101
|
Yue P, Lopez-Tapia F, Paladino D, Li Y, Chen CH, Namanja AT, Hilliard T, Chen Y, Tius MA, Turkson J. Hydroxamic Acid and Benzoic Acid-Based STAT3 Inhibitors Suppress Human Glioma and Breast Cancer Phenotypes In Vitro and In Vivo. Cancer Res 2015; 76:652-63. [PMID: 26088127 DOI: 10.1158/0008-5472.can-14-3558] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/18/2015] [Indexed: 12/31/2022]
Abstract
STAT3 offers an attractive target for cancer therapy, but small-molecule inhibitors with appealing pharmacologic properties have been elusive. Here, we report hydroxamic acid-based and benzoic acid-based inhibitors (SH5-07 and SH4-54, respectively) with robust bioactivity. Both inhibitors blocked STAT3 DNA-binding activity in vitro and in human glioma, breast, and prostate cancer cells and in v-Src-transformed murine fibroblasts. STAT3-dependent gene transcription was blocked along with Bcl-2, Bcl-xL, Mcl-1, cyclin D1, c-Myc, and survivin expression. Nuclear magnetic resonance analysis of STAT3-inhibitor complexes defined interactions with the SH2 and DNA-binding domains of STAT3. Ectopic expression of the SH2 domain in cells was sufficient to counter the STAT3-inhibitory effects of SH4-54. Neither compound appreciably affected STAT1 or STAT5 DNA-binding activities, STAT3-independent gene transcription, or activation of a panel of oncogenic kinases in malignant cells. Each compound decreased the proliferation and viability of glioma, breast, and prostate cancer cells and v-Src-transformed murine fibroblasts harboring constitutively active STAT3. Further, in mouse xenograft models of glioma and breast cancer, administration of SH5-07 or SH4-54 effectively inhibited tumor growth. Our results offer preclinical proof of concept for SH5-07 and SH4-54 as candidates for further development as cancer therapeutics.
Collapse
Affiliation(s)
- Peibin Yue
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Francisco Lopez-Tapia
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Department of Chemistry, University of Hawaii, Manoa, Honolulu, Hawaii
| | - David Paladino
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Yifei Li
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Chih-Hong Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Andrew T Namanja
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Tyvette Hilliard
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii
| | - Yuan Chen
- Department of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Marcus A Tius
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Department of Chemistry, University of Hawaii, Manoa, Honolulu, Hawaii
| | - James Turkson
- Natural Products and Experimental Therapeutics, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii. Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii, Manoa, Honolulu, Hawaii.
| |
Collapse
|
102
|
Huang W, Dong Z, Chen Y, Wang F, Wang CJ, Peng H, He Y, Hangoc G, Pollok K, Sandusky G, Fu XY, Broxmeyer HE, Zhang ZY, Liu JY, Zhang JT. Small-molecule inhibitors targeting the DNA-binding domain of STAT3 suppress tumor growth, metastasis and STAT3 target gene expression in vivo. Oncogene 2015; 35:783-92. [PMID: 26073084 DOI: 10.1038/onc.2015.215] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/26/2015] [Accepted: 05/10/2015] [Indexed: 12/30/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in malignant tumors and has important roles in multiple aspects of cancer aggressiveness. Thus targeting STAT3 promises to be an attractive strategy for treatment of advanced metastatic tumors. Although many STAT3 inhibitors targeting the SH2 domain have been reported, few have moved into clinical trials. Targeting the DNA-binding domain (DBD) of STAT3, however, has been avoided due to its 'undruggable' nature and potentially limited selectivity. In a previous study, we reported an improved in silico approach targeting the DBD of STAT3 that resulted in a small-molecule STAT3 inhibitor (inS3-54). Further studies, however, showed that inS3-54 has off-target effect although it is selective to STAT3 over STAT1. In this study, we describe an extensive structure and activity-guided hit optimization and mechanistic characterization effort, which led to identification of an improved lead compound (inS3-54A18) with increased specificity and pharmacological properties. InS3-54A18 not only binds directly to the DBD and inhibits the DNA-binding activity of STAT3 both in vitro and in situ but also effectively inhibits the constitutive and interleukin-6-stimulated expression of STAT3 downstream target genes. InS3-54A18 is completely soluble in an oral formulation and effectively inhibits lung xenograft tumor growth and metastasis with little adverse effect on animals. Thus inS3-54A18 may serve as a potential candidate for further development as anticancer therapeutics targeting the DBD of human STAT3 and DBD of transcription factors may not be 'undruggable' as previously thought.
Collapse
Affiliation(s)
- W Huang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Z Dong
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Y Chen
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - F Wang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - C J Wang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H Peng
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Y He
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - G Hangoc
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - K Pollok
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - G Sandusky
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pathology and Molecular Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - X-Y Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Z-Y Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J-Y Liu
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Computer and Information Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - J-T Zhang
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
103
|
Blaylock RL. Cancer microenvironment, inflammation and cancer stem cells: A hypothesis for a paradigm change and new targets in cancer control. Surg Neurol Int 2015; 6:92. [PMID: 26097771 PMCID: PMC4455122 DOI: 10.4103/2152-7806.157890] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/03/2015] [Indexed: 12/13/2022] Open
Abstract
Since President Nixon officially declared a war on cancer with the National Cancer Act, billions of dollars have been spent on research in hopes of finding a cure for cancer. Recent reviews have pointed out that over the ensuing 42 years, cancer death rates have barely changed for the major cancers. Recently, several researchers have questioned the prevailing cancer paradigm based on recent discoveries concerning the mechanism of carcinogenesis and the origins of cancer. Over the past decade we have learned a great deal concerning both of these central issues. Cell signaling has taken center stage, particularly as regards the links between chronic inflammation and cancer development. It is now evident that the common factor among a great number of carcinogenic agents is activation of genes controlling inflammation cell-signaling pathways and that these signals control all aspects of the cancer process. Of these pathways, the most important and common to all cancers is the NFκB and STAT3 pathways. The second discovery of critical importance is that mutated stem cells appear to be in charge of the cancer process. Most chemotherapy agents and radiotherapy kill daughter cells of the cancer stem cell, many of which are not tumorigenic themselves. Most cancer stem cells are completely resistant to conventional treatments, which explain dormancy and the poor cure rate with metastatic tumors. A growing number of studies are finding that several polyphenol extracts can kill cancer stem cells as well as daughter cells and can enhance the effectiveness and safety of conventional treatments. These new discoveries provide the clinician with a whole new set of targets for cancer control and cure.
Collapse
Affiliation(s)
- Russell L. Blaylock
- Theoretical Neuroscience Research, LLC, Assistant Editor-in-Chief, Surgical Neurology International, 315 Rolling Meadows Rd, Ridgeland, MS 39157, USA
| |
Collapse
|
104
|
Zuo M, Li C, Lin J, Javle M. LLL12, a novel small inhibitor targeting STAT3 for hepatocellular carcinoma therapy. Oncotarget 2015; 6:10940-9. [PMID: 25883212 PMCID: PMC4484430 DOI: 10.18632/oncotarget.3458] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 02/24/2015] [Indexed: 01/19/2023] Open
Abstract
The constitutive activation of signal transducer and activator of transcription 3 (STAT3) is frequently detected in clinical incidences of hepatocellular carcinoma (HCC) but not in normal human hepatocytes. STAT3 signaling plays pivotal roles in angiogenesis, survival, metastasis, and growth of HCC. Recent evidence suggests that the blockade of aberrant STAT3 pathways can be exploited as a therapeutic strategy for HCC. We have developed the novel small molecular STAT3 inhibitor LLL12 on the basis of curcumin structure using computer-aided rational design. LLL12 has shown antitumor activity in various solid tumors including breast, brain, pancreatic cancer, and glioblastoma in vitro and in vivo. In this study, we hypothesized LLL12 inhibits STAT3 phosphorylation at tyrosine 705 (Y705) in HCC and show antitumor activity in HCC in vitro and in vivo. Our results show that LLL12 selectively inhibited HCC cell proliferation and induced apoptosis in SNU387, SNU398, SNU449, and Hep3B HCC cells in vitro. Furthermore, LLL12 at 5 mg/kg/day significantly inhibited the growth of SNU398 xenografts in nude mice. Collectively, our results indicate that LLL12 could be used to target STAT3 for the effective prevention or treatment of HCC.
Collapse
Affiliation(s)
- Mingxin Zuo
- 1 Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chenglong Li
- 2 Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Jiayuh Lin
- 3 Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43205, USA
| | - Milind Javle
- 1 Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
105
|
STAT3 inhibition reduces toxicity of oncolytic VSV and provides a potentially synergistic combination therapy for hepatocellular carcinoma. Cancer Gene Ther 2015; 22:317-25. [PMID: 25930184 DOI: 10.1038/cgt.2015.23] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 03/27/2015] [Accepted: 03/28/2015] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is a refractory malignancy with a high mortality and increasing worldwide incidence rates, including the United States and central Europe. In this study, we demonstrate that a specific inhibitor of signal transducer and activator of transcription 3 (STAT3), NSC74859, efficiently reduces HCC cell proliferation and can be successfully combined with oncolytic virotherapy using vesicular stomatitis virus (VSV). The potential benefits of this combination treatment are strengthened by the ability of NSC74859 to protect primary hepatocytes and nervous system cells against virus-induced cytotoxicity, with an elevation of the VSV maximum tolerated dose in mice. Hereby we propose a strategy for improving the current regimen for HCC treatment and seek to further explore the molecular mechanisms underlying selective oncolytic specificity of VSV.
Collapse
|
106
|
Ouédraogo ZG, Müller-Barthélémy M, Kemeny JL, Dedieu V, Biau J, Khalil T, Raoelfils LI, Granzotto A, Pereira B, Beaudoin C, Guissou IP, Berger M, Morel L, Chautard E, Verrelle P. STAT3 Serine 727 Phosphorylation: A Relevant Target to Radiosensitize Human Glioblastoma. Brain Pathol 2015; 26:18-30. [PMID: 25736961 DOI: 10.1111/bpa.12254] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/03/2015] [Indexed: 01/23/2023] Open
Abstract
Radiotherapy is an essential component of glioma standard treatment. Glioblastomas (GBM), however, display an important radioresistance leading to tumor recurrence. To improve patient prognosis, there is a need to radiosensitize GBM cells and to circumvent the mechanisms of resistance caused by interactions between tumor cells and their microenvironment. STAT3 has been identified as a therapeutic target in glioma because of its involvement in mechanisms sustaining tumor escape to both standard treatment and immune control. Here, we studied the role of STAT3 activation on tyrosine 705 (Y705) and serine 727 (S727) in glioma radioresistance. This study explored STAT3 phosphorylation on Y705 (pSTAT3-Y705) and S727 (pSTAT3-S727) in glioma cell lines and in clinical samples. Radiosensitizing effect of STAT3 activation down-modulation by Gö6976 was explored. In a panel of 15 human glioma cell lines, we found that the level of pSTAT3-S727 was correlated to intrinsic radioresistance. Moreover, treating GBM cells with Gö6976 resulted in a highly significant radiosensitization associated to a concomitant pSTAT3-S727 down-modulation only in GBM cell lines that exhibited no or weak pSTAT3-Y705. We report the constitutive activation of STAT3-S727 in all GBM clinical samples. Targeting pSTAT3-S727 mainly in pSTAT3-Y705-negative GBM could be a relevant approach to improve radiation therapy.
Collapse
Affiliation(s)
- Zangbéwendé Guy Ouédraogo
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France.,Laboratoire de Pharmacologie, de Toxicologie et de Chimie Thérapeutique, Université de Ouagadougou, 03 BP 7021, OUAGADOUGOU 03, BURKINA FASO
| | - Mélanie Müller-Barthélémy
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France
| | - Jean-Louis Kemeny
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,CHU Clermont-Ferrand, Service d'Anatomopathologie, F-63003, CLERMONT-FERRAND, France
| | - Véronique Dedieu
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France
| | - Julian Biau
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France.,Institut Curie, CNRS UMR3347, INSERM U2021, 91405, Orsay, France
| | - Toufic Khalil
- CHU Clermont-Ferrand, Service de Neurochirurgie, F-63003, CLERMONT-FERRAND, France.,Clermont Université, Université d'Auvergne, EA 7282, IGCNC, BP 10448, F-63000, CLERMONT-FERRAND, France
| | - Lala Ines Raoelfils
- Centre Jean Perrin, Service D'anatomopathologie, F-63000, CLERMONT-FERRAND, France
| | | | - Bruno Pereira
- CHU Clermont-Ferrand, Biostatistics unit, DRCI, F-63003, CLERMONT-FERRAND, France
| | - Claude Beaudoin
- Clermont Université, Université Blaise-Pascal, GReD, UMR CNRS 6293, INSERM U1103, 24 Avenue des Landais BP80026, 63171 Aubière 63177, AUBIERE, France
| | - Innocent Pierre Guissou
- Laboratoire de Pharmacologie, de Toxicologie et de Chimie Thérapeutique, Université de Ouagadougou, 03 BP 7021, OUAGADOUGOU 03, BURKINA FASO
| | - Marc Berger
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,CHU Clermont-Ferrand, Service d'Hématologie Biologique/Immunologie, F-63003, CLERMONT-FERRAND, France
| | - Laurent Morel
- Clermont Université, Université Blaise-Pascal, GReD, UMR CNRS 6293, INSERM U1103, 24 Avenue des Landais BP80026, 63171 Aubière 63177, AUBIERE, France
| | - Emmanuel Chautard
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France
| | - Pierre Verrelle
- Clermont Université, Université d'Auvergne, EA 7283, CREaT, BP 10448, F-63000, CLERMONT-FERRAND, France.,Centre Jean Perrin, Service Radiothérapie, Laboratoire de Radio-Oncologie Expérimentale, F-63000, CLERMONT-FERRAND, France
| |
Collapse
|
107
|
Wake MS, Watson CJ. STAT3 the oncogene - still eluding therapy? FEBS J 2015; 282:2600-11. [DOI: 10.1111/febs.13285] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/04/2015] [Accepted: 03/26/2015] [Indexed: 02/06/2023]
|
108
|
Zhou C, Jiao Y, Wang R, Ren SG, Wawrowsky K, Melmed S. STAT3 upregulation in pituitary somatotroph adenomas induces growth hormone hypersecretion. J Clin Invest 2015; 125:1692-702. [PMID: 25774503 DOI: 10.1172/jci78173] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 01/29/2015] [Indexed: 01/09/2023] Open
Abstract
Pituitary somatotroph adenomas result in dysregulated growth hormone (GH) hypersecretion and acromegaly; however, regulatory mechanisms that promote GH hypersecretion remain elusive. Here, we provide evidence that STAT3 directly induces somatotroph tumor cell GH. Evaluation of pituitary tumors revealed that STAT3 expression was enhanced in human GH-secreting adenomas compared with that in nonsecreting pituitary tumors. Moreover, STAT3 and GH expression were concordant in a somatotroph adenoma tissue array. Promoter and expression analysis in a GH-secreting rat cell line (GH3) revealed that STAT3 specifically binds the Gh promoter and induces transcription. Stable expression of STAT3 in GH3 cells induced expression of endogenous GH, and expression of a constitutively active STAT3 further enhanced GH production. Conversely, expression of dominant-negative STAT3 abrogated GH expression. In primary human somatotroph adenoma-derived cell cultures, STAT3 suppression with the specific inhibitor S3I-201 attenuated GH transcription and reduced GH secretion in the majority of derivative cultures. In addition, S3I-201 attenuated somatotroph tumor growth and GH secretion in a rat xenograft model. GH induced STAT3 phosphorylation and nuclear translocation, indicating a positive feedback loop between STAT3 and GH in somatotroph tumor cells. Together, these results indicate that adenoma GH hypersecretion is the result of STAT3-dependent GH induction, which in turn promotes STAT3 expression, and suggest STAT3 as a potential therapeutic target for pituitary somatotroph adenomas.
Collapse
|
109
|
Szelag M, Czerwoniec A, Wesoly J, Bluyssen HAR. Identification of STAT1 and STAT3 specific inhibitors using comparative virtual screening and docking validation. PLoS One 2015; 10:e0116688. [PMID: 25710482 PMCID: PMC4339377 DOI: 10.1371/journal.pone.0116688] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/15/2014] [Indexed: 12/31/2022] Open
Abstract
Signal transducers and activators of transcription (STATs) facilitate action of cytokines, growth factors and pathogens. STAT activation is mediated by a highly conserved SH2 domain, which interacts with phosphotyrosine motifs for specific STAT-receptor contacts and STAT dimerization. The active dimers induce gene transcription in the nucleus by binding to a specific DNA-response element in the promoter of target genes. Abnormal activation of STAT signaling pathways is implicated in many human diseases, like cancer, inflammation and auto-immunity. Searches for STAT-targeting compounds, exploring the phosphotyrosine (pTyr)-SH2 interaction site, yielded many small molecules for STAT3 but sparsely for other STATs. However, many of these inhibitors seem not STAT3-specific, thereby questioning the present modeling and selection strategies of SH2 domain-based STAT inhibitors. We generated new 3D structure models for all human (h)STATs and developed a comparative in silico docking strategy to obtain further insight into STAT-SH2 cross-binding specificity of a selection of previously identified STAT3 inhibitors. Indeed, by primarily targeting the highly conserved pTyr-SH2 binding pocket the majority of these compounds exhibited similar binding affinity and tendency scores for all STATs. By comparative screening of a natural product library we provided initial proof for the possibility to identify STAT1 as well as STAT3-specific inhibitors, introducing the ‘STAT-comparative binding affinity value’ and ‘ligand binding pose variation’ as selection criteria. In silico screening of a multi-million clean leads (CL) compound library for binding of all STATs, likewise identified potential specific inhibitors for STAT1 and STAT3 after docking validation. Based on comparative virtual screening and docking validation, we developed a novel STAT inhibitor screening tool that allows identification of specific STAT1 and STAT3 inhibitory compounds. This could increase our understanding of the functional role of these STATs in different diseases and benefit the clinical need for more drugable STAT inhibitors with high specificity, potency and excellent bioavailability.
Collapse
Affiliation(s)
- Malgorzata Szelag
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Anna Czerwoniec
- Bioinformatics Laboratory, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Hans A. R. Bluyssen
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
- * E-mail:
| |
Collapse
|
110
|
Dual-inhibitors of STAT5 and STAT3: studies from molecular docking and molecular dynamics simulations. J Mol Model 2014; 20:2399. [PMID: 25098340 DOI: 10.1007/s00894-014-2399-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 07/23/2014] [Indexed: 10/24/2022]
Abstract
Although molecularly targeted therapy with imatinib has improved treatments of chronic myeloid leukemia (CML), clinical resistance gradually develops in patients with accelerated or blast phase CML. The inability of imatinib to cure CML suggests that inactivation of BCR-ABL kinase activity alone is not sufficient to control the disease. Aberrant STAT signaling and constitutive STAT5 or STAT3 activation are frequently found in both acute and chronic leukemia. Constitutive activation of STAT5 and STAT3 are associated with imatinib resistance on leukemia cells. Development of drugs targeting SH2 domains of STAT5 and STAT3 provides a novel strategy for the treatment of the imatinib-resistant CML. Here, molecular docking and molecular dynamics simulations were used to investigate the interactions of the drugs targeting STAT3 and STAT5 receptors at molecular level. The calculated binding free energies are consistent with the ranking of the experimental affinities and our simulations also explained their differences in binding energy. Then virtual screening based on molecular docking and molecular dynamics was applied to screen a set of ~1500 compounds for dual inhibitors of the SH2 domains of STAT5 and STAT3. Three top score compounds obtained in virtual screening were compound 660, 304, and 561. Results show that the three predicted dual-inhibitors are well fitted within the two binding domains and are predicted to present improved STAT5 and STAT3 SH2 inhibitory activity.
Collapse
|
111
|
STAT3 inhibitor NSC74859 radiosensitizes esophageal cancer via the downregulation of HIF-1α. Tumour Biol 2014; 35:9793-9. [PMID: 24981247 DOI: 10.1007/s13277-014-2207-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
Radiotherapy is the main therapy for inoperable and locally advanced esophageal squamous cell carcinoma (ESCC). However, radioresistance in ESCC remains a challenge. The aim of this study is to investigate the radiosensitizing effects of STAT3 inhibitor NSC74859 on ESCC and explore the underlying mechanisms. ECA109 and TE13 cells were exposed to hypoxia, and treated with NSC74859 or radiation, alone or in combination. Cell proliferation, survival, apoptosis, and double-stranded DNA breaks (DSBs) were examined. Nude mice model of ECA109 xenograft was treated with radiation and/or NSC74859. The levels of STAT3, p-STAT3, HIF-1α, and VEGF were detected by Western blot analysis. NSC74859 efficiently radiosensitized ESCC cells and xenografts in nude mice, and inhibited hypoxia-/radiation-induced activation of STAT3 and upregulation of HIF-1α and VEGF expression. NSC74859 confers radiosensitivity in ESCC via the inhibition of STAT3 activation and the downregulation of HIF-1α and VEGF expression. NSC74859 may become a promising radiosensitizer for ESCC radiotherapy.
Collapse
|
112
|
Szeląg M, Czerwoniec A, Wesoly J, Bluyssen HAR. Comparative screening and validation as a novel tool to identify STAT-specific inhibitors. Eur J Pharmacol 2014; 740:417-20. [PMID: 25183399 DOI: 10.1016/j.ejphar.2014.05.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 05/16/2014] [Accepted: 05/20/2014] [Indexed: 01/04/2023]
Abstract
Signal transducers and activators of transcription (STATs) facilitate action of cytokines, growth factors and pathogens. STAT activation is mediated by a highly conserved SH2 domain, which interacts with phosphotyrosine (pTyr) motifs for specific STAT-receptor contacts and STAT dimerization. The active dimers induce gene transcription in the nucleus by binding to specific DNA-response elements of target genes. Abnormal activation of STAT signaling pathways is implicated in many human diseases, like cancer, inflammation and auto-immunity. STAT inhibitory strategies mostly focus on inhibiting STAT dimerization using small molecules identified by molecular modeling, virtual or library screening, or natural products. Searches for STAT-targeting compounds, exploring the pTyr-SH2 interaction area, yielded many small molecules for STAT3 but sparsely for other STATs. So far, no STAT-targeting drug is approved by the FDA. Moreover, many of these inhibitors do not seem STAT-specific, thereby questioning the present selection strategies of SH2 domain-based STAT inhibitors. This illustrates the need for better models, and screening and validation tools for more druggable STAT inhibitors with high specificity, potency and excellent bioavailability. Based on newly developed 3D structure models for all human (h)STATs, we propose a pipeline approach that combines comparative in silico docking of STAT-SH2 models with an in vitro STAT phosphorylation assay, as a novel tool to screen multi-million compound libraries and identify specific inhibitors for different STATs. Identification of specific and effective STAT inhibitory compounds could provide a tool to increase our understanding of their functional role in different diseases, and serve as therapeutic strategies in cancer, inflammation and auto-immunity.
Collapse
Affiliation(s)
- Małgorzata Szeląg
- Department of Human Molecular Genetics, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Anna Czerwoniec
- Bioinformatics Laboratory, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Joanna Wesoly
- Laboratory of High Throughput Technologies, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland
| | - Hans A R Bluyssen
- Department of Human Molecular Genetics, Adam Mickiewicz University in Poznan, Umultowska 89, 61-614 Poznan, Poland.
| |
Collapse
|
113
|
Lin GS, Chen YP, Lin ZX, Wang XF, Zheng ZQ, Chen L. STAT3 serine 727 phosphorylation influences clinical outcome in glioblastoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:3141-3149. [PMID: 25031733 PMCID: PMC4097241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/26/2014] [Indexed: 06/03/2023]
Abstract
Besides STAT3 tyrosine 705 phosphorylation (pTyr705-STAT3), phosphorylation of STAT3 at serine 727 (pSer727-STAT3) is shown to contribute to tumorigenesis and be closely related with resistance to radiotherapy and chemotherapy in glioma, but there is currently no study regarding its relevance to prognosis in glioblastoma (GBM). Here, the expression of phosphorylated STAT3 was detected in tumor specimens from 88 patients with newly diagnosed GBM by immunohistochemistry, the Kaplan-Meier survival curve and COX proportional hazards regression model were applied to estimate its influences on progression-free survival (PFS) and overall survival (OS). Immunohistochemical assay showed elevated expression of pSer727-STAT3 in GBM compared with normal brain tissue. Univariate analysis indicated significant correlations of high percentage of pSer727-STAT3 positive tumor cells with shorter PFS (P = 0.006) and OS (P = 0.002). In multivariate analysis, high pSer727-STAT3 expression was demonstrated as an independent unfavorable prognostic indicator for PFS (HR 1.830, P = 0.022) and OS (HR 1.797, P = 0.040). And patients with high expression of both pTyr705-STAT3 and pSer727-STAT3 had a poorer prognosis compared with the remainder (P < 0.005). In conclusion, the high proportion of pSer727-STAT3 positive neoplastic cells in GBM is an independent unfavorable prognostic factor, and increased expression of both pTyr705-STAT3 and pSer727-STAT3 is predictive of poorer clinical outcome, thereby adding to the growing evidence that STAT3 inhibition may be a potential therapeutic strategy in glioblastoma.
Collapse
Affiliation(s)
- Guo-Shi Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical UniversityFuzhou, Fujian, China
| | - Yu-Peng Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou, Fujian, China
| | - Zhi-Xiong Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical UniversityFuzhou, Fujian, China
| | - Xing-Fu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical UniversityFuzhou, Fujian, China
| | - Zong-Qing Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical UniversityFuzhou, Fujian, China
| | - Long Chen
- Tumor Invasion Microecological Laboratory, Fujian Medical UniversityFuzhou, Fujian, China
| |
Collapse
|
114
|
The Multifaceted Roles of STAT3 Signaling in the Progression of Prostate Cancer. Cancers (Basel) 2014; 6:829-59. [PMID: 24722453 PMCID: PMC4074806 DOI: 10.3390/cancers6020829] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 03/11/2014] [Accepted: 03/17/2014] [Indexed: 01/09/2023] Open
Abstract
The signal transducer and activator of transcription (STAT)3 governs essential functions of epithelial and hematopoietic cells that are often dysregulated in cancer. While the role for STAT3 in promoting the progression of many solid and hematopoietic malignancies is well established, this review will focus on the importance of STAT3 in prostate cancer progression to the incurable metastatic castration-resistant prostate cancer (mCRPC). Indeed, STAT3 integrates different signaling pathways involved in the reactivation of androgen receptor pathway, stem like cells and the epithelial to mesenchymal transition that drive progression to mCRPC. As equally important, STAT3 regulates interactions between tumor cells and the microenvironment as well as immune cell activation. This makes it a major factor in facilitating prostate cancer escape from detection of the immune response, promoting an immunosuppressive environment that allows growth and metastasis. Based on the multifaceted nature of STAT3 signaling in the progression to mCRPC, the promise of STAT3 as a therapeutic target to prevent prostate cancer progression and the variety of STAT3 inhibitors used in cancer therapies is discussed.
Collapse
|
115
|
Lin GS, Yang LJ, Wang XF, Chen YP, Tang WL, Chen L, Lin ZX. STAT3 Tyr705 phosphorylation affects clinical outcome in patients with newly diagnosed supratentorial glioblastoma. Med Oncol 2014; 31:924. [DOI: 10.1007/s12032-014-0924-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/11/2014] [Indexed: 12/22/2022]
|
116
|
Li H, Xiao H, Lin L, Jou D, Kumari V, Lin J, Li C. Drug Design Targeting Protein–Protein Interactions (PPIs) Using Multiple Ligand Simultaneous Docking (MLSD) and Drug Repositioning: Discovery of Raloxifene and Bazedoxifene as Novel Inhibitors of IL-6/GP130 Interface. J Med Chem 2014; 57:632-41. [DOI: 10.1021/jm401144z] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Huameng Li
- Biophysics
Graduate Program, The Ohio State University, Columbus, Ohio 43210, United States
- Division
of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Hui Xiao
- Center
for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio 43205, United States
| | - Li Lin
- Division
of Cardiology, Department of Internal Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - David Jou
- Center
for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio 43205, United States
| | - Vandana Kumari
- Division
of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jiayuh Lin
- Center
for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio 43205, United States
| | - Chenglong Li
- Biophysics
Graduate Program, The Ohio State University, Columbus, Ohio 43210, United States
- Division
of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
117
|
Lavecchia A, Di Giovanni C, Cerchia C. Novel inhibitors of signal transducer and activator of transcription 3 signaling pathway: an update on the recent patent literature. Expert Opin Ther Pat 2014; 24:383-400. [PMID: 24432979 DOI: 10.1517/13543776.2014.877443] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The signal transducer and activator of transcription 3 (STAT3) is a transcription factor that plays a key role in normal cell growth and is constitutively activated in about 70% of solid and hematological cancers. Thus, the development of potent and selective inhibitors that target STAT3 is of interest especially in the cancer therapeutic area. AREAS COVERED This review updates new patents claiming STAT3 inhibitors and their uses published from 2011 to 2013. Pre-2011 patents have been extensively covered in previous reviews. Comments on the context of each chemical series are given where applicable to orientate the readers on the bewildering array of molecular designs now available. EXPERT OPINION The growing number of preclinical studies in numerous malignances as well as the first clinical trials of STAT3 inhibitors suggest that STAT3 remains a valid target for the treatment of human cancers as well as inflammatory diseases and/or autoimmune disorders. So, the future looks bright for patients because many new drugs are being developed and now combinations of STAT3 inhibitors with other targeted agents can diminish the resistance to traditional chemotherapy. These advances are expected to lead to further significant progress improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Antonio Lavecchia
- University of Naples Federico II, Department of Pharmacy, "Drug Discovery" Laboratory , Via D. Montesano 49, 80131 Naples , Italy +39 081 678613 ; +39 081 678012 ;
| | | | | |
Collapse
|
118
|
Gurbuz V, Konac E, Varol N, Yilmaz A, Gurocak S, Menevse S, Sozen S. Effects of AG490 and S3I-201 on regulation of the JAK/STAT3 signaling pathway in relation to angiogenesis in TRAIL-resistant prostate cancer cells in vitro.. Oncol Lett 2014; 7:755-763. [PMID: 24520293 PMCID: PMC3919920 DOI: 10.3892/ol.2014.1795] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/20/2013] [Indexed: 01/14/2023] Open
Abstract
The aim of the present study was to analyze the molecular mechanisms involved in blocking the signaling pathway and the effects of this on the progression of prostate cancer (CaP) cells in vitro. LNCaP human CaP cell line was stimulated with interleukin-6 (IL-6) in the presence/absence of Janus kinase (JAK) 2 (AG490), signal transducer and activator of transcription 3 [(STAT3) S3I-201] inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Cytotoxic activity, the activation of phosphorylated (p)-STAT3 protein, caspase (CASP) 3 activity at protein level, vascular endothelial growth factor (VEGF) A, VEGFC, vascular endothelial growth factor receptor 2, STAT3, matrix metalloproteinase-2, myeloid cell leukemia sequence 1 (MCL-1), CASP8 and CASP9 messenger RNA (mRNA) levels were determined. Morphology and apoptosis were confirmed by DAPI staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. IL-6 rapidly induced the phosphorylation of STAT3 in a dose- and time-dependent manner with a peak expression at 3 h at a concentration of 25 ng/ml. In addition, AG490 (50 μM) and S3I-201 (300 μM) inhibited STAT3 activation. Western blotting results revealed that p-STAT3 protein expression decreased significantly with AG490 and S3I-201 treatment in LNCaP cells. AG490 and S3I-201 induced the downregulation of VEGFA, MCL-1 and STAT3 and the upregulation of CASP8 and CASP9 mRNA transcription levels. In addition, the inhibitors increased the level of CASP3 protein. Combinations of AG490- and S3I-201-TRAIL did not result in an increase in this effect. Parallel results were found by DAPI staining and TUNEL assay. To the best of our knowledge, this is the first study to investigate the possible clinical use of AG490 or S3I-201, together with the reduced use of chemotherapeutic agents with high cytotoxicity, for their ability to exert an apoptotic effect, targeting the JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Venhar Gurbuz
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey ; Department of Pediatric Infectious Diseases, Faculty of Medicine, Hacettepe University Ankara, Ankara 06100, Turkey
| | - Ece Konac
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| | - Nuray Varol
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| | - Akin Yilmaz
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| | - Serhat Gurocak
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey ; Department of Urology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| | - Sevda Menevse
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| | - Sinan Sozen
- Department of Urology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey
| |
Collapse
|
119
|
Yu W, Li C. Regioselective one-pot C–N coupling of substituted naphthoquinones: selective intramolecular ring fusion of sulfonamides. Tetrahedron 2014. [DOI: 10.1016/j.tet.2013.11.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
120
|
Lin L, Hutzen B, Lee HF, Peng Z, Wang W, Zhao C, Lin HJ, Sun D, Li PK, Li C, Korkaya H, Wicha MS, Lin J. Evaluation of STAT3 signaling in ALDH+ and ALDH+/CD44+/CD24- subpopulations of breast cancer cells. PLoS One 2013; 8:e82821. [PMID: 24376586 PMCID: PMC3871589 DOI: 10.1371/journal.pone.0082821] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 11/06/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND STAT3 activation is frequently detected in breast cancer and this pathway has emerged as an attractive molecular target for cancer treatment. Recent experimental evidence suggests ALDH-positive (ALDH(+)), or cell surface molecule CD44-positive (CD44(+)) but CD24-negative (CD24(-)) breast cancer cells have cancer stem cell properties. However, the role of STAT3 signaling in ALDH(+) and ALDH(+)/CD44(+)/CD24(-) subpopulations of breast cancer cells is unknown. METHODS AND RESULTS We examined STAT3 activation in ALDH(+) and ALDH(+)/CD44(+)/CD24(-) subpopulations of breast cancer cells by sorting with flow cytometer. We observed ALDH-positive (ALDH(+)) cells expressed higher levels of phosphorylated STAT3 compared to ALDH-negative (ALDH(-)) cells. There was a significant correlation between the nuclear staining of phosphorylated STAT3 and the expression of ALDH1 in breast cancer tissues. These results suggest that STAT3 is activated in ALDH(+) subpopulations of breast cancer cells. STAT3 inhibitors Stattic and LLL12 inhibited STAT3 phosphorylation, reduced the ALDH(+) subpopulation, inhibited breast cancer stem-like cell viability, and retarded tumorisphere-forming capacity in vitro. Similar inhibition of STAT3 phosphorylation, and breast cancer stem cell viability were observed using STAT3 ShRNA. In addition, LLL12 inhibited STAT3 downstream target gene expression and induced apoptosis in ALDH(+) subpopulations of breast cancer cells. Furthermore, LLL12 inhibited STAT3 phosphorylation and tumor cell proliferation, induced apoptosis, and suppressed tumor growth in xenograft and mammary fat pad mouse models from ALDH(+) breast cancer cells. Similar in vitro and tumor growth in vivo results were obtained when ALDH(+) cells were further selected for the stem cell markers CD44(+) and CD24(-). CONCLUSION These studies demonstrate an important role for STAT3 signaling in ALDH(+) and ALDH(+)/CD44(+)/CD24(-) subpopulations of breast cancer cells which may have cancer stem cell properties and suggest that pharmacologic inhibition of STAT3 represents an effective strategy to selectively target the cancer stem cell-like subpopulation.
Collapse
Affiliation(s)
- Li Lin
- Divison of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, Columbus, Ohio, United States of America
- * E-mail: (JL); (LL)
| | - Brian Hutzen
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Hsiu-Fang Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zhengang Peng
- Medical Technology Division, School of Allied Medical Professions, Columbus, Ohio, United States of America
| | - Wenlong Wang
- Divison of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chongqiang Zhao
- Divison of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Huey-Jen Lin
- Medical Technology Division, School of Allied Medical Professions, Columbus, Ohio, United States of America
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Delaware, Newark, Delaware, United States of America
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Hasan Korkaya
- Department of Internal Medicine, The University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, United States of America
| | - Max S. Wicha
- Department of Internal Medicine, The University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, United States of America
| | - Jiayuh Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail: (JL); (LL)
| |
Collapse
|
121
|
Furqan M, Akinleye A, Mukhi N, Mittal V, Chen Y, Liu D. STAT inhibitors for cancer therapy. J Hematol Oncol 2013; 6:90. [PMID: 24308725 PMCID: PMC4029528 DOI: 10.1186/1756-8722-6-90] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/02/2013] [Indexed: 12/24/2022] Open
Abstract
Signal Transducer and Activator of Transcription (STAT) proteins are a family of cytoplasmic transcription factors consisting of 7 members, STAT1 to STAT6, including STAT5a and STAT5b. STAT proteins are thought to be ideal targets for anti-cancer therapy since cancer cells are more dependent on the STAT activity than their normal counterparts. Inhibitors targeting STAT3 and STAT5 have been developed. These included peptidomimetics, small molecule inhibitors and oligonucleotides. This review summarized advances in preclinical and clinical development of these compounds.
Collapse
Affiliation(s)
- Muhammad Furqan
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
| | - Akintunde Akinleye
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
| | - Nikhil Mukhi
- Department of Medicine, SUNY Downstate Medical Center Brooklyn, Brooklyn, NY 11203, USA
| | - Varun Mittal
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
| | - Yamei Chen
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
- Department of Hematology, Xiamen Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Delong Liu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
- Division of Hematology and Oncology, Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY 10595, USA
| |
Collapse
|
122
|
Bhasin D, Etter JP, Chettiar SN, Mok M, Li PK. Antiproliferative activities and SAR studies of substituted anthraquinones and 1,4-naphthoquinones. Bioorg Med Chem Lett 2013; 23:6864-7. [DOI: 10.1016/j.bmcl.2013.09.098] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/25/2013] [Accepted: 09/30/2013] [Indexed: 01/14/2023]
|
123
|
Nguyen HB, Hung LW, Yeates TO, Terwilliger TC, Waldo GS. Split green fluorescent protein as a modular binding partner for protein crystallization. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2013; 69:2513-23. [PMID: 24311592 PMCID: PMC3852656 DOI: 10.1107/s0907444913024608] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/03/2013] [Indexed: 02/08/2023]
Abstract
A modular strategy for protein crystallization using split green fluorescent protein (GFP) as a crystallization partner is demonstrated. Insertion of a hairpin containing GFP β-strands 10 and 11 into a surface loop of a target protein provides two chain crossings between the target and the reconstituted GFP compared with the single connection afforded by terminal GFP fusions. This strategy was tested by inserting this hairpin into a loop of another fluorescent protein, sfCherry. The crystal structure of the sfCherry-GFP(10-11) hairpin in complex with GFP(1-9) was determined at a resolution of 2.6 Å. Analysis of the complex shows that the reconstituted GFP is attached to the target protein (sfCherry) in a structurally ordered way. This work opens the way to rapidly creating crystallization variants by reconstituting a target protein bearing the GFP(10-11) hairpin with a variety of GFP(1-9) mutants engineered for favorable crystallization.
Collapse
Affiliation(s)
- Hau B. Nguyen
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| | - Li-Wei Hung
- Physics Division, Los Alamos National Laboratory, MS D454, Los Alamos, NM 87545, USA
| | - Todd O. Yeates
- Department of Chemistry and Biochemistry, University of California, PO Box 951569, Los Angeles, CA 90095, USA
| | - Thomas C. Terwilliger
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| | - Geoffrey S. Waldo
- Bioscience Division, Los Alamos National Laboratory, MS M888, Los Alamos, NM 87545, USA
| |
Collapse
|
124
|
Bid HK, Kibler A, Phelps DA, Manap S, Xiao L, Lin J, Capper D, Oswald D, Geier B, DeWire M, Smith PD, Kurmasheva RT, Mo X, Fernandez S, Houghton PJ. Development, characterization, and reversal of acquired resistance to the MEK1 inhibitor selumetinib (AZD6244) in an in vivo model of childhood astrocytoma. Clin Cancer Res 2013; 19:6716-29. [PMID: 24132923 DOI: 10.1158/1078-0432.ccr-13-0842] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE The BT-40 low-grade childhood astrocytoma xenograft model expresses mutated BRAF(V600E) and is highly sensitive to the MEK inhibitor selumetinib (AZD6244). In this study, we developed and characterized selumetinib resistance and explored approaches to circumventing the mechanisms of acquired resistance. EXPERIMENTAL DESIGN BT-40 xenografts were selected in vivo for selumetinib resistance. Resistant tumors were obtained and characterized, as were tumors that reverted to sensitivity. Characterization included expression profiling, assessment of MEK signature and compensatory pathways, MEK inhibition, BRAF expression, and cytokine levels. Combination treatment of BT-40/AZD-resistant tumors with the MEK inhibitor and a STAT3 inhibitor (LLL12) was assessed. RESULTS Resistance was unstable, tumors reverting to selumetinib sensitivity when passaged in untreated mice, and MEK was equally inhibited in sensitive and resistant tumors by selumetinib. Drug resistance was associated with an enhanced MEK signature and increased interleukin (IL)-6 and IL-8 expression. Selumetinib treatment induced phosphorylation of STAT3 (Y705) only in resistant xenografts, and similar results were observed in BRAF(V600E) astrocytic cell lines intrinsically resistant to selumetinib. Treatment of BT-40-resistant tumors with selumetinib or LLL12 had no significant effect, whereas combined treatment induced complete regressions of BT-40/AZD-resistant xenografts. CONCLUSIONS Resistance to selumetinib selected in vivo in BT-40 tumor xenografts was unstable. In resistant tumors, selumetinib activated STAT3, and combined treatment with selumetinib and LLL12 induced complete responses in resistant BT-40 tumors. These results suggest dual targeting BRAF (V600E) signaling and STAT3 signaling may be effective in selumetinib-resistant tumors or may retard or prevent onset of resistance.
Collapse
Affiliation(s)
- Hemant K Bid
- Authors' Affiliations: Center for Childhood Cancer & Blood Diseases, Nationwide Children's Hospital; Center for Biostatistics, The Ohio State University, Columbus, Ohio; Institut of Pathology, Department Neuropathology, Ruprecht-Karls University and Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany; and Astrazeneca Ltd., Oncology iMed, Macclesfield, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Okemoto K, Wagner B, Meisen H, Haseley A, Kaur B, Chiocca EA. STAT3 activation promotes oncolytic HSV1 replication in glioma cells. PLoS One 2013; 8:e71932. [PMID: 23936533 PMCID: PMC3732216 DOI: 10.1371/journal.pone.0071932] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/04/2013] [Indexed: 12/05/2022] Open
Abstract
Recent studies report that STAT3 signaling is a master regulator of mesenchymal transformation of gliomas and that STAT3 modulated genes are highly expressed in the mesenchymal transcriptome of gliomas. A currently studied experimental treatment for gliomas consists of intratumoral injection of oncolytic viruses (OV), such as oncolytic herpes simplex virus type 1 (oHSV). We have described one particular oHSV (rQNestin34.5) that exhibits potent anti-glioma activity in animal models. Here, we hypothesized that alterations in STAT3 signaling in glioma cells may affect the replicative ability of rQNestin34.5. In fact, human U251 glioma cells engineered to either over-express STAT3 or with genetic down-regulation of STAT3 supported oHSV replication to a significantly higher or lesser degree, respectively, when compared to controls. Administration of pharmacologic agents that increase STAT3 phosphorylation/activation (Valproic Acid) or increase STAT3 levels (Interleukin 6) also significantly enhanced oHSV replication. Instead, administration of inhibitors of STAT3 phosphorylation/activation (LLL12) significantly reduced oHSV replication. STAT3 led to a reduction in interferon signaling in oHSV infected cells and inhibition of interferon signaling abolished the effect of STAT3 on oHSV replication. These data thus indicate that STAT3 signaling in malignant gliomas enhances oHSV replication, likely by inhibiting the interferon response in infected glioma cells, thus suggesting avenues for possible potentiation of oncolytic virotherapy.
Collapse
Affiliation(s)
- Kazuo Okemoto
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
| | - Benjamin Wagner
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
| | - Hans Meisen
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
| | - Amy Haseley
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
| | - Balveen Kaur
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
| | - Ennio Antonio Chiocca
- Dardinger Center for Neuro-Oncology and Neurosciences, Department of Neurological Surgery, James Cancer Hospital/Solove Research Institute/Comprehensive Cancer Center and Wexner Medical Center, the Ohio State University, Columbus, Ohio, United States of America
- Harvey Cushing Neuro-Oncology Laboratories, Department of Neurosurgery, Institute for the Neurosciences at the Brigham, Brigham and Women’s/Faulkner Hospital and Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
126
|
Overcoming intratumor heterogeneity of polygenic cancer drug resistance with improved biomarker integration. Neoplasia 2013; 14:1278-89. [PMID: 23308059 DOI: 10.1593/neo.122096] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 12/11/2012] [Accepted: 12/11/2012] [Indexed: 12/14/2022] Open
Abstract
Improvements in technology and resources are helping to advance our understanding of cancer-initiating events as well as factors involved with tumor progression, adaptation, and evasion of therapy. Tumors are well known to contain diverse cell populations and intratumor heterogeneity affords neoplasms with a diverse set of biologic characteristics that can be used to evolve and adapt. Intratumor heterogeneity has emerged as a major hindrance to improving cancer patient care. Polygenic cancer drug resistance necessitates reconsidering drug designs to include polypharmacology in pursuit of novel combinatorial agents having multitarget activity to overcome the diverse and compensatory signaling pathways in which cancer cells use to survive and evade therapy. Advances will require integration of different biomarkers such as genomics and imaging to provide for more adequate elucidation of the spatially varying location, type, and extent of diverse intratumor signaling molecules to provide for a rationale-based personalized cancer medicine strategy.
Collapse
|
127
|
Kroon P, Berry PA, Stower MJ, Rodrigues G, Mann VM, Simms M, Bhasin D, Chettiar S, Li C, Li PK, Maitland NJ, Collins AT. JAK-STAT blockade inhibits tumor initiation and clonogenic recovery of prostate cancer stem-like cells. Cancer Res 2013; 73:5288-98. [PMID: 23824741 DOI: 10.1158/0008-5472.can-13-0874] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Interleukin (IL)-6 overexpression and constitutive STAT3 activation occur in many cancers, including prostate cancer. However, their contribution to prostate stem and progenitor cells has not been explored. In this study, we show that stem-like cells from patients with prostate cancer secrete higher levels of IL-6 than their counterparts in non-neoplastic prostate. Tumor grade did not influence the levels of expression or secretion. Stem-like and progenitor cells expressed the IL-6 receptor gp80 with concomitant expression of pSTAT3. Blockade of activated STAT3, by either anti-IL-6 antibody siltuximab (CNTO 328) or LLL12, a specific pSTAT3 inhibitor, suppressed the clonogenicity of the stem-like cells in patients with high-grade disease. In a murine xenograft model used to determine the in vivo effects of pSTAT3 suppression, LLL12 treatment effectively abolished outgrowth of a patient-derived castrate-resistant tumor. Our results indicate that the most primitive cells in prostate cancer require pSTAT3 for survival, rationalizing STAT3 as a therapeutic target to treat advanced prostate cancer.
Collapse
Affiliation(s)
- Paula Kroon
- Yorkshire Cancer Research Unit, Department of Biology, York
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Bhasin D, Chettiar SN, Etter JP, Mok M, Li PK. Anticancer activity and SAR studies of substituted 1,4-naphthoquinones. Bioorg Med Chem 2013; 21:4662-9. [PMID: 23791367 DOI: 10.1016/j.bmc.2013.05.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/30/2013] [Accepted: 05/10/2013] [Indexed: 02/04/2023]
Abstract
In this paper, we report the structure-activity relationship studies of substituted 1,4-naphthoquinones for its anticancer properties. 1,4-Naphthoquinone, Juglone, Menadione, Plumbagin and LLL12.1 were used as lead molecules to design PD compounds. Most of the PD compounds showed improved antiproliferative activity in comparison to the lead molecule in prostate (DU-145), breast (MDA-MB-231) and colon (HT-29) cancer cell lines. PD9, PD10, PD11, PD13, PD14 and PD15 were found to be the most potent compound with an IC₀ value of 1-3 μM in all cancer cell lines. Fluorescent polarization assay was employed to study the inhibition of STAT3 dimerization by PD compounds. PD9 and PD18 were found to be potent STAT3 dimerization inhibitors.
Collapse
Affiliation(s)
- Deepak Bhasin
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Rm 338 Parks Hall, 500 West 12th Avenue, Columbus, OH 43210-1291, United States
| | | | | | | | | |
Collapse
|
129
|
Yu W, Xiao H, Lin J, Li C. Discovery of Novel STAT3 Small Molecule Inhibitors via in Silico Site-Directed Fragment-Based Drug Design. J Med Chem 2013; 56:4402-12. [DOI: 10.1021/jm400080c] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Wenying Yu
- Division of Medicinal Chemistry
and Pharmacognosy, College of Pharmacy, The Ohio State University,
Columbus, Ohio 43210, United States
| | - Hui Xiao
- Center for Childhood Cancer,
The Research Institute at Nationwide Children’s Hospital, Department
of Pediatrics, College of Medicine, The Ohio State University, Columbus,
Ohio 43205, United States
| | - Jiayuh Lin
- Center for Childhood Cancer,
The Research Institute at Nationwide Children’s Hospital, Department
of Pediatrics, College of Medicine, The Ohio State University, Columbus,
Ohio 43205, United States
| | - Chenglong Li
- Division of Medicinal Chemistry
and Pharmacognosy, College of Pharmacy, The Ohio State University,
Columbus, Ohio 43210, United States
| |
Collapse
|
130
|
Katz E, Sims AH, Sproul D, Caldwell H, Dixon MJ, Meehan RR, Harrison DJ. Targeting of Rac GTPases blocks the spread of intact human breast cancer. Oncotarget 2013; 3:608-19. [PMID: 22689141 PMCID: PMC3442288 DOI: 10.18632/oncotarget.520] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
High expression of Rac small GTPases in invasive breast ductal carcinoma is associated with poor prognosis, but its therapeutic value in human cancers is not clear. The aim of the current study was to determine the response of human primary breast cancers to Rac-based drug treatments ex vivo. Three-dimensional organotypic cultures were used to assess candidate therapeutic avenues in invasive breast cancers. Uniquely, in these primary cultures, the tumour is not disaggregated, with both epithelial and mesenchymal components maintained within a three-dimensional matrix of type I collagen. EHT 1864, a small molecule inhibitor of Rac GTPases, prevents spread of breast cancers in this setting, and also reduces proliferation at the invading edge. Rac1+ epithelial cells in breast tumours also contain high levels of the phosphorylated form of the transcription factor STAT3. The small molecule Stattic inhibits activation of STAT3 and induces effects similar to those seen with EHT 1864. Pan-Rac inhibition of proliferation precedes down-regulation of STAT3 activity, defining it as the last step in Rac activation during human breast cancer invasion. Our data highlights the potential use of Rac and STAT3 inhibition in treatment of invasive human breast cancer and the benefit of studying novel cancer treatments using three-dimensional primary tumour tissue explant cultures.
Collapse
Affiliation(s)
- Elad Katz
- Breakthrough Breast Cancer Research Unit, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
131
|
|
132
|
Biologic activity of the novel small molecule STAT3 inhibitor LLL12 against canine osteosarcoma cell lines. BMC Vet Res 2012; 8:244. [PMID: 23244668 PMCID: PMC3585923 DOI: 10.1186/1746-6148-8-244] [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: 09/07/2012] [Accepted: 11/28/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND STAT3 [1] has been shown to be dysregulated in nearly every major cancer, including osteosarcoma (OS). Constitutive activation of STAT3, via aberrant phosphorylation, leads to proliferation, cell survival and resistance to apoptosis. The present study sought to characterize the biologic activity of a novel allosteric STAT3 inhibitor, LLL12, in canine OS cell lines. RESULTS We evaluated the effects of LLL12 treatment on 4 canine OS cell lines and found that LLL12 inhibited proliferation, induced apoptosis, reduced STAT3 phosphorylation, and decreased the expression of several transcriptional targets of STAT3 in these cells. Lastly, LLL12 exhibited synergistic anti-proliferative activity with the chemotherapeutic doxorubicin in the OS lines. CONCLUSION LLL12 exhibits biologic activity against canine OS cell lines through inhibition of STAT3 related cellular functions supporting its potential use as a novel therapy for OS.
Collapse
|
133
|
Liu SH, Wang KB, Lan KH, Lee WJ, Pan HC, Wu SM, Peng YC, Chen YC, Shen CC, Cheng HC, Liao KK, Sheu ML. Calpain/SHP-1 interaction by honokiol dampening peritoneal dissemination of gastric cancer in nu/nu mice. PLoS One 2012; 7:e43711. [PMID: 22937084 PMCID: PMC3427156 DOI: 10.1371/journal.pone.0043711] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Accepted: 07/24/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Honokiol, a small-molecular weight natural product, has previously been reported to activate apoptosis and inhibit gastric tumorigenesis. Whether honokiol inhibits the angiogenesis and metastasis of gastric cancer cells remains unknown. METHODOLOGY/PRINCIPAL FINDINGS We tested the effects of honokiol on angiogenic activity and peritoneal dissemination using in vivo, ex vivo and in vitro assay systems. The signaling responses in human gastric cancer cells, human umbilical vascular endothelial cells (HUVECs), and isolated tumors were detected and analyzed. In a xenograft gastric tumor mouse model, honokiol significantly inhibited the peritoneal dissemination detected by PET/CT technique. Honokiol also effectively attenuated the angiogenesis detected by chick chorioallantoic membrane assay, mouse matrigel plug assay, rat aortic ring endothelial cell sprouting assay, and endothelial cell tube formation assay. Furthermore, honokiol effectively enhanced signal transducer and activator of transcription (STAT-3) dephosphorylation and inhibited STAT-3 DNA binding activity in human gastric cancer cells and HUVECs, which was correlated with the up-regulation of the activity and protein expression of Src homology 2 (SH2)-containing tyrosine phosphatase-1 (SHP-1). Calpain-II inhibitor and siRNA transfection significantly reversed the honokiol-induced SHP-1 activity. The decreased STAT-3 phosphorylation and increased SHP-1 expression were also shown in isolated peritoneal metastatic tumors. Honokiol was also capable of inhibiting VEGF generation, which could be reversed by SHP-1 siRNA transfection. CONCLUSIONS/SIGNIFICANCE Honokiol increases expression and activity of SPH-1 that further deactivates STAT3 pathway. These findings also suggest that honokiol is a novel and potent inhibitor of angiogenesis and peritoneal dissemination of gastric cancer cells, providing support for the application potential of honokiol in gastric cancer therapy.
Collapse
Affiliation(s)
- Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh Bin Wang
- Department of Nuclear Medicine, Kuang Tien General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Keng Hsin Lan
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen Jane Lee
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hung Chuan Pan
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Sheng Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yen Chun Peng
- Division of Gastroenterology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi Ching Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chin Chang Shen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Hsu Chen Cheng
- Department of life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Ko Kaung Liao
- Department of Anatomy, Chung Shan Medical University, Taichung, Taiwan
| | - Meei Ling Sheu
- Department of Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
134
|
Wang X, Crowe PJ, Goldstein D, Yang JL. STAT3 inhibition, a novel approach to enhancing targeted therapy in human cancers (review). Int J Oncol 2012; 41:1181-91. [PMID: 22842992 DOI: 10.3892/ijo.2012.1568] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 02/10/2012] [Indexed: 11/06/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) regulates many critical functions in human normal and malignant tissues, such as differentiation, proliferation, survival, angiogenesis and immune function. Constitutive activation of STAT3 is implicated in a wide range of human cancers. As such, STAT3 has been studied as a tumour therapeutic target. This review aimed principally to summarise the updated research on STAT3 inhibition studies and their therapeutic potential in solid tumours. Recent literature associated with STAT3 inhibition was reviewed through PubMed and Medline database, followed by critical comparison and analysis. Constitutive activation of STAT3 has been identified as abnormal and oncogenic. The pathway of STAT3 activation and signal transduction identifies 3 approaches for inhibition: modulating upstream positive or negative regulators, regulating RNA (DN-STAT3, anti-sense RNA, siRNA and microRNA) or targeting STAT3 protein at different domains. The last approach using small molecule STAT3 inhibitors has been the most examined so far with both preclinical and clinical studies. Targeting STAT3 using a specific inhibitor may be a useful cancer treatment approach, with the potential for a broad clinical impact.
Collapse
Affiliation(s)
- Xiaochun Wang
- Sarcoma Research Group, Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Randwick, NSW, Australia
| | | | | | | |
Collapse
|
135
|
Debnath B, Xu S, Neamati N. Small molecule inhibitors of signal transducer and activator of transcription 3 (Stat3) protein. J Med Chem 2012; 55:6645-68. [PMID: 22650325 DOI: 10.1021/jm300207s] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Bikash Debnath
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90089, United States
| | | | | |
Collapse
|
136
|
Jackson C, Ruzevick J, Amin AG, Lim M. Potential role for STAT3 inhibitors in glioblastoma. Neurosurg Clin N Am 2012; 23:379-89. [PMID: 22748651 DOI: 10.1016/j.nec.2012.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor. Signal transducers and activators of transcription 3 (STAT3) is a transcription factor that translocates to the nucleus to modulate the expression of a variety of genes associated with cell survival, differentiation, proliferation, angiogenesis, and immune function. Several cancers induce constitutive STAT3 activation. Most studies have reported that STAT3 inhibition has antineoplastic activity; however, emerging evidence suggests that the role of STAT3 activity in GBM may be more nuanced than initially appreciated. The authors review the roles of STAT3 in GBM and discuss potential strategies for targeting STAT3.
Collapse
Affiliation(s)
- Christopher Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
137
|
Yeh YT, Lee KT, Tsai CJ, Chen YJ, Wang SN. Prolactin promotes hepatocellular carcinoma through Janus kinase 2. World J Surg 2012; 36:1128-1135. [PMID: 22392353 DOI: 10.1007/s00268-012-1505-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one human cancer with obvious gender disparity. This study investigated the association of aberrant prolactin levels with HCC risk and the potential impacts on HCC of the prolactin receptor (PRLR)/Janus kinase 2 (JAK2) signaling. METHODS Serum prolactin of 63 HCC patients and 162 subjects without HCC was measured by radioimmunoassay. The expressions of PRLR and phosphorylated JAK2 (p-JAK2) in 82 retrospectively collected HCC specimens were evaluated by immunohistochemistry and further incorporated into the survival analysis. The immunoblotting and proliferation assays were used to analyze the effects of PRLR/JAK2 signaling on liver cancer cells with prolactin treatment. RESULTS Serum prolactin level was significantly higher in HCC patients than in controls. Hepatocellular carcinoma patients with high p-JAK2 expression had a significantly higher postoperative risk than those with low p-JAK2 expression. Moreover, results from the multivariate analysis indicated the prognostic role of p-JAK2 expression with respect to overall survival in HCC patients. In addition, the Kaplan-Meier survival curve showed that high p-JAK2 expression was associated with poor survival in HCC patients with high PRLR expression. The immunoblotting assay showed that prolactin induced the expression of both p-JAK2 and cyclin D1 in Hep-G2 cells. Importantly, the proliferative effects induced by prolactin could be effectively attenuated by adding AG490, a JAK2 inhibitor. CONCLUSIONS Increased circulating prolactin was found in HCC patients and high p-JAK2 expression could predict poor overall survival in those patients expressing high PRLR. In addition, prolactin contributed to the proliferation of liver cancer cells through PRLR/JAK2 signaling.
Collapse
Affiliation(s)
- Yao-Tsung Yeh
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - King-Teh Lee
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Jung Tsai
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Jie Chen
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shen-Nien Wang
- Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Cancer Center, Kaohsiung Medical University Hospital, No. 100, Tzyou 1st Rd., Kaohsiung, Taiwan.
| |
Collapse
|
138
|
Forsbach A, Müller C, Montino C, Kritzler A, Nguyen T, Weeratna R, Jurk M, Vollmer J. Negative regulation of the type I interferon signaling pathway by synthetic Toll-like receptor 7 ligands. J Interferon Cytokine Res 2012; 32:254-68. [PMID: 22540943 DOI: 10.1089/jir.2011.0091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Ten Toll-like receptor (TLR) family members have been reported in humans. Here, the endoplasmatic receptors TLR9, TLR8, TLR7, and TLR3 respond to nucleic acids and derivatives or to small molecules (TLR7 and 8). Another cytoplasmic RNA receptor, retinoic acid inducible gene I (RIG-I), is stimulated by 5' triphosphate double-stranded RNA. We discovered that TLR7 small-molecule agonists inhibit nucleic acid-mediated TLR3, TLR7, TLR9, or RIG-I-dependent interferon-α (IFN-α) immune response. Other cytokines and chemokines stimulated by nucleic acid agonists remained unaffected. The observed blockage of TLR3, TLR7, TLR9, and RIG-I-mediated IFN-α response appears to be driven by a competitive mechanism at the type I IFN pathway. Besides type I IFN, IFN response genes such as IFIT-1, Mx1, OAS1, or IRF7 were affected, which indicates that the key element driving the inhibition is located in the type I IFN pathway. Indeed, the heterotrimeric complex formation of phosphor-signal transducer and activator of transcription factor 1 (STAT1), phosphor-STAT2, and IRF9 (called ISGF3, IFN-stimulated gene factor 3) is inhibited through the TLR7 small-molecule agonists by phosphor-STAT2 blockage. These findings provide novel insights into the use of synthetic TLR7 or TLR7/8 small molecules as ligands for immune activation and suppression.
Collapse
Affiliation(s)
- Alexandra Forsbach
- Pfizer Oligonucleotide Therapeutics Unit-Coley Pharmaceutical GmbH , Düsseldorf, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Bid HK, Oswald D, Li C, London CA, Lin J, Houghton PJ. Anti-angiogenic activity of a small molecule STAT3 inhibitor LLL12. PLoS One 2012; 7:e35513. [PMID: 22530037 PMCID: PMC3328460 DOI: 10.1371/journal.pone.0035513] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/16/2012] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Recent data indicate the Signal Transducer and Activator of Transcription 3 (STAT3) pathway is required for VEGF production and angiogenesis in various types of cancers. STAT3 inhibitors have been shown to reduce tumor microvessel density in tumors but a direct anti-angiogenic activity has not been described. METHODOLOGY/PRINCIPAL FINDINGS We investigated the direct action of a small molecule inhibitor of STAT3 (LLL12) in human umbilical cord vascular endothelial cells (HUVECs) in vitro, in a Matrigel model for angiogenesis in vivo, and its antitumor activity in a xenograft model of osteosarcoma. LLL12 (100 nM) significantly inhibited VEGF-stimulated STAT3 phosphorylation in HUVECs, reduced their proliferation/migration and inhibited VEGF-induced tube formation. Morphologic analysis of LLL12 treated HUVECs demonstrated marked changes in actin/tubulin distribution and bundling. In scid mice, LLL12 reduced microvessel invasion into VEGF-infused Matrigel plugs by ∼90% at a dose of 5 mg/kg daily. Following a period of tumor progression (2 weeks), LLL12 completely suppressed further growth of established OS-1 osteosarcoma xenografts. Pharmacodynamic studies showed robust phosphorylated STAT3 in control tumors, whereas phospho-STAT3 was not detected in LLL12-treated OS-1 tumors. Treated tumors demonstrated decreased proliferation (Ki67 staining), and decreased microvessel density (CD34 staining), but no significant increase in apoptosis (TUNEL staining), relative to controls. Assay of angiogenic factors, using an antibody array, showed VEGF, MMP-9, Angiopoietin1/2, Tissue Factor and FGF-1 expression were dramatically reduced in LLL12-treated tumors compared to control tumors. CONCLUSIONS These findings provide the first evidence that LLL12 effectively inhibits tumor angiogenesis both in vitro and in vivo.
Collapse
Affiliation(s)
- Hemant K. Bid
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Duane Oswald
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Chenglong Li
- College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Cheryl A. London
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jiayuh Lin
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, United States of America
| | - Peter J. Houghton
- Center for Childhood Cancer, Nationwide Children's Hospital, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
140
|
Lin L, Benson DM, DeAngelis S, Bakan CE, Li PK, Li C, Lin J. A small molecule, LLL12 inhibits constitutive STAT3 and IL-6-induced STAT3 signaling and exhibits potent growth suppressive activity in human multiple myeloma cells. Int J Cancer 2012; 130:1459-69. [PMID: 21520044 PMCID: PMC3228889 DOI: 10.1002/ijc.26152] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 03/31/2011] [Indexed: 12/27/2022]
Abstract
We characterized the effects of a newly developed signal transducers and activators of transcription 3 (STAT3) inhibitor, LLL12 in multiple myeloma (MM) cells. LLL12 specifically inhibited STAT3 phosphorylation, nuclear localization, DNA binding activity, down-regulated STAT3 downstream genes, and induced apoptosis in MM cells. Importantly, LLL12 significantly inhibited STAT3 phosphorylation, induced apoptosis in primary MM cells which came from patients that were clinically resistant to lenalidomide and bortezomib. LLL12 is a potent inhibitor of cell proliferation with IC50 values ranging between 0.26 and 1.96 μM in MM and primary MM cells. LLL12 also inhibited STAT3 phosphorylation induced by interleukin-6 (IL-6) and interferon-α but not STAT1, STAT2, STAT4 and STAT6 phosphorylation induced by interferon-α, interferon-γ and IL-4 indicating the selectivity of LLL12 for STAT3. The selectively of LLL12 on STAT3 was further demonstrated on 21 protein kinases, which LLL12 had IC50 values ≥ 73.92 μM. In addition, the pretreatment of LLL12 blocked the promotion of the cell proliferation and resistance to lenalidomide by IL-6. Furthermore, LLL12 significantly blocked tumor growth of MM cells in mouse model. Our results indicate that LLL12 blocks constitutive STAT3 and IL-6 induced STAT3 signaling and may be a potential therapeutic agent for MM.
Collapse
Affiliation(s)
- Li Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, 700 Children’s Drive, Columbus, OH 43205
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Don M. Benson
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus OH 43210
| | - Stephanie DeAngelis
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, 700 Children’s Drive, Columbus, OH 43205
| | - Courtney E. Bakan
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus OH 43210
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus OH 43210
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus OH 43210
| | - Jiayuh Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Department of Pediatrics, 700 Children’s Drive, Columbus, OH 43205
| |
Collapse
|
141
|
Glioma cell migration on three-dimensional nanofiber scaffolds is regulated by substrate topography and abolished by inhibition of STAT3 signaling. Neoplasia 2012; 13:831-40. [PMID: 21969816 DOI: 10.1593/neo.11612] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 07/15/2011] [Accepted: 07/21/2011] [Indexed: 02/06/2023] Open
Abstract
A hallmark of malignant gliomas is their ability to disperse through neural tissue, leading to long-term failure of all known therapies. Identifying new antimigratory targets could reduce glioma recurrence and improve therapeutic efficacy, but screens based on conventional migration assays are hampered by the limited ability of these assays to reproduce native cell motility. Here, we have analyzed the motility, gene expression, and sensitivity to migration inhibitors of glioma cells cultured on scaffolds formed by submicron-sized fibers (nanofibers) mimicking the neural topography. Glioma cells cultured on aligned nanofiber scaffolds reproduced the elongated morphology of cells migrating in white matter tissue and were highly sensitive to myosin II inhibition but only moderately affected by stress fiber disruption. In contrast, the same cells displayed a flat morphology and opposite sensitivity to myosin II and actin inhibition when cultured on conventional tissue culture polystyrene. Gene expression analysis indicated a correlation between migration on aligned nanofibers and increased STAT3 signaling, a known driver of glioma progression. Accordingly, cell migration out of glioblastoma-derived neurospheres and tumor explants was reduced by STAT3 inhibitors at subtoxic concentrations. Remarkably, these inhibitors were ineffective when tested at the same concentrations in a conventional two-dimensional migration assay. We conclude that migration of glioma cells is regulated by topographical cues that affect cell adhesion and gene expression. Cell migration analysis using nanofiber scaffolds could be used to reproduce native mechanisms of migration and to identify antimigratory strategies not disclosed by other in vitro models.
Collapse
|
142
|
The interconnectedness of cancer cell signaling. Neoplasia 2012; 13:1183-93. [PMID: 22241964 DOI: 10.1593/neo.111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 12/14/2011] [Accepted: 12/14/2011] [Indexed: 11/18/2022] Open
Abstract
The elegance of fundamental and applied research activities have begun to reveal a myriad of spatial and temporal alterations in downstream signaling networks affected by cell surface receptor stimulation including G protein-coupled receptors and receptor tyrosine kinases. Interconnected biochemical pathways serve to integrate and distribute the signaling information throughout the cell by orchestration of complex biochemical circuits consisting of protein interactions and covalent modification processes. It is clear that scientific literature summarizing results from both fundamental and applied scientific research activities has served to provide a broad foundational biologic database that has been instrumental in advancing our continued understanding of underlying cancer biology. This article reflects on historical advances and the role of innovation in the competitive world of grant-sponsored research.
Collapse
|
143
|
McCormack D, Schneider J, McDonald D, McFadden D. The antiproliferative effects of pterostilbene on breast cancer in vitro are via inhibition of constitutive and leptin-induced Janus kinase/signal transducer and activator of transcription activation. Am J Surg 2011; 202:541-4. [DOI: 10.1016/j.amjsurg.2011.06.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/23/2011] [Accepted: 06/27/2011] [Indexed: 12/01/2022]
|
144
|
Lin L, Liu A, Peng Z, Lin HJ, Li PK, Li C, Lin J. STAT3 is necessary for proliferation and survival in colon cancer-initiating cells. Cancer Res 2011; 71:7226-37. [PMID: 21900397 DOI: 10.1158/0008-5472.can-10-4660] [Citation(s) in RCA: 240] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
STAT3 is constitutively activated in colon cancer but its contributions in cancer-initiating cells have not been explored. In this study, we characterized STAT3 in aldehyde dehydrogenase (ALDH)-positive (ALDH(+)) and CD133-positive (CD133(+)) subpopulations of human colon tumor cells that exhibited more potent tumor-initiating ability than ALDH(-)/CD133(-) cells in tumor xenograft assays in mice. We found that ALDH(+)/CD133(+) cells expressed higher levels of the active phosphorylated form of STAT3 than either ALDH(-)/CD133(-) or unfractionated colon cancer cells. STAT3 inhibition by RNA interference-mediated knockdown or small-molecule inhibitors LLL12 or Stattic blocked downstream target gene expression, cell viability, and tumorsphere-forming capacity in cancer-initiating cells. Similarly, treatment of mouse tumor xenografts with STAT3 short hairpin RNA (shRNA), interleukin 6 shRNA, or LLL12 inhibited tumor growth. Our results establish that STAT3 is constitutively activated in colon cancer-initiating cells and that these cells are sensitive to STAT3 inhibition. These findings establish a powerful rationale to develop STAT3 inhibitory strategies for treating advanced colorectal cancers.
Collapse
Affiliation(s)
- Li Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43205, USA.
| | | | | | | | | | | | | |
Collapse
|
145
|
Signal transducer and activator of transcription 3 (STAT3): a promising target for anticancer therapy. Future Med Chem 2011; 3:567-97. [PMID: 21526897 DOI: 10.4155/fmc.11.22] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an oncogenic protein whose inhibition is sought for the prevention and treatment of cancer. In this review, the validated therapeutic strategy to block aberrant activity of STAT3 in many tumor cell lines is evaluated by presenting the most promising inhibitors to date. The compounds are discussed in classes based on their different mechanisms of action, which are critically explained. In addition, their future clinical development as anticancer agents is considered. Furthermore, the efforts devoted to the comprehension of the structure-activity relationships and to the identification of the biological effects are brought to attention. The synthetic and technological approaches recently developed to overcome the difficulties in the obtainment of clinically suitable drugs are also presented.
Collapse
|
146
|
Fletcher S, Page BDG, Zhang X, Yue P, Li ZH, Sharmeen S, Singh J, Zhao W, Schimmer AD, Trudel S, Turkson J, Gunning PT. Antagonism of the Stat3-Stat3 protein dimer with salicylic acid based small molecules. ChemMedChem 2011; 6:1459-70. [PMID: 21618433 PMCID: PMC3192013 DOI: 10.1002/cmdc.201100194] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Indexed: 12/15/2022]
Abstract
More than 50 new inhibitors of the oncogenic Stat3 protein were identified through a structure-activity relationship (SAR) study based on the previously identified inhibitor S3I-201 (IC₅₀ =86 μM, K(i) >300 μM). A key structural feature of these inhibitors is a salicylic acid moiety, which, by acting as a phosphotyrosine mimetic, is believed to facilitate binding to the Stat3 SH2 domain. Several of the analogues exhibit higher potency than the lead compound in inhibiting Stat3 DNA binding activity, with an in vitro IC₅₀ range of 18.7-51.9 μM, and disruption of Stat3-pTyr peptide interactions with K(i) values in the 15.5-41 μM range. One agent in particular exhibited potent inhibition of Stat3 phosphorylation in both breast and multiple myeloma tumor cells, suppressed the expression of Stat3 target genes, and induced antitumor effects in tumor cells harboring activated Stat3 protein.
Collapse
Affiliation(s)
- Steven Fletcher
- Department of Chemistry, University of Toronto Mississauga, Mississauga, ON, L5L 1C6 (Canada)
| | - Brent D. G. Page
- Department of Chemistry, University of Toronto Mississauga, Mississauga, ON, L5L 1C6 (Canada)
| | - Xialoei Zhang
- Department of Molecular Biology and Microbiology, Burnett College of Biomedical Sciences, University of Central Florida, Orlando, FL, 32826 (USA)
| | - Peibin Yue
- Department of Molecular Biology and Microbiology, Burnett College of Biomedical Sciences, University of Central Florida, Orlando, FL, 32826 (USA)
| | - Zhi Hua Li
- Division of Medical Oncology and Hematology, University Health Network, Princess Margaret Hospital, McLaughlin Centre of Molecular Medicine, 620 University Ave, Toronto, ON, M5G 2C1 (Canada)
| | - Sumaiya Sharmeen
- Ontario Cancer Institute/Princess Margaret Hospital, 610 University Avenue, Toronto, ON, M5G 2M9 (Canada)
| | - Jagdeep Singh
- Department of Chemistry, University of Toronto Mississauga, Mississauga, ON, L5L 1C6 (Canada)
| | - Wei Zhao
- Department of Molecular Biology and Microbiology, Burnett College of Biomedical Sciences, University of Central Florida, Orlando, FL, 32826 (USA)
| | - Aaron D. Schimmer
- Ontario Cancer Institute/Princess Margaret Hospital, 610 University Avenue, Toronto, ON, M5G 2M9 (Canada)
| | - Suzanne Trudel
- Division of Medical Oncology and Hematology, University Health Network, Princess Margaret Hospital, McLaughlin Centre of Molecular Medicine, 620 University Ave, Toronto, ON, M5G 2C1 (Canada)
| | - James Turkson
- Department of Molecular Biology and Microbiology, Burnett College of Biomedical Sciences, University of Central Florida, Orlando, FL, 32826 (USA)
| | - Patrick T. Gunning
- Department of Chemistry, University of Toronto Mississauga, Mississauga, ON, L5L 1C6 (Canada)
| |
Collapse
|
147
|
Li H, Liu A, Zhao Z, Xu Y, Lin J, Jou D, Li C. Fragment-based drug design and drug repositioning using multiple ligand simultaneous docking (MLSD): identifying celecoxib and template compounds as novel inhibitors of signal transducer and activator of transcription 3 (STAT3). J Med Chem 2011; 54:5592-6. [PMID: 21678971 DOI: 10.1021/jm101330h] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We describe a novel method of drug discovery using MLSD and drug repositioning, with cancer target STAT3 being used as a test case. Multiple drug scaffolds were simultaneously docked into hot spots of STAT3 by MLSD, followed by tethering to generate virtual template compounds. Similarity search of virtual hits on drug database identified celecoxib as a novel inhibitor of STAT3. Furthermore, we designed two novel lead inhibitors based on one of the lead templates and celecoxib.
Collapse
Affiliation(s)
- Huameng Li
- Biophysics Graduate Program, The Ohio State University, Columbus, Ohio 43210, United States
| | | | | | | | | | | | | |
Collapse
|
148
|
Liu A, Liu Y, Li PK, Li C, Lin J. LLL12 inhibits endogenous and exogenous interleukin-6-induced STAT3 phosphorylation in human pancreatic cancer cells. Anticancer Res 2011; 31:2029-2035. [PMID: 21737619 PMCID: PMC4288000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Pancreatic cancer is one of the most serious types of cancer, with a five-year survival rate at only 6%. There is a critical need to develop more effective treatments for pancreatic cancer. Growing evidence shows that chronic inflammation plays a crucial role in tumor initiation and progression. Here we demonstrated that the endogenous expression of the inflammatory cytokine interleukin-6 (IL-6) correlates with signal transducer and activator of transcription 3 (STAT3) phosphorylation in human pancreatic cancer cells. Inhibition of the endogenous IL-6/STAT3 pathway reduces cell viability. Exogenous IL-6 induces STAT3 phosphorylation, but differently induces phosphorylation of STAT3 upstream kinases, Janus kinase 1(JAK1), JAK2, and tyrosine kinase 2 (TYK2). Interestingly, LLL12, a nonpeptide, cell-permeable small molecule, selectively blocked exogenous IL-6-induced STAT3 phosphorylation and nuclear translocation in both PANC-1 and ASPC-1 pancreatic cancer cell lines independently of the phosphorylation of JAK1, JAK2, and TYK2. These results suggest that the inhibition of endogenous and exogenous IL-6-mediated STAT3 signaling may be a potential therapeutic approach for pancreatic cancer.
Collapse
Affiliation(s)
- Aiguo Liu
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, OH, U.S.A
- Department of Pediatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R.C
| | - Yan Liu
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, OH, U.S.A
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, U.S.A
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, U.S.A
| | - Jiayuh Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, OH, U.S.A
| |
Collapse
|
149
|
Ball S, Li C, Li PK, Lin J. The small molecule, LLL12, inhibits STAT3 phosphorylation and induces apoptosis in medulloblastoma and glioblastoma cells. PLoS One 2011; 6:e18820. [PMID: 21526200 PMCID: PMC3079737 DOI: 10.1371/journal.pone.0018820] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Accepted: 03/10/2011] [Indexed: 01/05/2023] Open
Abstract
Tumors of the central nervous system represent a major source of cancer-related deaths, with medulloblastoma and glioblastoma being the most common malignant brain tumors in children and adults respectively. While significant advances in treatment have been made, with the 5-year survival rate for medulloblastoma at 70-80%, treating patients under 3 years of age still poses a problem due to the deleterious effects of radiation on the developing brain, and the median survival for patients with glioblastoma is only 15 months. The transcription factor, STAT3, has been found constitutively activated in a wide variety of cancers and in recent years it has become an attractive therapeutic target. We designed a non-peptide small molecule STAT3 inhibitor, LLL12, using structure-based design. LLL12 was able to inhibit STAT3 phosphorylation, decrease cell viability and induce apoptosis in medulloblastoma and glioblastoma cell lines with elevated levels of p-STAT3 (Y705). IC(50) values for LLL12 were found to be between 1.07 µM and 5.98 µM in the five cell lines expressing phosphorylated STAT3. STAT3 target genes were found to be downregulated and a decrease in STAT3 DNA binding was observed following LLL12 treatment, indicating that LLL12 is an effective STAT3 inhibitor. LLL12 was also able to inhibit colony formation, wound healing and decreased IL-6 and LIF secretion. Our results suggest that LLL12 is a potent STAT3 inhibitor and that it may be a potential therapeutic treatment for medulloblastoma and glioblastoma.
Collapse
Affiliation(s)
- Sarah Ball
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Chenglong Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Pui-Kai Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - Jiayuh Lin
- Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
150
|
Lesina M, Kurkowski MU, Ludes K, Rose-John S, Treiber M, Klöppel G, Yoshimura A, Reindl W, Sipos B, Akira S, Schmid RM, Algül H. Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer. Cancer Cell 2011; 19:456-69. [PMID: 21481788 DOI: 10.1016/j.ccr.2011.03.009] [Citation(s) in RCA: 668] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Revised: 12/01/2010] [Accepted: 03/07/2011] [Indexed: 02/08/2023]
Abstract
Physiological levels of Kras(G12D) are sufficient to induce pancreatic intraepithelial neoplasias (PanINs); the mechanisms that drive PanIN progression are unknown. Here, we establish that, in addition to oncogenic Kras(G12D), IL-6 transsignaling-dependent activation of Stat3/Socs3 is required to promote PanIN progression and pancreatic ductal adenocarcinoma (PDAC). Myeloid compartment induces Stat3 activation by secreting IL-6; consequently, IL-6 transsignaling activates Stat3 in the pancreas. Using genetic tools, we show that inactivation of IL-6 transsignaling or Stat3 inhibits PanIN progression and reduces the development of PDAC. Aberrant activation of Stat3 through homozygous deletion of Socs3 in the pancreas accelerates PanIN progression and PDAC development. Our data describe the involvement of IL-6 transsignaling/Stat3/Socs3 in PanIN progression and PDAC development.
Collapse
Affiliation(s)
- Marina Lesina
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|