1
|
Liu MM, Chen XH, Lu XM, Wang FF, Wang C, Liu Y, Li PL, Du BT, Liang S, Gong PD, Wang YX. Variations in the Profiles of Vascular-Related Factors Among Different Sub-Types of Polycystic Ovarian Syndrome in Northern China. Front Endocrinol (Lausanne) 2021; 11:527592. [PMID: 33716949 PMCID: PMC7953058 DOI: 10.3389/fendo.2020.527592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 12/04/2020] [Indexed: 11/13/2022] Open
Abstract
Recently, a growing body of evidence has suggested that abnormal ovarian angiogenesis, secondary to the imbalance between various angiogenic markers, is involved in the pathogenesis of PCOS, and this has led to the use of various interventions (such as Diane-35) to restore the normal ovarian angiogenesis. Therefore, we conducted the current investigation to determine the role of such markers (endothelial growth factor (VEGF), endostatin (ES), and thrombospondin-1 (TSP-1)) in the pathogenesis of PCOS along with the associated changes in ovarian blood flow in patients with PCOS compared to healthy controls, both before and after a course of oral contraception. A total of 381 patients with PCOS and 98 healthy females of childbearing age were recruited from July 2014 to June 2017 at the Reproductive Center of the Second Affiliated Hospital of Harbin Medical University. The serum levels of VEGF, ES, and TSP-1 were determined by enzyme-linked immunosorbent assay, while ovarian perfusion was measured by the pulsatility index (PI) and resistance index (RI) by using transvaginal color Doppler ultrasound. Repeated analyses were carried out after 3 months of Diane-35 treatment. Post-treatment serum levels of luteinizing hormone (LH)/follicle stimulating hormone (FSH) ratio of patients with PCOS decreased significantly (P <0.05). The RI values of most PCOS patients increased after treatment (P<0.05), while PI was significantly increased in all patients (P<0.05). However, variable changes in the serum levels of TSP-1, VEGF, and ES after treatment were observed. Serum VEGF levels showed a negative correlation with serum LH/FSH ratio, T concentration, and ES (P <0.05), while ES levels were negatively correlated with serum T concentrations only (P<0.05). The markers of angiogenesis (VEGF, ES, and TSP-1) were expressed differently among PCOS patients, who also responded differently to the same course of Diane-35 treatment. This field still warrants further investigation to reach a more definitive conclusion.
Collapse
Affiliation(s)
- Mei-mei Liu
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Wang S, Zhou D, Xu Z, Song J, Qian X, Lv X, Luan J. Anti-tumor Drug Targets Analysis: Current Insight and Future Prospect. Curr Drug Targets 2020; 20:1180-1202. [PMID: 30947670 DOI: 10.2174/1389450120666190402145325] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/21/2019] [Accepted: 03/22/2019] [Indexed: 12/13/2022]
Abstract
The incidence and mortality of malignant tumors are on the rise, which has become the second leading cause of death in the world. At present, anti-tumor drugs are one of the most common methods for treating cancer. In recent years, with the in-depth study of tumor biology and related disciplines, it has been gradually discovered that the essence of cell carcinogenesis is the infinite proliferation of cells caused by the disorder of cell signal transduction pathways, followed by a major shift in the concept of anti-tumor drugs research and development. The focus of research and development is shifting from traditional cytotoxic drugs to a new generation of anti-tumor drugs targeted at abnormal signaling system targets in tumor cells. In this review, we summarize the targets of anti-tumor drugs and analyse the molecular mechanisms of their effects, which lay a foundation for subsequent treatment, research and development.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Dexi Zhou
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Jing Song
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xueyi Qian
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| | - Xiongwen Lv
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Institute for Liver Disease of Anhui Medical University, Hefei, Anhui Province, China
| | - Jiajie Luan
- Department of Pharmacy, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, China
| |
Collapse
|
3
|
Mapping Tyrosine Kinase Receptor Dimerization to Receptor Expression and Ligand Affinities. Processes (Basel) 2019. [DOI: 10.3390/pr7050288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Tyrosine kinase receptor (RTK) ligation and dimerization is a key mechanism for translating external cell stimuli into internal signaling events. This process is critical to several key cell and physiological processes, such as in angiogenesis and embryogenesis, among others. While modulating RTK activation is a promising therapeutic target, RTK signaling axes have been shown to involve complicated interactions between ligands and receptors both within and across different protein families. In angiogenesis, for example, several signaling protein families, including vascular endothelial growth factors and platelet-derived growth factors, exhibit significant cross-family interactions that can influence pathway activation. Computational approaches can provide key insight to detangle these signaling pathways but have been limited by the sparse knowledge of these cross-family interactions. Here, we present a framework for studying known and potential non-canonical interactions. We constructed generalized models of RTK ligation and dimerization for systems of two, three and four receptor types and different degrees of cross-family ligation. Across each model, we developed parameter-space maps that fully determine relative pathway activation for any set of ligand-receptor binding constants, ligand concentrations and receptor concentrations. Therefore, our generalized models serve as a powerful reference tool for predicting not only known ligand: Receptor axes but also how unknown interactions could alter signaling dimerization patterns. Accordingly, it will drive the exploration of cross-family interactions and help guide therapeutic developments across processes like cancer and cardiovascular diseases, which depend on RTK-mediated signaling.
Collapse
|
4
|
Design, synthesis and preclinical evaluation of 5-methyl-N4-aryl-furo[2,3-d]pyrimidines as single agents with combination chemotherapy potential. Bioorg Med Chem Lett 2018; 28:3085-3093. [DOI: 10.1016/j.bmcl.2018.07.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/19/2018] [Accepted: 07/26/2018] [Indexed: 01/16/2023]
|
5
|
Xu J, Xie L, Guo W. PDGF/PDGFR effects in osteosarcoma and the "add-on" strategy. Clin Sarcoma Res 2018; 8:15. [PMID: 30083310 PMCID: PMC6071404 DOI: 10.1186/s13569-018-0102-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/18/2018] [Indexed: 01/12/2023] Open
Abstract
New treatment options for advanced osteosarcoma have remained limited. The platelet-derived growth factor (PDGF)/platelet-derived growth factor receptor (PDGFR) pathway plays an important role in the development and metastasis of osteosarcoma, via either direct autocrine stimulation of tumor cells, or paracrine stimulation on tumor stromal cells. It promotes angiogenesis to overcome hypoxia in the tumor microenvironment, and modulates tumor interstitial fluid pressure to control the influx and efflux of other agents. Targeting the PDGF/PDGFR pathway is a promising therapeutic method to overcome drug resistance and improve patients' outcome in osteosarcoma. Further evidence is needed to define the detailed mechanism. Results from clinical trials using PDGF/PDGFR inhibitor as a single agent were disappointing, both in osteosarcoma and soft tissue sarcoma. However, when combined with other agents, named as "add-on" strategy, a synergistic antitumor effect has been confirmed in soft tissue sarcoma, and should be attempted in osteosarcoma.
Collapse
Affiliation(s)
- Jie Xu
- Peking University People's Hospital, Beijing, 100044 China
| | - Lu Xie
- Peking University People's Hospital, Beijing, 100044 China
| | - Wei Guo
- Peking University People's Hospital, Beijing, 100044 China
| |
Collapse
|
6
|
Di Pietro M, Pascuali N, Parborell F, Abramovich D. Ovarian angiogenesis in polycystic ovary syndrome. Reproduction 2018; 155:R199-R209. [PMID: 29386378 DOI: 10.1530/rep-17-0597] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine pathology among women in reproductive age. Its main symptoms are oligo or amenorrhea, hyperandrogenism and the presence of ovarian cysts. It is also associated with infertility, obesity and insulin resistance. Mainly due to its heterogeneity, PCOS treatments are directed to manage its symptoms and to prevent associated diseases. The correct formation and regression of blood vessels during each ovarian cycle is indispensable for proper follicular development, ovulation and corpus luteum formation. The importance of these processes opened a new and promising field: ovarian angiogenesis. Vascular alterations characterize numerous pathologies, either with increased, decreased or abnormal angiogenesis. In the last years, several anomalies of ovarian angiogenesis have been described in women with PCOS. Therefore, it has been suggested that these alterations may be associated with the decreased - or lack of - ovulation rates and for the formation of cysts in the PCOS ovaries. Restoration of a proper vessel formation in the ovaries may lead to improved follicular development and ovulation in these patients. In the present review, we attempt to summarize the alterations in ovarian angiogenesis that have been described in women with PCOS. We also discuss the therapeutic approaches aimed to correct these alterations and their beneficial effects on the treatment of infertility in PCOS.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| |
Collapse
|
7
|
Xie Y, Li C, Huang Y, Jia Z, Cao J. A novel multikinase inhibitor R8 exhibits potent inhibition on cancer cells through both apoptosis and autophagic cell death. Oncotarget 2017; 8:87209-87220. [PMID: 29152075 PMCID: PMC5675627 DOI: 10.18632/oncotarget.20257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/13/2017] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy is an important treatment for cancer patients, especially for those with unresectable lesions. Targeted therapy of cancer by specific inhibition of aberrant tyrosine kinase activities in cancer cells with chemically synthesized tyrosine kinase inhibitors (TKIs), shows better responses while less side effects than traditional chemotherapeutic drugs. It is common that cancer cells often exhibit deregulation of several tyrosine kinases simultaneously, multikinase TKIs (MKIs) therefore have greater advantages over single-target TKIs. Currently more MKIs are under developing for better efficacy for different types of cancer. In the present work, we evaluated the in vitro therapeutic potential of a novel MKI, namely R8, with comparison to the clinically available MKI Sunitinib. Results showed that R8 has stronger inhibition on six different types of cancer cell lines with lower IC50 than Sunitinib does. Cell cycle analysis showed that R8 induced significant G0/G1 arrest phase of lung cancer A549 and NCI-H226 cells. The inhibition was also confirmed by colony formation and migration assays in both lung cancer cell lines in a dose-dependent manner. R8 could significantly inhibit the phosphorylation of multiple receptor tyrosine kinases (RTKs) included PDGFRβ, VEGFR2, EGFR and C-Kit, leading to the down-regulation of PI3K-Akt-mTOR signaling. Further analysis revealed that R8 treatment induced more significant apoptosis than Sunitinib did, which might be the consequence of the autophagic cell death. In conclusion, this work suggested R8 to be a promising novel anticancer MKI, and provided the basis for further in vivo investigation on its potential in treatment of lung cancer.
Collapse
Affiliation(s)
- Yuqiong Xie
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chunchun Li
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yali Huang
- Institute of Hygiene, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Zhenyu Jia
- Institute of Occupational Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Jiang Cao
- Clinical Research Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
8
|
Pavana RK, Choudhary S, Bastian A, Ihnat MA, Bai R, Hamel E, Gangjee A. Discovery and preclinical evaluation of 7-benzyl-N-(substituted)-pyrrolo[3,2-d]pyrimidin-4-amines as single agents with microtubule targeting effects along with triple-acting angiokinase inhibition as antitumor agents. Bioorg Med Chem 2017; 25:545-556. [PMID: 27894589 PMCID: PMC5191990 DOI: 10.1016/j.bmc.2016.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 02/07/2023]
Abstract
The utility of cytostatic antiangiogenic agents (AA) in cancer chemotherapy lies in their combination with cytotoxic chemotherapeutic agents. Clinical combinations of AA with microtubule targeting agents (MTAs) have been particularly successful. The discovery, synthesis and biological evaluations of a series of 7-benzyl-N-substituted-pyrrolo[3,2-d]pyrimidin-4-amines are reported. Novel compounds which inhibit proangiogenic receptor tyrosine kinases (RTKs) including vascular endothelial growth factor receptor-2 (VEGFR-2), platelet-derived growth factor receptor-β (PDGFR-β) and epidermal growth factor receptor (EGFR), along with microtubule targeting in single molecules are described. These compounds also inhibited blood vessel formation in the chicken chorioallantoic membrane (CAM) assay, and some potently inhibited tubulin assembly (with activity comparable to that of combretastatin A-4 (CA)). In addition, some of the analogs circumvent the most clinically relevant tumor resistance mechanisms (P-glycoprotein and β-III tubulin expression) to microtubule targeting agents (MTA). These MTAs bind at the colchicine site on tubulin. Two analogs displayed two to three digit nanomolar GI50 values across the entire NCI 60 tumor cell panel and one of these, compound 7, freely water soluble as its HCl salt, afforded excellent in vivo antitumor activity against an orthotopic triple negative 4T1 breast cancer model and was superior to doxorubicin.
Collapse
Affiliation(s)
- Roheeth Kumar Pavana
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Shruti Choudhary
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States
| | - Anja Bastian
- Department of Physiology, University of Oklahoma College of Medicine, Oklahoma City, OK 73104, United States
| | - Michael A Ihnat
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Oklahoma City, OK 73117, United States
| | - Ruoli Bai
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Aleem Gangjee
- Division of Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA 15282, United States.
| |
Collapse
|
9
|
Platelet-derived growth factor receptor/platelet-derived growth factor (PDGFR/PDGF) system is a prognostic and treatment response biomarker with multifarious therapeutic targets in cancers. Tumour Biol 2016; 37:10053-66. [PMID: 27193823 DOI: 10.1007/s13277-016-5069-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/05/2016] [Indexed: 02/06/2023] Open
Abstract
Progress in cancer biology has led to an increasing discovery of oncogenic alterations of the platelet-derived growth factor receptors (PDGFRs) in cancers. In addition, their overexpression in numerous cancers invariably makes PDGFRs and platelet-derived growth factors (PDGFs) prognostic and treatment markers in some cancers. The oncologic alterations of the PDGFR/PDGF system affect the extracellular, transmembrane and tyrosine kinase domains as well as the juxtamembrane segment of the receptor. The receptor is also involved in fusions with intracellular proteins and receptor tyrosine kinase. These discoveries undoubtedly make the system an attractive oncologic therapeutic target. This review covers elementary biology of PDGFR/PDGF system and its role as a prognostic and treatment marker in cancers. In addition, the multifarious therapeutic targets of PDGFR/PDGF system are discussed. Great potential exists in the role of PDGFR/PDGF system as a prognostic and treatment marker and for further exploration of its multifarious therapeutic targets in safe and efficacious management of cancer treatments.
Collapse
|
10
|
Wang Y, Appiah-Kubi K, Wu M, Yao X, Qian H, Wu Y, Chen Y. The platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) are major players in oncogenesis, drug resistance, and attractive oncologic targets in cancer. Growth Factors 2016; 34:64-71. [PMID: 27170215 DOI: 10.1080/08977194.2016.1180293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) play a key role in signaling pathways in oncogenesis. The overexpression of PDGFs and PDGFRs and the oncogenic alterations of these receptors have been implicated in human cancers and correlated significantly with poor outcomes. This review discusses the biology of the PDGF isoforms and receptors briefly, and their role in oncogenesis. Also, the attractiveness of targeting PDGFs and PDGFRs, based on a wide display of oncologic alterations in cancers, diverse therapeutic strategies, their roles in resistance to cancer treatments with prospects of overcoming drug resistance, and the extent to which validated biomarkers have been developed for effective PDGFs and PDGFRs-based cancer management are discussed.
Collapse
Affiliation(s)
- Ying Wang
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Kwaku Appiah-Kubi
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
- b Department of Applied Biology , University for Development Studies , Navrongo , Ghana , and
| | - Min Wu
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Xiaoyuan Yao
- c Basic Medical Department, Changchun Medical College , Jilin , People's Republic of China
| | - Hai Qian
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Yan Wu
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| | - Yongchang Chen
- a Department of Physiology , School of Medicine, Jiangsu University , Jiangsu , People's Republic of China
| |
Collapse
|
11
|
Dadrich M, Nicolay NH, Flechsig P, Bickelhaupt S, Hoeltgen L, Roeder F, Hauser K, Tietz A, Jenne J, Lopez R, Roehrich M, Wirkner U, Lahn M, Huber PE. Combined inhibition of TGFβ and PDGF signaling attenuates radiation-induced pulmonary fibrosis. Oncoimmunology 2015; 5:e1123366. [PMID: 27467922 PMCID: PMC4910723 DOI: 10.1080/2162402x.2015.1123366] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/11/2015] [Accepted: 11/18/2015] [Indexed: 11/25/2022] Open
Abstract
Background: Radiotherapy (RT) is a mainstay for the treatment of lung cancer, but the effective dose is often limited by the development of radiation-induced pneumonitis and pulmonary fibrosis. Transforming growth factor β (TGFβ) and platelet-derived growth factor (PDGF) play crucial roles in the development of these diseases, but the effects of dual growth factor inhibition on pulmonary fibrosis development remain unclear. Methods: C57BL/6 mice were treated with 20 Gy to the thorax to induce pulmonary fibrosis. PDGF receptor inhibitors SU9518 and SU14816 (imatinib) and TGFβ receptor inhibitor galunisertib were applied individually or in combinations after RT. Lung density and septal fibrosis were measured by high-resolution CT and MRI. Lung histology and gene expression analyses were performed and Osteopontin levels were studied. Results: Treatment with SU9518, SU14816 or galunisertib individually attenuated radiation-induced pulmonary inflammation and fibrosis and decreased radiological and histological signs of lung damage. Combining PDGF and TGFβ inhibitors showed to be feasible and safe in a mouse model, and dual inhibition significantly attenuated radiation-induced lung damage and extended mouse survival compared to blockage of either pathway alone. Gene expression analysis of irradiated lung tissue showed upregulation of PDGF and TGFβ-dependent signaling components by thoracic irradiation, and upregulation patterns show crosstalk between downstream mediators of the PDGF and TGFβ pathways. Conclusion: Combined small-molecule inhibition of PDGF and TGFβ signaling is a safe and effective treatment for radiation-induced pulmonary inflammation and fibrosis in mice and may offer a novel approach for treatment of fibrotic lung diseases in humans. Translational statement: RT is an effective treatment modality for cancer with limitations due to acute and chronic toxicities, where TGFβ and PDGF play a key role. Here, we show that a combined inhibition of TGFβ and PDGF signaling is more effective in attenuating radiation-induced lung damage compared to blocking either pathway alone. We used the TGFβ-receptor I inhibitor galunisertib, an effective anticancer compound in preclinical models and the PDGFR inhibitors imatinib and SU9518, a sunitinib analog. Our signaling data suggest that the reduction of TGFβ and PDGF signaling and the attenuation of SPP1 (Osteopontin) expression may be responsible for the observed benefits. With the clinical availability of similar compounds currently in phase-I/II trials as cancer therapeutics or already approved for certain cancers or idiopathic lung fibrosis (IPF), our study suggests that the combined application of small molecule inhibitors of TGFβ and PDGF signaling may offer a promising approach to treat radiation-associated toxicity in RT of lung cancer.
Collapse
Affiliation(s)
- Monika Dadrich
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Paul Flechsig
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiology, University Hospital Center, 400 INF, Heidelberg, Germany
| | - Sebastian Bickelhaupt
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Line Hoeltgen
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Falk Roeder
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany; Department of Radiation Oncology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Kai Hauser
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Alexandra Tietz
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Jürgen Jenne
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Ramon Lopez
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Manuel Roehrich
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Ute Wirkner
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF , Heidelberg, Germany
| | - Michael Lahn
- Oncology Early Clinical Investigation, Lilly Research Laboratories, Indianapolis , IN, USA
| | - Peter E Huber
- Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), 280 INF, Heidelberg, Germany; Department of Radiation Oncology, University Hospital Center, 400 INF, Heidelberg, Germany
| |
Collapse
|
12
|
Fragment virtual screening based on Bayesian categorization for discovering novel VEGFR-2 scaffolds. Mol Divers 2015; 19:895-913. [DOI: 10.1007/s11030-015-9592-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/25/2015] [Indexed: 12/24/2022]
|
13
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
14
|
Johnson DK, Karanicolas J. Selectivity by small-molecule inhibitors of protein interactions can be driven by protein surface fluctuations. PLoS Comput Biol 2015; 11:e1004081. [PMID: 25706586 PMCID: PMC4338137 DOI: 10.1371/journal.pcbi.1004081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022] Open
Abstract
Small-molecules that inhibit interactions between specific pairs of proteins have long represented a promising avenue for therapeutic intervention in a variety of settings. Structural studies have shown that in many cases, the inhibitor-bound protein adopts a conformation that is distinct from its unbound and its protein-bound conformations. This plasticity of the protein surface presents a major challenge in predicting which members of a protein family will be inhibited by a given ligand. Here, we use biased simulations of Bcl-2-family proteins to generate ensembles of low-energy conformations that contain surface pockets suitable for small molecule binding. We find that the resulting conformational ensembles include surface pockets that mimic those observed in inhibitor-bound crystal structures. Next, we find that the ensembles generated using different members of this protein family are overlapping but distinct, and that the activity of a given compound against a particular family member (ligand selectivity) can be predicted from whether the corresponding ensemble samples a complementary surface pocket. Finally, we find that each ensemble includes certain surface pockets that are not shared by any other family member: while no inhibitors have yet been identified to take advantage of these pockets, we expect that chemical scaffolds complementing these “distinct” pockets will prove highly selective for their targets. The opportunity to achieve target selectivity within a protein family by exploiting differences in surface fluctuations represents a new paradigm that may facilitate design of family-selective small-molecule inhibitors of protein-protein interactions. Despite intense interest and considerable effort, there are few examples of small molecules that directly inhibit protein-protein interactions. Crystal structures of early successes have highlighted the plasticity of the protein surface, as some inhibitor-bound proteins are captured in conformations that are distinct from both their unbound and their protein-bound conformations. The lack of a single well-defined structure presents a challenge for predicting the members of a protein family to which a given compound will show activity (ligand selectivity). Here we generate ensembles of conformations from simulation, and show that we can predict ligand selectivity based on which family members sample conformations complementary to the ligand. This approach may present a new avenue for designing highly-selective inhibitors of protein-protein interactions.
Collapse
Affiliation(s)
- David K. Johnson
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, United States of America
| | - John Karanicolas
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, United States of America
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
15
|
Lu YY, Zhao CR, Wang RQ, Li WB, Qu XJ. A novel anticancer diarylurea derivative HL-40 as a multi-kinases inhibitor with good pharmacokinetics in Wistar rats. Biomed Pharmacother 2014; 69:255-9. [PMID: 25661367 DOI: 10.1016/j.biopha.2014.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 11/05/2014] [Indexed: 12/11/2022] Open
Abstract
HL-40, N-(4-(1-(4-chlorine indazole)) phenyl)-N-(4-chloro-3-three fluorine methyl phenyl) urea, is a novel diarylurea derivative. In this study, we investigated the kinases activities and binding constants, pharmacokinetics of HL-40, and then evaluated its anticancer efficacy by both in vitro and in vivo methods. Enzyme activities assays in vitro were employed to identify eight candidate kinase targets. The competition binding assays against eight candidate kinases suggested that HL-40 showed strong affinity to c-Kit, PDGFRβ and FLT3. The pharmacokinetic studies in Wistar rats showed that HL-40 could maintain high compound concentration and long residence time in the blood circulation. HL-40 possessed strong inhibition activities against 12 human cancer cells. Meanwhile, HL-40 effectively delayed the growth of cancer xenografts without significant toxicity to mice. Based on these in vitro and in vivo results, we suggested that HL-40 might be developed as a potential multi-kinases inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Yu-Yin Lu
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Cui-Rong Zhao
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Rui-Qi Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Wen-Bao Li
- School of Medicine and Pharmacy, Marine Drug R&D Department QMBRI, Ocean University of China, Qingdao 26603, China.
| | - Xian-Jun Qu
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| |
Collapse
|
16
|
Heldin CH. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal 2013; 11:97. [PMID: 24359404 PMCID: PMC3878225 DOI: 10.1186/1478-811x-11-97] [Citation(s) in RCA: 365] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 12/11/2013] [Indexed: 01/15/2023] Open
Abstract
Platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of certain cell types during embryonal development and e.g. tissue repair in the adult. Overactivity of PDGF receptor signaling, by overexpression or mutational events, may drive tumor cell growth. In addition, pericytes of the vasculature and fibroblasts and myofibroblasts of the stroma of solid tumors express PDGF receptors, and PDGF stimulation of such cells promotes tumorigenesis. Inhibition of PDGF receptor signaling has proven to useful for the treatment of patients with certain rare tumors. Whether treatment with PDGF/PDGF receptor antagonists will be beneficial for more common malignancies is the subject for ongoing studies.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for life laboratory, Uppsala University, Box 595SE-751 24 Uppsala, Sweden.
| |
Collapse
|
17
|
Juliachs M, Vidal A, Del Muro XG, Piulats JM, Condom E, Casanovas O, Graupera M, Germà JR, Villanueva A, Viñals F. Effectivity of pazopanib treatment in orthotopic models of human testicular germ cell tumors. BMC Cancer 2013; 13:382. [PMID: 23937707 PMCID: PMC3751347 DOI: 10.1186/1471-2407-13-382] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 08/02/2013] [Indexed: 01/23/2023] Open
Abstract
Background Cisplatin (CDDP) resistance in testicular germ cell tumors (GCTs) is still a clinical challenge, and one associated with poor prognosis. The purpose of this work was to test pazopanib, an anti-tumoral and anti-angiogenic multikinase inhibitor, and its combination with lapatinib (an anti-ErbB inhibitor) in mouse orthotopic models of human testicular GCTs. Methods We used two different models of human testicular GCTs orthotopically grown in nude mice; a CDDP-sensitive choriocarcinoma (TGT38) and a new orthotopic model generated from a metastatic GCT refractory to first-line CDDP chemotherapy (TGT44). Nude mice implanted with these orthotopic tumors were treated with the inhibitors and the effect on tumoral growth and angiogenesis was evaluated. Results TGT44 refractory tumor had an immunohistochemical profile similar to the original metastasis, with characteristics of yolk sac tumor. TGT44 did not respond when treated with cisplatin. In contrast, pazopanib had an anti-angiogenic effect and anti-tumor efficacy in this model. Pazopanib in combination with lapatinib in TGT38, an orthotopic model of choriocarcinoma had an additive effect blocking tumor growth. Conclusions We present pazopanib as a possible agent for the alternative treatment of CDDP-sensitive and CDDP-refractory GCT patients, alone or in combination with anti-ErbB therapies.
Collapse
Affiliation(s)
- Mercè Juliachs
- Laboratori de Recerca Translacional, Institut Català d'Oncologia, Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, 08908, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
King JW, Lee SM. Axitinib for the treatment of advanced non-small-cell lung cancer. Expert Opin Investig Drugs 2013; 22:765-73. [DOI: 10.1517/13543784.2013.775243] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
19
|
Abstract
BACKGROUND Activins control the growth of several tumour types including thoracic malignancies. In the present study, we investigated their expression and function in malignant pleural mesothelioma (MPM). METHODS The expression of activins and activin receptors was analysed by quantitative PCR in a panel of MPM cell lines. Activin A expression was further analysed by immunohistochemistry in MPM tissue specimens (N=53). Subsequently, MPM cells were treated with activin A, activin receptor inhibitors or activin-targeting siRNA and the impact on cell viability, proliferation, migration and signalling was assessed. RESULTS Concomitant expression of activin subunits and receptors was found in all cell lines, and activin A was overexpressed in most cell lines compared with non-malignant mesothelial cells. Similarly, immunohistochemistry demonstrated intense staining of tumour cells for activin A in a subset of patients. Treatment with activin A induced SMAD2 phosphorylation and stimulated clonogenic growth of mesothelioma cells. In contrast, treatment with kinase inhibitors of activin receptors (SB-431542, A-8301) inhibited MPM cell viability, clonogenicity and migration. Silencing of activin A expression by siRNA oligonucleotides further confirmed these results and led to reduced cyclin D1/3 expression. CONCLUSION Our study suggests that activin A contributes to the malignant phenotype of MPM cells via regulation of cyclin D and may represent a valuable candidate for therapeutic interference.
Collapse
|
20
|
[Antiangiogenic agents: current limits in thoracic oncology]. Bull Cancer 2012; 99:1083-91. [PMID: 23113985 DOI: 10.1684/bdc.2012.1655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Antiangiogenic agents appear as major therapeutic options in renal, colorectal and breast cancer. Their part in thoracic oncology is still limited today except for bevacizumab. We review the current limits of antiangiogenic agents in terms of efficacy, activity, tolerance and therapeutic strategies. Problems about predictive biomarkers and cost-effectiveness of antiangiogenic agents in thoracic oncology are also mentioned.
Collapse
|
21
|
Powers AD, Liu B, Lee AG, Palecek SP. Macroporous hydrogel micropillars for quantifying Met kinase activity in cancer cell lysates. Analyst 2012; 137:4052-61. [PMID: 22814332 PMCID: PMC3438145 DOI: 10.1039/c2an35464k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Overactive and overexpressed kinases have been implicated in the cause and progression of many cancers. Kinase inhibitors offer a targeted approach for treating cancers associated with increased or deregulated kinase activity. Often, however, cancer cells exhibit initial resistance to these inhibitors or evolve to develop resistance during treatment. Additionally, cancers of any one tissue type are typically heterogeneous in their oncogenesis mechanisms, and thus diagnosis of a particular type of cancer does not necessarily provide insight into what kinase therapies may be effective. For example, while some lung cancer cells that overexpress the epidermal growth factor receptor (EFGR) respond to treatment with EGFR kinase inhibitors, overexpression or hyperactivity of Met kinase correlates with resistance to EGFR kinase inhibitors. Here we describe a microfluidic-based assay for quantifying Met kinase activity in cancer cell lysates with the eventual goals of predicting cancer cell responsiveness to kinase inhibitors and monitoring development of resistance to these inhibitors. In this assay, we immobilized a phosphorylation substrate for Met kinase into macroporous hydrogel micropillars. We then exposed the micropillars to a cancer cell lysate and detected substrate phosphorylation using a fluorescently conjugated antibody. This assay is able to quantify Met kinase activity in whole cell lysate from as few as 150 cancer cells. It can also detect cells expressing overactive Met kinase in a background of up to 75% non-cancerous cells. Additionally, the assay can quantify kinase inhibition by the Met-specific kinase inhibitors SU11274 and PHA665752, suggesting predictive capability for cellular response to kinase inhibitors.
Collapse
Affiliation(s)
- Alicia D. Powers
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison
| | - Bi Liu
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison
| | - Andrew G. Lee
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison
| |
Collapse
|
22
|
Abramovich D, Irusta G, Bas D, Cataldi NI, Parborell F, Tesone M. Angiopoietins/TIE2 system and VEGF are involved in ovarian function in a DHEA rat model of polycystic ovary syndrome. Endocrinology 2012; 153:3446-56. [PMID: 22577112 DOI: 10.1210/en.2012-1105] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrinological pathology among women of reproductive age. It is characterized by anovulation, oligo- or amenorrhea, hyperandrogenism, obesity, and insulin resistance. PCOS patients present with elevated levels of vascular endothelial growth factor (VEGF) in serum and follicular fluid. In this study, we examined the ovarian expression of angiopoietins (ANGPT) and their receptor tyrosine kinase receptor (TIE2), involved in the stabilization of blood vessels, in a rat model of dehydroepiandrosterone-induced PCOS. We also analyzed the effect of ovarian VEGF inhibition on ANGPT/TIE2, follicular development, and vascular stability. VEGF levels were increased in the PCOS ovaries, whereas the levels of its receptor fetal liver kinase-1 were decreased. In addition, the periendothelial cell area and the ANGPT1 to ANGPT2 ratio in the ovary were increased in the PCOS group. Percentage of primary follicles was increased and the percentage of preantral follicles and corpora lutea was decreased in the PCOS group. VEGF inhibition decreased the percentage of primary follicles close to control values. Interestingly, despite the presence of cysts in the ovaries from VEGF inhibitor-treated PCOS rats, its percentage was lower than the PCOS group without treatment. In summary, this study describes an alteration not only in the VEGF/fetal liver kinase-1 system but also in the ANGPT/TIE2 system in a dehydroepiandrosterone-induced PCOS rat model. This leads to an increase in periendothelial cell recruitment. We also demonstrated that ovarian VEGF inhibition can partially restore the accumulation of small follicles in PCOS rats and reduces cyst formation, improving ovulation and follicular development. Therefore, the inhibition of VEGF could be considered, in addition to other currently applied treatments, as a new strategy to be studied in PCOS patients to restore ovarian function.
Collapse
Affiliation(s)
- Dalhia Abramovich
- Instituto de Biología y Medicina Experimental-Consejo Nacional de Investigaciones Cientificas y Técnicas, Universidad de Buenos Aires, C1428ADN, Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
23
|
El-Nikhely N, Larzabal L, Seeger W, Calvo A, Savai R. Tumor–stromal interactions in lung cancer: novel candidate targets for therapeutic intervention. Expert Opin Investig Drugs 2012; 21:1107-22. [DOI: 10.1517/13543784.2012.693478] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
24
|
Novel small molecule Raf kinase inhibitors for targeted cancer therapeutics. Arch Pharm Res 2012; 35:605-15. [PMID: 22553052 DOI: 10.1007/s12272-012-0403-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/14/2011] [Accepted: 12/14/2011] [Indexed: 12/13/2022]
Abstract
Aberrant activation of Raf signaling pathway is frequently found in various human tumors, it has been considered as distinct and promising molecular target for cancer therapeutics. B-Raf is most attractive drug target out of three Raf isoforms (A-Raf, B-Raf and C-Raf) because it exhibits high kinase activity due to frequent mutations in human tumors. However, most recently, it has been reported that Raf isoforms show the cross-activation in the presence of specific B-Raf inhibitors, which brings about the paradoxical p-ERK activation as well as tumor promoting effect. According to these findings, it remains controversy whether pan-Raf kinase inhibitor is more valuable and promising rather than specific B-Raf inhibitor under certain conditions in terms of cancer therapeutics. In this short review, novel Raf kinase inhibitors undergoing clinical investigation are introduced. Moreover, the paradoxical p-ERK activation is discussed with specific B-Raf inhibitors, PLX4032/4720 compounds.
Collapse
|