK) inhibitors or gene silencing and examined cell viabilities. ATP-binding Cassette Sub-family B Member 1 (ABCB1) was highly expressed in VR cells. A pathway analysis and western blot analysis revealed the high expression of integrins β1 and β3 and the activation of FA pathway components, including Src family kinase (SFK) and AKT, in VR cells. SFK involvement in VRB resistance was confirmed by the recovery of VRB sensitivity in FYN knockdown A549 VR cells. Saracatinib, a dual inhibitor of SFK and ABCB1, had a synergistic effect with VRB in VR cells. In conclusion, ABCB1 is the primary cause of VRB resistance. Additionally, the FA pathway, particularly integrin, and SFK, are promising targets for VRB-resistant lung cancer. Further studies are needed to identify clinically applicable target drugs and biomarkers that will improve disease prognoses and predict therapeutic efficacies.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenosine Triphosphate/pharmacology
- Adult
- Aged
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis
- Biomarkers, Tumor/metabolism
- Carcinoma, Large Cell/drug therapy
- Carcinoma, Large Cell/metabolism
- Carcinoma, Large Cell/pathology
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Proliferation
- Drug Resistance, Neoplasm
- Female
- Focal Adhesions/drug effects
- Focal Adhesions/metabolism
- Focal Adhesions/pathology
- Follow-Up Studies
- Humans
- Lung Neoplasms/drug therapy
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Prognosis
- Signal Transduction/drug effects
- Survival Rate
- Tumor Cells, Cultured
- Vinorelbine/pharmacology
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Takao Nakanishi
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
- Department of Thoracic SurgeryKobe‐City Nishi‐Kobe Medical CenterKobeJapan
| | - Toshi Menju
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Shigeto Nishikawa
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Koji Takahashi
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Ryo Miyata
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Kei Shikuma
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Terumasa Sowa
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Naoto Imamura
- Department of Thoracic SurgeryJapanese Red Cross Wakayama Medical CenterWakayamaJapan
| | - Masatsugu Hamaji
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Hideki Motoyama
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Kyoko Hijiya
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Akihiro Aoyama
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Toshihiko Sato
- Institute for Advancement of Clinical and Translational ScienceKyoto University HospitalKyotoJapan
| | | | - Makoto Sonobe
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Hiroshi Date
- Department of Thoracic SurgeryGraduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
37
|
Bhummaphan N, Pongrakhananon V, Sritularak B, Chanvorachote P. Cancer Stem Cell-Suppressing Activity of Chrysotoxine, a Bibenzyl from Dendrobium pulchellum. J Pharmacol Exp Ther 2018; 364:332-346. [PMID: 29217540 DOI: 10.1124/jpet.117.244467] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/28/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) have been recognized as rare populations driving cancer progression, metastasis, and drug resistance in leading cancers. Attempts have been made toward identifying compounds that specifically target these CSCs. Therefore, investigations of novel therapeutic strategies for CSC targeting are required. The cytotoxic effects of chrysotoxine on human non-small cell lung cancer-derived H460 and H23 cells were evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. The effects of chrysotoxine suppression of CSC-like phenotypes were determined in CSC-rich populations and primary CSCs in three-dimensional (3D) culture and in an extreme limiting dilution assay. Expression of CSC markers and associated proteins was determined by Western blot analyse and flow cytometry. We have reported herein the CSC-suppressing activity of chrysotoxine, a bibenzyl compound isolated from Dendrobium pulchellum We have shown, to our knowledge for the first time, that chrysotoxine dramatically suppresses CSC-like phenotypes of H460 and H23 cells. Treatment with chrysotoxine significantly reduced the viability of 3D CSC-rich populations and concomitantly decreased known CSC markers. Chrysotoxine suppressed CSC phenotypes through downregulation of Src/protein kinase B (Akt) signaling. Active (phosphorylated Y416) Src was shown to regulate cancer stemness, since ectopic overexpression of Src strongly activated Akt and subsequently enhanced pluripotency transcription factor SRY (sex-determining region Y)-box 2 (Sox2)- mediating CSC phenotypes, whereas the short hairpin RNA of Src and an Src inhibitor (dasatinib) suppressed Akt, Sox2, and CSC properties. Importantly, chrysotoxine was shown to suppress active Src/Akt signaling and in turn depleted Sox2-mediated CSCs. Our findings indicate a novel CSC-targeted role of chrysotoxine and its regulation by Src/Akt and Sox2, which may be exploited for cancer treatment.
Collapse
Affiliation(s)
- Narumol Bhummaphan
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Department of Pharmacognosy and Pharmaceutical Botany; Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology; and Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Varisa Pongrakhananon
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Department of Pharmacognosy and Pharmaceutical Botany; Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology; and Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Boonchoo Sritularak
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Department of Pharmacognosy and Pharmaceutical Botany; Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology; and Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| | - Pithi Chanvorachote
- Inter-Department Program of Biomedical Sciences, Faculty of Graduate School, Department of Pharmacognosy and Pharmaceutical Botany; Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology; and Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, Thailand
| |
Collapse
|
38
|
Rizeq B, Zakaria Z, Ouhtit A. Towards understanding the mechanisms of actions of carcinoembryonic antigen-related cell adhesion molecule 6 in cancer progression. Cancer Sci 2018; 109:33-42. [PMID: 29110374 PMCID: PMC5765285 DOI: 10.1111/cas.13437] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/21/2022] Open
Abstract
Human carcinoembryonic antigen (CEA) is the prototypic member of a family of highly related cell surface glycoproteins that includes carcinoembryonic antigen‐related cell adhesion molecule 6 (CEACAM6) and others. CEACAM6 (formerly NCA), which belongs to the immunoglobulin superfamily, is a cell adhesion protein of the CEA family. It is normally expressed on the epithelial surfaces and on the surface of myeloid cells (CD66c). CEACAM6 is a multi‐functional glycoprotein that mediates homotypic binding with other CEA family members and heterotypic binding with integrin receptors. It functions by organizing tissue architecture and regulating different signal transduction, while aberrant expression leads to the development of human malignancies. It was first discovered in proliferating cells of adenomas and hyperplastic polyps in comparison to benign colonic tissue when overexpressed on the surface of various cell types in model systems. CEACAM6 functions as a pan‐inhibitor of cell differentiation and cell polarization, and it also causes distortion of tissue architecture. Moreover, overexpression of CEACAM6 modulates cancer progression through aberrant cell differentiation, anti‐apoptosis, cell growth and resistance to therapeutic agents. In addition, CEACAM6 overexpression in multiple malignancies promotes cell invasion and metastasis, thereby representing an acquired advantage of tumor cells directly responsible for an invasive phenotype. This review focuses on the findings supporting the mechanisms of actions linking the oncogenic potential of CEACAM6 to the onset of cancer progression and pathogenesis, especially in breast cancer, and to validating CEACAM6 as a target to pave the way towards the design of efficient therapeutic strategies against breast cancer.
Collapse
Affiliation(s)
- Balsam Rizeq
- Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Zain Zakaria
- Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| |
Collapse
|
39
|
Isfort S, Crysandt M, Gezer D, Koschmieder S, Brümmendorf TH, Wolf D. Bosutinib: A Potent Second-Generation Tyrosine Kinase Inhibitor. Recent Results Cancer Res 2018; 212:87-108. [PMID: 30069626 DOI: 10.1007/978-3-319-91439-8_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bosutinib is one of the five tyrosine kinase inhibitors which are currently approved for the treatment of chronic myeloid leukemia. By its dual inhibition of Src and ABL kinase and also targeting further kinases, it creates a unique target portfolio which also explains its unique side effect profile. The approval of bosutinib in 2013 made the drug available for patients previously treated with one or more tyrosine kinase inhibitor(s) and for whom imatinib, nilotinib, and dasatinib are not considered appropriate treatment options. As initially the first-line clinical trial comparing bosutinib with imatinib in CML patients in chronic phase did not reach its primary endpoint and therefore the product was not licensed for first-line therapy, a second first-line trial, the so-called BFORE study, was performed and just recently the promising results have been published predicting a quick expansion of the existing label. In comparison with the other approved TKIs, bosutinib harbors a distinct side effect profile with only very few cardiovascular and thromboembolic events and minimal long-term safety issues with most adverse events happening during the first months of treatment. On the other hand, gastrointestinal side effects are very common (e.g., diarrhea rates in more than 80% of the patients) with bosutinib surprising some of the investigators during the early clinical trials evaluating bosutinib. Until then, several approaches have been used to face this problem resulting in extensive supportive efforts (such as early loperamid treatment) as well as new trials testing alternative dosing strategies with early dose adjustment schedules. This article reports preclinical and clinical data available for bosutinib both in hematologic diseases such as CML or ALL and solid tumours as well as other diseases and envisions future perspectives including additional patient groups in which bosutinib might be of clinical benefit.
Collapse
Affiliation(s)
- Susanne Isfort
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Dominik Wolf
- Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127, Bonn, Germany
| |
Collapse
|
40
|
Tsao AS, Lin H, Carter BW, Lee JJ, Rice D, Vaporcyan A, Swisher S, Mehran R, Heymach J, Nilsson M, Fan Y, Nunez M, Diao L, Wang J, Fujimoto J, Wistuba II, Hong WK. Biomarker-Integrated Neoadjuvant Dasatinib Trial in Resectable Malignant Pleural Mesothelioma. J Thorac Oncol 2017; 13:246-257. [PMID: 29313814 DOI: 10.1016/j.jtho.2017.10.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Window of opportunity trials in malignant pleural mesothelioma (MPM) are challenging but can yield important translational information about a novel agent. METHODS We treated patients with MPM (N = 24) with 4 weeks of oral dasatinib followed by surgery with or without radiotherapy and then an optional 2 years of maintenance dasatinib. The primary end point was biomarker modulation of phosphorylated (p) SrcTyr419. RESULTS For all patients, the median progression-free survival (PFS) was 7.5 months and the median overall survival was 19.1 months. No significant responses were seen after 4 weeks of dasatinib therapy; however, modulation of median p-SrcTyr419 immunohistochemistry (IHC) scores was seen: the median pretreatment score was 70 (interquartile range 37.5-110), and the median posttreatment score was 41.9 (interquartile range 4.2-60) (p = 0.004). A decrease in p-SrcTyr419 levels after dasatinib correlated with improved median PFS (6.9 months versus 0.94 months [p = 0.03]), suggesting that p-SrcTyr419 is a viable pharmacodynamic biomarker for dasatinib in MPM. Platelet-derived growth factor receptor (PDGFR) pathway analysis correlated high PDGFR beta [PDGFRB) level (in the cytoplasm [hazard ratio] (HR) = 2.54, p = 0.05], stroma [HR = 2.79, p = 0.03], and nucleus [HR = 6.79, p = 0.023]) with a shorter PFS. Low (less than the median) cytoplasmic p-PDGFR alpha IHC levels were predictive of a decrease in positron emission tomography/computed tomography standard uptake values levels after dasatinib therapy (p = 0.04), whereas higher-than-median IHC scores of PDGFRB (cytoplasmic [HR = 2.8, p = 0.03] and nuclear [HR = 6.795, p = 0.02]) were correlated with rising standard uptake values levels. CONCLUSIONS In conclusion, there was no significant efficacy signal, and dasatinib monotherapy will not continue to be studied in MPM. However, our study demonstrated that PDGFR subtypes (platelet-derived growth factor receptor alpha and PDGFRB) may have differential roles in prognosis and resistance to antiangiogenic tyrosine kinase inhibitors and are important potential therapeutic targets that require further investigation.
Collapse
Affiliation(s)
- Anne S Tsao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas.
| | - Heather Lin
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Brett W Carter
- Department of Diagnostic Radiology Thoracic Imaging, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - J Jack Lee
- Department of Biostatistics, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - David Rice
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Ara Vaporcyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Steven Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Reza Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Monique Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Youhong Fan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Maria Nunez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Waun Ki Hong
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| |
Collapse
|
41
|
Lai YH, Lin SY, Wu YS, Chen HW, Chen JJW. AC-93253 iodide, a novel Src inhibitor, suppresses NSCLC progression by modulating multiple Src-related signaling pathways. J Hematol Oncol 2017; 10:172. [PMID: 29132432 PMCID: PMC5683468 DOI: 10.1186/s13045-017-0539-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023] Open
Abstract
Background The tyrosine kinase Src is involved in the progression of many cancers. Moreover, inhibiting Src activity has been shown to obstruct several signaling pathways regulated by the EGFR. Thus, Src is a valuable target molecule in drug development. The purpose of this study was to identify compounds that directly or indirectly modulate Src to suppress lung cancer cell growth and motility and to investigate the molecular mechanisms underlying the effects of these compounds. Methods Human non-small cell lung cancer (NSCLC) cell lines (PC9, PC9/gef, A549, and H1975) with different EGFR statuses were tested by cytotoxicity and proliferation assays after AC-93253 iodide treatment. Src and Src-related protein expression in AC-93253 iodide-treated PC9, PC9/gef, and A549 cells were assessed by western blotting. The effects of AC-93253 iodide on cancer cell colony formation, invasion, and migration were assessed in PC9 and PC9/gef cells. The synergistic effects of gefitinib and AC-93253 iodide were evaluated by combination index (CI)-isobologram analysis in gefitinib-resistant cell lines. The efficacy of AC-93253 iodide in vivo was determined using nude mice treated with either the compound or the vehicle. Results Among the compounds, AC-93253 iodide exhibited the most potent dose-independent inhibitory effects on the activity of Src as well as on that of the Src-related proteins EGFR, STAT3, and FAK. Furthermore, AC-93253 iodide significantly suppressed cancer cell proliferation, colony formation, invasion, and migration in vitro and tumor growth in vivo. AC-93253 iodide sensitized tumor cells to gefitinib treatment regardless of whether the cells were gefitinib-sensitive (PC9) or resistant (H1975 and PC9/gef), indicating that it may exert synergistic effects when used in combination with established therapeutic agents. Our findings also suggested that the inhibitory effects of AC-93253 iodide on lung cancer progression may be attributable to its ability to modulate multiple proteins, including Src, PI3K, JNK, Paxillin, p130cas, MEK, ERK, and EGFR. Conclusions Our data suggest that AC-93253 iodide inhibits NSCLC cell growth and motility by regulating multiple Src-related pathways. Our findings may facilitate the development of therapeutic strategies and anti-tumor drugs that may be useful for treating lung cancer in the future. Electronic supplementary material The online version of this article (10.1186/s13045-017-0539-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yi-Hua Lai
- Institute of Biomedical Sciences, National Chung Hsing University, No. 145, Xingda Rd., South Dist, Taichung, 40227, Taiwan, Republic of China
| | - Sih-Yin Lin
- Institute of Biomedical Sciences, National Chung Hsing University, No. 145, Xingda Rd., South Dist, Taichung, 40227, Taiwan, Republic of China
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, No. 145, Xingda Rd., South Dist, Taichung, 40227, Taiwan, Republic of China. .,Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan. .,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan. .,Department of Biotechnology, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
42
|
Zhong L, Yang J, Cao Z, Chen X, Hu Y, Li L, Yang S. Preclinical pharmacodynamic evaluation of drug candidate SKLB-178 in the treatment of non-small cell lung cancer. Oncotarget 2017; 8:12843-12854. [PMID: 28086226 PMCID: PMC5355060 DOI: 10.18632/oncotarget.14597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/23/2016] [Indexed: 02/05/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a serious life-threatening malignancy. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors, such as Gefitinib and Erlotinib, are effective clinical medicines for advanced NSCLC patients harboring EGFR-activating mutations. However, this therapy just benefits a small percentage of sufferers. Worse still, all patients treated with drugs ultimately develop resistance. Hence, there is still an unmet medical need among patients with NSCLC. In this account, we report a novel multikinase inhibitor SKLB-178, which potently inhibits both EGFR-activating and resistant mutations, as well as the activities of Src and VEGFR2 kinases. SKLB-178 potently inhibited cancer cell growth in both Gefitinib-sensitive and resistant NSCLC cells. Meanwhile, SKLB-178 significantly suppressed the migration, invasion and tube formation of endothelial cells, and the growth of intersegmental vessel in zebrafish. The in vivo pharmacodynamic studies further demonstrated that SKLB-178 had wider potency than Gefitinib, and could significantly prolong survival of animals in A549 experimental metastasis model. These advantages together with the low toxicity of SKLB-178 indicate that SKLB-178 deserves to be further developed as a potential drug candidate for NSCLC therapy.
Collapse
Affiliation(s)
- Lei Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center of Biotherapy, Sichuan 610041, China.,Personalized Drug Therapy Key Laboratory of Sichuan Province, Hospital of the University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Sichuan 610072, China
| | - Jiao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center of Biotherapy, Sichuan 610041, China
| | - Zhixing Cao
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Sichuan 611137, China
| | - Xin Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center of Biotherapy, Sichuan 610041, China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center of Biotherapy, Sichuan 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of pharmacy, Sichuan University, Sichuan 610041, China
| | - Shengyong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University/Collaborative Innovation Center of Biotherapy, Sichuan 610041, China
| |
Collapse
|
43
|
Watanabe S, Yoshida T, Kawakami H, Takegawa N, Tanizaki J, Hayashi H, Takeda M, Yonesaka K, Tsurutani J, Nakagawa K. T790M-Selective EGFR-TKI Combined with Dasatinib as an Optimal Strategy for Overcoming EGFR-TKI Resistance in T790M-Positive Non-Small Cell Lung Cancer. Mol Cancer Ther 2017; 16:2563-2571. [PMID: 28839001 DOI: 10.1158/1535-7163.mct-17-0351] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/02/2017] [Accepted: 08/09/2017] [Indexed: 11/16/2022]
Abstract
T790M mutation-selective EGFR tyrosine kinase inhibitors (EGFR-TKI) have demonstrated clinical benefits in non-small cell lung cancer (NSCLC) patients harboring T790M mutation, which is the major cause of resistance to EGFR-TKI. However, their efficacy is limited, possibly due to the emergence of apoptosis resistance in T790M-positive NSCLC. We previously identified Src family kinases as cooncogenic drivers along with T790M and found that the Src inhibitor dasatinib combined with an irreversible or a preclinical T790M-selective EGFR-TKI enhanced antitumor activity in T790M-positive cells. In the current study, we evaluated the efficacy of dasatinib combined with the clinically relevant T790M-selective EGFR-TKI ASP8273 or osimertinib in EGFR mutation-positive NSCLC with or without T790M mutation. A cell viability assay revealed that dasatinib had synergistic effects with these TKIs in T790M-positive cells and simultaneously inhibited Src, Akt, and Erk, which remained activated upon single-agent treatment. Dasatinib also increased the rate of apoptosis in T790M-positive cells induced by T790M-selective EGFR-TKIs, as determined by the Annexin-V binding assay; this was associated with downregulation of the antiapoptotic Bcl-2 family member Bcl-xL, a finding that was confirmed in mice bearing T790M-positive xenografts. Our results suggest that Bcl-xL plays a key role in the apoptosis resistance of T790M-positive NSCLC, and that dasatinib combined with clinically relevant T790M-selective EGFR-TKIs is potentially effective in overcoming resistance to first-generation EGFR-TKIs in NSCLC patients with acquired T790M. Mol Cancer Ther; 16(11); 2563-71. ©2017 AACR.
Collapse
Affiliation(s)
- Satomi Watanabe
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takeshi Yoshida
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan.
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan.
| | - Naoki Takegawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Junko Tanizaki
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Junji Tsurutani
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
44
|
Rhodomycin A, a novel Src-targeted compound, can suppress lung cancer cell progression via modulating Src-related pathways. Oncotarget 2016; 6:26252-65. [PMID: 26312766 PMCID: PMC4694899 DOI: 10.18632/oncotarget.4761] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 06/25/2015] [Indexed: 02/06/2023] Open
Abstract
Src activation is involved in cancer progression and the interplay with EGFR. Inhibition of Src activity also represses the signalling pathways regulated by EGFR. Therefore, Src has been considered a target molecule for drug development. This study aimed to identify the compounds that target Src to suppress lung cancer tumourigenesis and metastasis and investigate their underlying molecular mechanisms. Using a molecular docking approach and the National Cancer Institute (NCI) compound dataset, eight candidate compounds were selected, and we evaluated their efficacy. Among them, rhodomycin A was the most efficient at reducing the activity and expression of Src in a dose-dependent manner, which was also the case for Src-associated proteins, including EGFR, STAT3, and FAK. Furthermore, rhodomycin A significantly suppressed cancer cell proliferation, migration, invasion, and clonogenicity in vitro and tumour growth in vivo. In addition, rhodomycin A rendered gefitinib-resistant lung adenocarcinoma cells more sensitive to gefitinib treatment, implying a synergistic effect of the combination therapy. Our data also reveal that the inhibitory effect of rhodomycin A on lung cancer progression may act through suppressing the Src-related multiple signalling pathways, including PI3K, JNK, Paxillin, and p130cas. These findings will assist the development of anti-tumour drugs to treat lung cancer.
Collapse
|
45
|
Formisano L, D'Amato V, Servetto A, Brillante S, Raimondo L, Di Mauro C, Marciano R, Orsini RC, Cosconati S, Randazzo A, Parsons SJ, Montuori N, Veneziani BM, De Placido S, Rosa R, Bianco R. Src inhibitors act through different mechanisms in Non-Small Cell Lung Cancer models depending on EGFR and RAS mutational status. Oncotarget 2016; 6:26090-103. [PMID: 26325669 PMCID: PMC4694888 DOI: 10.18632/oncotarget.4636] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 07/23/2015] [Indexed: 11/25/2022] Open
Abstract
Resistance to the EGFR tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib, often related to Ras or secondary EGFR mutations, is a relevant clinical issue in Non-Small Cell Lung Cancer (NSCLC). Although Src TK has been involved in such resistance, clinical development of its inhibitors has been so far limited. To better define the molecular targets of the Src TKIs saracatinib, dasatinib and bosutinib, we used a variety of in vitro/in vivo studies. Kinase assays supported by docking analysis demonstrated that all the compounds directly inhibit EGFR TK variants. However, in live cells only saracatinib efficiently reduced EGFR activation, while dasatinib was the most effective agent in inhibiting Src TK. Consistently, a pronounced anti-proliferative effect was achieved with saracatinib, in EGFR mutant cells, or with dasatinib, in wt EGFR/Ras mutant cells, poorly dependent on EGFR and erlotinib-resistant. We then identified the most effective drug combinations to overcome resistance to EGFR inhibitors, both in vitro and in nude mice: in T790M EGFR erlotinib-resistant cells, saracatinib with the anti-EGFR mAb cetuximab; in Ras mutant erlotinib-resistant models, dasatinib with the MEK inhibitor selumetinib. Src inhibitors may act with different mechanisms in NSCLCs, depending on EGFR/Ras mutational profile, and may be integrated with EGFR or MEK inhibitors for different cohorts of NSCLCs.
Collapse
Affiliation(s)
- Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Valentina D'Amato
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Simona Brillante
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Lucia Raimondo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Concetta Di Mauro
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Marciano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Clara Orsini
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Antonio Randazzo
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Sarah J Parsons
- Department of Microbiology, Immunology & Cancer Biology, Cancer Center, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Nunzia Montuori
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberta Rosa
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
46
|
Stewart TA, Azimi I, Brooks AJ, Thompson EW, Roberts-Thomson SJ, Monteith GR. Janus kinases and Src family kinases in the regulation of EGF-induced vimentin expression in MDA-MB-468 breast cancer cells. Int J Biochem Cell Biol 2016; 76:64-74. [PMID: 27163529 DOI: 10.1016/j.biocel.2016.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/07/2016] [Accepted: 05/06/2016] [Indexed: 12/20/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is an important process associated with the metastasis of breast cancer cells. Members of the Janus kinases (JAKs) and Src family kinases (SFKs) are implicated in the regulation of an invasive phenotype in various cancer cell types. Using the pharmacological inhibitors JAK Inhibitor I (a pan-JAK inhibitor) and PP2 we investigated the role of the JAKs and SFKs, respectively, in the regulation of EMT markers in the MDA-MB-468 breast cancer cell line model of epidermal growth factor (EGF)-induced EMT. We identified selective inhibition of EGF induction of the mesenchymal marker vimentin by PP2 and JAK Inhibitor I. The effect of JAK Inhibitor I on vimentin protein induction occurred at a concentration lower than that required to significantly inhibit EGF-mediated signal transducer and activator of transcription 3 (STAT3)-phosphorylation, suggesting involvement of a STAT3-independent mechanism of EGF-induced vimentin regulation by JAKs. Despite our identification of a role for the JAK family in EGF-induced vimentin protein expression, siRNA-mediated silencing of each member of the JAK family was unable to phenocopy pharmacological inhibition, indicating potential redundancy among the JAK family members in this pathway. While SFKs and JAKs do not represent global regulators of the EMT phenotype, our findings have identified a role for members of these signaling pathways in the regulation of EGF-induced vimentin expression in the MDA-MB-468 breast cancer cell line.
Collapse
Affiliation(s)
- Teneale A Stewart
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Iman Azimi
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia; Mater Research, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew J Brooks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia; The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Erik W Thompson
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, Australia; Australia and Translational Research Institute, Brisbane, QLD, Australia
| | | | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia; Mater Research, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
47
|
Chen CH, Chang WH, Su KY, Ku WH, Chang GC, Hong QS, Hsiao YJ, Chen HC, Chen HY, Wu R, Yang PC, Chen JJW, Yu SL. HLJ1 is an endogenous Src inhibitor suppressing cancer progression through dual mechanisms. Oncogene 2016; 35:5674-5685. [DOI: 10.1038/onc.2016.106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 12/30/2015] [Accepted: 01/04/2016] [Indexed: 12/30/2022]
|
48
|
Zhang T, Cui G, Yao YL, Guo Y, Wang QC, Li XN, Feng WM. Inhibition of Nonsmall Cell Lung Cancer Cell Migration by Protein Arginine Methyltransferase 1-small Hairpin RNA Through Inhibiting Epithelial-mesenchymal Transition, Extracellular Matrix Degradation, and Src Phosphorylation In Vitro. Chin Med J (Engl) 2016; 128:1202-8. [PMID: 25947404 PMCID: PMC4831548 DOI: 10.4103/0366-6999.156126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Protein arginine methyltransferases 1 (PRMT1) is over-expressed in a variety of cancers, including lung cancer, and is correlated with a poor prognosis of tumor development. This study aimed to investigate the role of PRMT1 in nonsmall cell lung cancer (NSCLC) migration in vitro. Methods: In this study, PRMT1 expression in the NSCLC cell line A549 was silenced using lentiviral vector-mediated short hairpin RNAs. Cell migration was measured using both scratch wound healing and transwell cell migration assays. The mRNA expression levels of matrix metalloproteinase 2 (MMP-2) and tissue inhibitor of metalloproteinase 1, 2 (TIMP1, 2) were measured using quantitative real-time reverse transcription-polymerase chain reaction. The expression levels of protein markers for epithelial-mesenchymal transition (EMT) (E-cadherin, N-cadherin), focal adhesion kinase (FAK), Src, AKT, and their corresponding phosphorylated states were detected by Western blot. Results: Cell migration was significantly inhibited in the PRMT1 silenced group compared to the control group. The mRNA expression of MMP-2 decreased while TIMP1 and TIMP2 increased significantly. E-cadherin mRNA expression also increased while N-cadherin decreased. Only phosphorylated Src levels decreased in the silenced group while FAK or AKT remained unchanged. Conclusions: PRMT1-small hairpin RNA inhibits the migration abilities of NSCLC A549 cells by inhibiting EMT, extracellular matrix degradation, and Src phosphorylation in vitro.
Collapse
Affiliation(s)
| | - Ge Cui
- Department of Pathology, Research Center, First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang 313000, China
| | | | | | | | | | | |
Collapse
|
49
|
AKT inactivation causes persistent drug tolerance to EGFR inhibitors. Pharmacol Res 2015; 102:132-7. [PMID: 26453958 DOI: 10.1016/j.phrs.2015.09.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/31/2022]
Abstract
Drug resistance is a major obstacle to the success of EGFR-targeted therapy. We recently studied the mechanism by which a small subset of EGFR mutant lung cancer cells remains viable after EGFR inhibition. We found that this drug-tolerant subpopulation develops because EGFR inhibition prevents AKT activity and thus inactivates Ets-1 function. In this article, we discuss how changes in intrinsic cell signaling after EGFR inhibition open a new avenue to drug resistance in NSCLCs, and comment on combined TKI and MEK inhibitor treatment to reduce the probability of emergent resistance to EGFR TKIs.
Collapse
|
50
|
Zhao N, Liu Y, Chang Z, Li K, Zhang R, Zhou Y, Qiu F, Han X, Xu Y. Identification of Biomarker and Co-Regulatory Motifs in Lung Adenocarcinoma Based on Differential Interactions. PLoS One 2015; 10:e0139165. [PMID: 26402252 PMCID: PMC4581687 DOI: 10.1371/journal.pone.0139165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/08/2015] [Indexed: 01/01/2023] Open
Abstract
Changes in intermolecular interactions (differential interactions) may influence the progression of cancer. Specific genes and their regulatory networks may be more closely associated with cancer when taking their transcriptional and post-transcriptional levels and dynamic and static interactions into account simultaneously. In this paper, a differential interaction analysis was performed to detect lung adenocarcinoma-related genes. Furthermore, a miRNA-TF (transcription factor) synergistic regulation network was constructed to identify three kinds of co-regulated motifs, namely, triplet, crosstalk and joint. Not only were the known cancer-related miRNAs and TFs (let-7, miR-15a, miR-17, TP53, ETS1, and so on) were detected in the motifs, but also the miR-15, let-7 and miR-17 families showed a tendency to regulate the triplet, crosstalk and joint motifs, respectively. Moreover, several biological functions (i.e., cell cycle, signaling pathways and hemopoiesis) associated with the three motifs were found to be frequently targeted by the drugs for lung adenocarcinoma. Specifically, the two 4-node motifs (crosstalk and joint) based on co-expression and interaction had a closer relationship to lung adenocarcinoma, and so further research was performed on them. A 10-gene biomarker (UBC, SRC, SP1, MYC, STAT3, JUN, NR3C1, RB1, GRB2 and MAPK1) was selected from the joint motif, and a survival analysis indicated its significant association with survival. Among the ten genes, JUN, NR3C1 and GRB2 are our newly detected candidate lung adenocarcinoma-related genes. The genes, regulators and regulatory motifs detected in this work will provide potential drug targets and new strategies for individual therapy.
Collapse
Affiliation(s)
- Ning Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yongjing Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Zhiqiang Chang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Kening Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Rui Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuanshuai Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Fujun Qiu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xiaole Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
- * E-mail:
| |
Collapse
|