101
|
Poulsen A, William AD, Dymock BW. Designed Macrocyclic Kinase Inhibitors. MACROCYCLES IN DRUG DISCOVERY 2014. [DOI: 10.1039/9781782623113-00141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cancer continues to present as an increasing and serious global unmet medical need in today's aging population.1 Macrocyclic kinase inhibitors have reached advanced clinical testing and are making an impact in oncologic conditions including myelofibrosis, lymphomas and leukemias. Rheumatoid arthritis (RA) is also beginning to be impacted with the first macrocycle having entered Phase I clinical evaluation in healthy volunteers. Increasing reports of innovative macrocycles in preclinical research are appearing in the literature. Desirable, selective, multi-kinase inhibitory profiles against specific kinases known to be abrogated in cancer, RA, and other diseases have been achieved in a first generation series of clinical stage compact small molecule macrocyclic kinase inhibitors. Herein we discuss their design, synthesis, structure activity relationships and assessment of the latest clinical data in a range of oncologic conditions. Macrocyclic kinase inhibitors have the potential to offer new hope to patients and their families.
Collapse
Affiliation(s)
- Anders Poulsen
- Experimental Therapeutics Centre, A*STAR 11 Biopolis Way, #03-10/11 The Helios 138667 Singapore
| | - Anthony D. William
- Institute of Chemical and Engineering Sciences, A*STAR 11 Biopolis Way, The Helios #03-08 138667 Singapore
| | - Brian W. Dymock
- Department of Pharmacy, National University of Singapore 18 Science Drive 4 117543 Singapore
| |
Collapse
|
102
|
Zhu Y, Hong H, Xu ZP, Li Z, Cai W. Quantum dot-based nanoprobes for in vivo targeted imaging. Curr Mol Med 2014; 13:1549-67. [PMID: 24206136 DOI: 10.2174/1566524013666131111121733] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 05/30/2013] [Accepted: 10/02/2013] [Indexed: 02/06/2023]
Abstract
Fluorescent semiconductor quantum dots (QDs) have attracted tremendous attention over the last decade. The superior optical properties of QDs over conventional organic dyes make them attractive labels for a wide variety of biomedical applications, whereas their potential toxicity and instability in biological environment have puzzled scientific researchers. Much research effort has been devoted to surface modification and functionalization of QDs to make them versatile probes for biomedical applications, and significant progress has been made over the last several years. This review article aims to describe the current state-of-the-art of the synthesis, modification, bioconjugation, and applications of QDs for in vivo targeted imaging. In addition, QD-based multifunctional nanoprobes are also summarized.
Collapse
Affiliation(s)
- Y Zhu
- (W. Cai) Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Room 7137, 1111 Highland Avenue, Madison, WI 53705-2275, USA.
| | | | | | | | | |
Collapse
|
103
|
Martin V, Corso S, Comoglio PM, Giordano S. Increase of MET gene copy number confers resistance to a monovalent MET antibody and establishes drug dependence. Mol Oncol 2014; 8:1561-74. [PMID: 25011627 DOI: 10.1016/j.molonc.2014.06.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/16/2014] [Accepted: 06/16/2014] [Indexed: 12/14/2022] Open
Abstract
The relevant role in cancer played by the tyrosine kinase receptor encoded by the MET oncogene led to the development of specific inhibitors, some of which are now in advanced phases of clinical trials. Previous experience has shown that the main limit to the efficacy of most targeted treatments is the advent of resistance. Mechanisms underlying resistance to MET-specific small tyrosine kinase inhibitors (TKIs) have been already described, while nothing is known about resistance to MET monoclonal antibodies, nor about bypassing resistance to chemical TKIs by antibodies or vice-versa. EBC1 lung cancer cells are MET-addicted as a consequence of gene amplification and thus sensitive to MET inhibitors, including the monovalent form of a MET monoclonal antibody (MV-DN30). We generated cells resistant to this antibody and found that resistance was due to a further increase of gene copy number and a dramatic overexpression of the MET receptor. Such an excess of expression saturated the 'shedding' activity of MV-DN30, and prevented both the efficient down-regulation of the MET receptor from the surface and the inhibition of the ensuing constitutive activation. Notably, antibody-resistant cells remained MET-'addicted' and were still sensitive to MET TKIs. Moreover, antibody-resistant cells became 'drug-dependent', since the removal of MV-DN30 led them to death due to excess of signal. In the mirror experiment, cells made resistant to MET-specific TKIs were still sensitive to treatment with the antibody MV-DN30. These findings suggest that a discontinuous, combined treatment by antibodies and chemical kinase inhibitors may increase the clinical response and bypass resistance to anti-MET targeted therapies.
Collapse
Affiliation(s)
- Valentina Martin
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy
| | - Simona Corso
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy.
| | - Paolo M Comoglio
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy
| | - Silvia Giordano
- University of Torino, Department of Oncology, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy; Candiolo Cancer Institute-FPO, IRCCS, S.P. 142, Km 3.95, 10060 Candiolo, Torino, Italy.
| |
Collapse
|
104
|
Rosanio S, Pelliccia F, Gaudio C, Greco C, Keylani AM, D'Agostino DC. Pulmonary arterial hypertension in adults: novel drugs and catheter ablation techniques show promise? Systematic review on pharmacotherapy and interventional strategies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:743868. [PMID: 25013799 PMCID: PMC4072027 DOI: 10.1155/2014/743868] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 04/18/2014] [Indexed: 11/25/2022]
Abstract
This systematic review aims to provide an update on pharmacological and interventional strategies for the treatment of pulmonary arterial hypertension in adults. Currently US Food and Drug Administration approved drugs including prostanoids, endothelin-receptor antagonists, phosphodiesterase type-5 inhibitors, and soluble guanylate-cyclase stimulators. These agents have transformed the prognosis for pulmonary arterial hypertension patients from symptomatic improvements in exercise tolerance ten years ago to delayed disease progression today. On the other hand, percutaneous balloon atrioseptostomy by using radiofrequency perforation, cutting balloon dilatation, or insertion of butterfly stents and pulmonary artery catheter-based denervation, both associated with very low rate of major complications and death, should be considered in combination with specific drugs at an earlier stage rather than late in the progression of pulmonary arterial hypertension and before the occurrence of overt right-sided heart failure.
Collapse
Affiliation(s)
- Salvatore Rosanio
- Division of Cardiology, Department of Internal Medicine, University of North Texas Health Science Center, 855 Montgomery Street, PCC Room 315, Fort Worth, TX 76107, USA
| | - Francesco Pelliccia
- Department of Heart and Great Vessels “Attilio Reale”, La Sapienza University, Rome, Italy
| | - Carlo Gaudio
- Department of Heart and Great Vessels “Attilio Reale”, La Sapienza University, Rome, Italy
| | - Cesare Greco
- Department of Heart and Great Vessels “Attilio Reale”, La Sapienza University, Rome, Italy
| | - Abdul M. Keylani
- Division of Cardiology, Department of Internal Medicine, University of North Texas Health Science Center, 855 Montgomery Street, PCC Room 315, Fort Worth, TX 76107, USA
| | - Darrin C. D'Agostino
- Division of Cardiology, Department of Internal Medicine, University of North Texas Health Science Center, 855 Montgomery Street, PCC Room 315, Fort Worth, TX 76107, USA
| |
Collapse
|
105
|
Kaumaya PTP. Bridging oncology and immunology: expanding horizons with innovative peptide vaccines and peptidomimetics. Immunotherapy 2014; 5:1159-63. [PMID: 24188668 DOI: 10.2217/imt.13.128] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Pravin T P Kaumaya
- The Ohio State University Wexner Medical Center, Department of Obstetrics & Gynecology, 410 W 10th Avenue N729, Columbus, OH 43210, USA and The Comprehensive Cancer Center, The Ohio State University, 300 W 10th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
106
|
Deng J, Shao J, Markowitz JS, An G. ABC Transporters in Multi-Drug Resistance and ADME-Tox of Small Molecule Tyrosine Kinase Inhibitors. Pharm Res 2014; 31:2237-55. [DOI: 10.1007/s11095-014-1389-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 04/15/2014] [Indexed: 12/31/2022]
|
107
|
Li J, Pang Q. Oxidative stress-associated protein tyrosine kinases and phosphatases in Fanconi anemia. Antioxid Redox Signal 2014; 20:2290-301. [PMID: 24206276 PMCID: PMC3995293 DOI: 10.1089/ars.2013.5715] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
SIGNIFICANCE Fanconi anemia (FA) is a genetic disorder featuring chromosomal instability, developmental defects, progressive bone marrow failure, and predisposition to cancer. Besides the predominant role in DNA damage response and/or repair, many studies have linked FA proteins to oxidative stress. Oxidative stress, defined as imbalance in pro-oxidant and antioxidant homeostasis, has been considered to contribute to disease development, including FA. RECENT ADVANCES A variety of signaling pathways may be influenced by oxidative stress, particularly the equilibrium between protein kinases and phosphatases, consequently leading to an aberrant phosphorylation state of cellular proteins. Dysfunction of kinases/phosphatases has been implicated in the pathophysiology of human diseases. In FA, evidence is emerging that links abnormal phosphorylation/de-phosphorylation of signaling molecules to clinical complications and malformations. CRITICAL ISSUES In this study, we review the recent findings on the oxidative stress-related kinases and phosphatases, particularly tyrosine phosphatases in FA. FUTURE DIRECTIONS Understanding the role of oxidative stress-related kinases and phosphatases in FA may provide unique and generic possibilities for the future development of therapeutic strategies by targeting the dysregulated protein kinases and phosphatases in a clinical setting.
Collapse
Affiliation(s)
- Jie Li
- 1 Division of Neurosurgery, Center for Theoretic and Applied Neuro-Oncology, Moores Cancer Center, University of California , San Diego, La Jolla, California
| | | |
Collapse
|
108
|
Gyurkovska V, Dimitrova P, Ivanovska N. Tyrosine Kinase Inhibitor Tyrphostin AG490 Inhibits Osteoclast Differentiation in Collagenase-Induced Osteoarthritis. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade plays a principal role in the signaling of a vast array of cytokines and growth factors which stimulates diverse cellular functions and immune responses. Osteoarthritis (OA) is the most common joint disease in the adult population. The present study was designed to evaluate the effects of tyrosine kinase inhibitor, tyrphostin AG490 in a mouse model of collagenase-induced osteoarthritis (CIOA). CIOA was provoked by two intraarticular (i.a.) injections of collagenase in mice and intraperitoneally (i.p.) treated with AG490 at a dose of 5 mg/kg at days 0, 5 and 10 and at a dose of 8 mg/kg at day 18. The administration of AG490 in CIOA mice inhibited osteoclast generation in bone and the loss of glycosaminoglycans and proteoglycans in cartilage. Tyrphostin decreased the levels of IFN-γ, IL1, IL-6 and IL-17 in the synovial fluid (SF) dependant on the time post AG490 administration. Limited numbers of CD11b positive Ly6G neutrophils in blood and SF along with a decrease of F4/80 positive cells in synovial fluid (SF) were observed in tyrphostin AG490-treated arthritic mice. AG490 inhibited M-CSF+RANKL-induced cytokine production by bone marrow (BM) cells and the differentiation of BM cells in vitro. Because of the findings presented, we argue that tyrphostin AG490 may hold promising therapeutic potential against important clinical conditions such as osteoarthritis (OA).
Collapse
Affiliation(s)
- V. Gyurkovska
- Department of Immunology, Institute of Microbiology, Sofia, Bulgaria
| | - P. Dimitrova
- Department of Immunology, Institute of Microbiology, Sofia, Bulgaria
| | - N. Ivanovska
- Department of Immunology, Institute of Microbiology, Sofia, Bulgaria
| |
Collapse
|
109
|
Tundidor Y, García-Hernández CP, Pupo A, Cabrera Infante Y, Rojas G. Delineating the functional map of the interaction between nimotuzumab and the epidermal growth factor receptor. MAbs 2014; 6:1013-25. [PMID: 24759767 DOI: 10.4161/mabs.28915] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Molecular details of epidermal growth factor receptor (EGFR) targeting by nimotuzumab, a therapeutic anti-cancer antibody, have been largely unknown. The current study delineated a functional map of their interface, based on phage display and extensive mutagenesis of both the target antigen and the Fv antibody fragment. Five residues in EGFR domain III (R353, S356, F357, T358, and H359T) and the third hypervariable region of nimotuzumab heavy chain were shown to be major functional contributors to the interaction. Fine specificity differences between nimotuzumab and other anti-EGFR antibodies were revealed. Mapping information guided the generation of a plausible in silico binding model. Knowledge about the epitope/paratope interface opens new avenues for the study of tumor sensitivity/resistance to nimotuzumab and for further engineering of its binding site. The developed mapping platform, also validated with the well-known cetuximab epitope, allows a comprehensive exploration of antigenic regions and could be expanded to map other anti-EGFR antibodies.
Collapse
Affiliation(s)
- Yaima Tundidor
- Systems Biology Department; Center of Molecular Immunology; Habana, Cuba
| | | | - Amaury Pupo
- Systems Biology Department; Center of Molecular Immunology; Habana, Cuba
| | | | - Gertrudis Rojas
- Systems Biology Department; Center of Molecular Immunology; Habana, Cuba
| |
Collapse
|
110
|
Hahn VS, Lenihan DJ, Ky B. Cancer therapy-induced cardiotoxicity: basic mechanisms and potential cardioprotective therapies. J Am Heart Assoc 2014; 3:e000665. [PMID: 24755151 PMCID: PMC4187516 DOI: 10.1161/jaha.113.000665] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/26/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Virginia Shalkey Hahn
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (V.S.H., B.K.)
| | - Daniel J. Lenihan
- Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (D.J.L.)
| | - Bonnie Ky
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (V.S.H., B.K.)
- Penn Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (B.K.)
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (B.K.)
| |
Collapse
|
111
|
Wang X, Yennawar N, Hankey PA. Autoinhibition of the Ron receptor tyrosine kinase by the juxtamembrane domain. Cell Commun Signal 2014; 12:28. [PMID: 24739671 PMCID: PMC4021555 DOI: 10.1186/1478-811x-12-28] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 02/05/2014] [Indexed: 01/21/2023] Open
Abstract
Background The Ron receptor tyrosine kinase (RTK) has been implicated in the progression of a number of carcinomas, thus understanding the regulatory mechanisms governing its activity is of potential therapeutic significance. A critical role for the juxtamembrane domain in regulating RTK activity is emerging, however the mechanism by which this regulation occurs varies considerably from receptor to receptor. Results Unlike other RTKs described to date, tyrosines in the juxtamembrane domain of Ron are inconsequential for receptor activation. Rather, we have identified an acidic region in the juxtamembrane domain of Ron that plays a central role in promoting receptor autoinhibition. Furthermore, our studies demonstrate that phosphorylation of Y1198 in the kinase domain promotes Ron activation, likely by relieving the inhibitory constraints imposed by the juxtamembrane domain. Conclusions Taken together, our experimental data and molecular modeling provide a better understanding of the mechanisms governing Ron activation, which will lay the groundwork for the development of novel therapeutic approaches for targeting Ron in human malignancies.
Collapse
Affiliation(s)
| | | | - Pamela A Hankey
- Graduate Program in Cell and Developmental Biology, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
112
|
Analysis of growth factor signaling in genetically diverse breast cancer lines. BMC Biol 2014; 12:20. [PMID: 24655548 PMCID: PMC4234128 DOI: 10.1186/1741-7007-12-20] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 03/17/2014] [Indexed: 01/22/2023] Open
Abstract
Background Soluble growth factors present in the microenvironment play a major role in tumor development, invasion, metastasis, and responsiveness to targeted therapies. While the biochemistry of growth factor-dependent signal transduction has been studied extensively in individual cell types, relatively little systematic data are available across genetically diverse cell lines. Results We describe a quantitative and comparative dataset focused on immediate-early signaling that regulates the AKT (AKT1/2/3) and ERK (MAPK1/3) pathways in a canonical panel of well-characterized breast cancer lines. We also provide interactive web-based tools to facilitate follow-on analysis of the data. Our findings show that breast cancers are diverse with respect to ligand sensitivity and signaling biochemistry. Surprisingly, triple negative breast cancers (TNBCs; which express low levels of ErbB2, progesterone and estrogen receptors) are the most broadly responsive to growth factors and HER2amp cancers (which overexpress ErbB2) the least. The ratio of ERK to AKT activation varies with ligand and subtype, with a systematic bias in favor of ERK in hormone receptor positive (HR+) cells. The factors that correlate with growth factor responsiveness depend on whether fold-change or absolute activity is considered the key biological variable, and they differ between ERK and AKT pathways. Conclusions Responses to growth factors are highly diverse across breast cancer cell lines, even within the same subtype. A simple four-part heuristic suggests that diversity arises from variation in receptor abundance, an ERK/AKT bias that depends on ligand identity, a set of factors common to all receptors that varies in abundance or activity with cell line, and an “indirect negative regulation” by ErbB2. This analysis sets the stage for the development of a mechanistic and predictive model of growth factor signaling in diverse cancer lines. Interactive tools for looking up these results and downloading raw data are available at http://lincs.hms.harvard.edu/niepel-bmcbiol-2014/.
Collapse
|
113
|
Rodríguez J, Castañeda G, Muñoz L, Navarro D, Villa JC. Simultaneous determination of erlotinib and metabolites in human urine using capillary electrophoresis. Electrophoresis 2014; 35:1489-95. [DOI: 10.1002/elps.201300573] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/10/2014] [Accepted: 01/27/2014] [Indexed: 01/29/2023]
Affiliation(s)
- Juana Rodríguez
- Department of Analytical Chemistry and Food Technology; University of Castilla-La Mancha; Ciudad Real Spain
| | - Gregorio Castañeda
- Department of Analytical Chemistry and Food Technology; University of Castilla-La Mancha; Ciudad Real Spain
| | - Lorena Muñoz
- Department of Analytical Chemistry and Food Technology; University of Castilla-La Mancha; Ciudad Real Spain
| | - Diana Navarro
- Department of Analytical Chemistry and Food Technology; University of Castilla-La Mancha; Ciudad Real Spain
| | - Jose C. Villa
- Department of Clinical Oncology; General University Hospital of Ciudad Real; Ciudad Real Spain
| |
Collapse
|
114
|
Casimiro MC, Velasco-Velázquez M, Aguirre-Alvarado C, Pestell RG. Overview of cyclins D1 function in cancer and the CDK inhibitor landscape: past and present. Expert Opin Investig Drugs 2014; 23:295-304. [PMID: 24387133 DOI: 10.1517/13543784.2014.867017] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Intensive efforts, over the last decade, have been made to inhibit the kinase activity of cyclins that act as mediators during cell-cycle progression. Activation of the cyclin D1 oncogene, often by amplification or rearrangement, is a major driver of multiple types of human tumors including breast and squamous cell cancers, B-cell lymphoma, myeloma and parathyroid adenoma. AREAS COVERED In this review, the authors summarize the activity of cyclins and cyclin-dependent kinases in cell-cycle progression and transcription. They focus on cyclin D1/CDK4/CDK6, a central mediator in the transition from G1 to S phase. Furthermore, the authors discuss the first generation of pan-cyclin-dependent kinase inhibitors that failed to meet expectation and discuss, in detail, the second generation of highly specific cyclin D1/CDK4/CDK6 inhibitors that are proving to be more efficacious. EXPERT OPINION The mechanism by which cyclin D1 drives tumorigenesis may be dependent on kinase and kinase-independent functions. Further evidence is necessary to delineate the roles of cyclin D1 in early pre-neoplastic lesions where its overexpression may promote genomic instability in a kinase-independent manner.
Collapse
Affiliation(s)
- Mathew C Casimiro
- Thomas Jefferson University & Hospital, Department of Cancer Biology , 233 South 10th Street, Philadelphia, PA 19107 , USA
| | | | | | | |
Collapse
|
115
|
Ji L, Zheng W, Lin Y, Wang X, Lü S, Hao X, Luo Q, Li X, Yang L, Wang F. Novel ruthenium complexes ligated with 4-anilinoquinazoline derivatives: synthesis, characterisation and preliminary evaluation of biological activity. Eur J Med Chem 2014; 77:110-20. [PMID: 24631730 DOI: 10.1016/j.ejmech.2014.02.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 02/24/2014] [Accepted: 02/28/2014] [Indexed: 10/25/2022]
Abstract
The ruthenium DMSO complexes cis-Ru(II)C12(DMSO)4 and [(DMSO)2H][trans-Ru(III)Cl4(DMSO)2] reacted with 4-(3'-chloro-4'-fluoroanilino)-6-(2-(2-aminoethyl)aminoethoxy)-7-methoxyquinazoline (L1), 4-(3'-chloro-4'-fluoroanilino)-6-(2-(1H-imidazol-1-yl)ethoxy)-7-methoxy quinazoline (L2), N-(benzo[d]imidazol-4-yl)-6,7-dimethoxyquinazolin-4-amine hydrochloride (L3), 5-(6,7-dimethoxyquinazolin-4-ylamino)quinolin-8-ol hydrochloride (L4), respectively, to afford [Ru(II)Cl2(DMSO)2(L1)] (1), [Ru(III)Cl3(DMSO)(L1)] (2), [Ru(III)Cl4(DMSO)(H-L2)] (3), [Ru(III)Cl4(DMSO)(H-L3)] (4), and [Ru(III)Cl3(DMSO)(H-L4)] (5), which were characterised by mass spectrometry, NMR, elementary analysis and single crystal X-ray diffraction (complex 1). Experimental screening (ELISA) showed that complexes 1, 2 and 3 are remarkably inhibitory towards epidermal growth factor receptor (EGFR) with IC50 values at submicromolar or nanomolar level. Docking studies indicated that complexation with ruthenium has little interference with the formation of the two essential H-bonds between the N3 of the quinazoline ring in L1 and L2 and O-H of Thr766 through a water molecule, and the N1 of the quinazoline ring and N-H of Met769 in EGFR. Moreover, complex 2 was shown to be more active against the EGF-stimulated proliferation of human breast cancer cell line MCF-7 than the better EGFR inhibitor 4-(3'-chloro-4'-fluoroanilino)-6,7-dimethoxyquinazoline, being more potential to induce early-stage apoptosis than gefitinib. These imply that apart from inhibiting EGFR, complex 2 may involve in regulating other biological events related to the proliferation of MCF-7, implicating a novel type of multi-targeting metal-based anticancer agents.
Collapse
Affiliation(s)
- Liyun Ji
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Wei Zheng
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Yu Lin
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiuli Wang
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Shuang Lü
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Xiang Hao
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| | - Xianchan Li
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China
| | - Ling Yang
- Laboratory of Pharmaceutical Resource Discovery, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences, PR China; CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, PR China.
| |
Collapse
|
116
|
Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev 2014; 66:2-25. [PMID: 24270007 PMCID: PMC4219254 DOI: 10.1016/j.addr.2013.11.009] [Citation(s) in RCA: 1990] [Impact Index Per Article: 180.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/23/2013] [Accepted: 11/13/2013] [Indexed: 12/17/2022]
Abstract
Cancer nanotherapeutics are progressing at a steady rate; research and development in the field has experienced an exponential growth since early 2000's. The path to the commercialization of oncology drugs is long and carries significant risk; however, there is considerable excitement that nanoparticle technologies may contribute to the success of cancer drug development. The pace at which pharmaceutical companies have formed partnerships to use proprietary nanoparticle technologies has considerably accelerated. It is now recognized that by enhancing the efficacy and/or tolerability of new drug candidates, nanotechnology can meaningfully contribute to create differentiated products and improve clinical outcome. This review describes the lessons learned since the commercialization of the first-generation nanomedicines including DOXIL® and Abraxane®. It explores our current understanding of targeted and non-targeted nanoparticles that are under various stages of development, including BIND-014 and MM-398. It highlights the opportunities and challenges faced by nanomedicines in contemporary oncology, where personalized medicine is increasingly the mainstay of cancer therapy. We revisit the fundamental concepts of enhanced permeability and retention effect (EPR) and explore the mechanisms proposed to enhance preferential "retention" in the tumor, whether using active targeting of nanoparticles, binding of drugs to their tumoral targets or the presence of tumor associated macrophages. The overall objective of this review is to enhance our understanding in the design and development of therapeutic nanoparticles for treatment of cancers.
Collapse
Affiliation(s)
- Nicolas Bertrand
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jun Wu
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Xiaoyang Xu
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Nazila Kamaly
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA
| | - Omid C Farokhzad
- Laboratory of Nanomedicine and Biomaterials, Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St., Boston, MA 02115, USA.
| |
Collapse
|
117
|
Schönherr M, Bhattacharya A, Kottek T, Szymczak S, Köberle M, Wickenhauser C, Siebolts U, Saalbach A, Koczan D, Magin TM, Simon JC, Kunz M. Genomewide RNAi screen identifies protein kinase Cb and new members of mitogen-activated protein kinase pathway as regulators of melanoma cell growth and metastasis. Pigment Cell Melanoma Res 2014; 27:418-30. [PMID: 24406113 DOI: 10.1111/pcmr.12216] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 01/07/2014] [Indexed: 01/13/2023]
Abstract
A large-scale RNAi screen was performed for eight different melanoma cell lines using a pooled whole-genome lentiviral shRNA library. shRNAs affecting proliferation of transduced melanoma cells were negatively selected during 10 days of culture. Overall, 617 shRNAs were identified by microarray hybridization. Pathway analyses identified mitogen-activated protein kinase (MAPK) pathway members such as ERK1/2, JNK1/2 and MAP3K7 and protein kinase C β (PKCβ) as candidate genes. Knockdown of PKCβ most consistently reduced cellular proliferation, colony formation and migratory capacity of melanoma cells and was selected for further validation. PKCβ showed enhanced expression in human primary melanomas and distant metastases as compared with benign melanocytic nevi. Moreover, treatment of melanoma cells with PKCβ-specific inhibitor enzastaurin reduced melanoma cell growth but had only small effects on benign fibroblasts. Finally, PKCβ-shRNA significantly reduced lung colonization capacity of stably transduced melanoma cells in mice. Taken together, this study identified new candidate genes for melanoma cell growth and proliferation. PKCβ seems to play an important role in these processes and might serve as a new target for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Madeleine Schönherr
- Department of Dermatology, Venereology and Allergology, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
New chemical scaffolds for human african trypanosomiasis lead discovery from a screen of tyrosine kinase inhibitor drugs. Antimicrob Agents Chemother 2014; 58:2202-10. [PMID: 24468788 DOI: 10.1128/aac.01691-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Human African trypanosomiasis (HAT) is caused by the protozoan Trypanosoma brucei. New drugs are needed to treat HAT because of undesirable side effects and difficulties in the administration of the antiquated drugs that are currently used. In human proliferative diseases, protein tyrosine kinase (PTK) inhibitors (PTKIs) have been developed into drugs (e.g., lapatinib and erlotinib) by optimization of a 4-anilinoquinazoline scaffold. Two sets of facts raise a possibility that drugs targeted against human PTKs could be "hits" for antitrypanosomal lead discoveries. First, trypanosome protein kinases bind some drugs, namely, lapatinib, CI-1033, and AEE788. Second, the pan-PTK inhibitor tyrphostin A47 blocks the endocytosis of transferrin and inhibits trypanosome replication. Following up on these concepts, we performed a focused screen of various PTKI drugs as possible antitrypanosomal hits. Lapatinib, CI-1033, erlotinib, axitinib, sunitinib, PKI-166, and AEE788 inhibited the replication of bloodstream T. brucei, with a 50% growth inhibitory concentration (GI50) between 1.3 μM and 2.5 μM. Imatinib had no effect (i.e., GI50>10 μM). To discover leads among the drugs, a mouse model of HAT was used in a proof-of-concept study. Orally administered lapatinib reduced parasitemia, extended the survival of all treated mice, and cured the trypanosomal infection in 25% of the mice. CI-1033 and AEE788 reduced parasitemia and extended the survival of the infected mice. On the strength of these data and noting their oral bioavailabilities, we propose that the 4-anilinoquinazoline and pyrrolopyrimidine scaffolds of lapatinib, CI-1033, and AEE788 are worth optimizing against T. brucei in medicinal chemistry campaigns (i.e., scaffold repurposing) to discover new drugs against HAT.
Collapse
|
119
|
Ding SJ, Qian WJ, Smith RD. Quantitative proteomic approaches for studying phosphotyrosine signaling. Expert Rev Proteomics 2014; 4:13-23. [PMID: 17288512 DOI: 10.1586/14789450.4.1.13] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Protein tyrosine phosphorylation is a fundamental mechanism for controlling many aspects of cellular processes, as well as aspects of human health and diseases. Compared with phosphoserine and phosphothreonine, phosphotyrosine signaling is more tightly regulated, but often more challenging to characterize, due to significantly lower levels of tyrosine phosphorylation (i.e., a relative abundance of 1800:200:1 was estimated for phosphoserine/phosphothreonine/phosphotyrosine in vertebrate cells). In this review, we outline recent advances in analytical methodologies for enrichment, identification and accurate quantitation of tyrosine-phosphorylated proteins and peptides. Advances in antibody-based technologies, capillary liquid chromatography coupled with mass spectrometry, and various stable isotope labeling strategies are discussed, as well as non-mass spectrometry-based methods, such as those using protein/peptide arrays. As a result of these advances, powerful tools now have the power to crack signal transduction codes at the system level, and provide a basis for discovering novel drug targets for human diseases.
Collapse
Affiliation(s)
- Shi-Jian Ding
- Pacific Northwest National Laboratory, Biological Science Division & Environmental Molecular Sciences Laboratory, Richland, WA 99352, USA.
| | | | | |
Collapse
|
120
|
Rahman AFMM, Korashy HM, Kassem MG. Gefitinib. PROFILES OF DRUG SUBSTANCES, EXCIPIENTS, AND RELATED METHODOLOGY 2014; 39:239-264. [PMID: 24794908 DOI: 10.1016/b978-0-12-800173-8.00005-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Gefitinib (Iressa®) is a selective inhibitor of epidermal growth factor, a growth factor that plays a pivotal role in the control of cell growth, apoptosis, and angiogenesis. Gefitinib is clinically used for the treatment of chemoresistant non-small cell lung cancer patients. Gefitinib is freely soluble in dimethylsulphoxide but slightly soluble in methanol and ethanol. Several methods of gefitinib synthesis are included in this review. UV spectroscopy of gefitinib showed a λmax of approximately 331nm, whereas IR spectroscopy principal peaks were observed at 3400cm(-1) (NH), 2956cm(-1) (CH2, CH, alkyl), 1625cm(-1) (CC, CN), 1500cm(-1) (HCCH, aryl), 1110cm(-1) (CO), 1028cm(-1) (CF). In addition, different analytical methods for determination of gefitinib are also described in this review. Pharmacokinetically, after oral administration, gefitinib is slowly absorbed with bioavailability of approximately 60% in human. Gefitinib is metabolized extensively in the liver into five metabolites by cytochrome P450s, primarily by CYP3A4 and to a lesser extent by CYP3A5 and CYP2D6. Gefitinib is eliminated mainly hepatically with total plasma clearance of 595mL/min after intravenous administration. Most of the adverse effects associated with gefitinib therapy are mild to moderate in severity and are usually reversible and manageable with appropriate intervention, such as diarrhea, dry skin, rash, nausea, and vomiting.
Collapse
Affiliation(s)
- A F M Motiur Rahman
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hesham M Korashy
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Gabr Kassem
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
121
|
Rettew AN, Getty PJ, Greenfield EM. Receptor tyrosine kinases in osteosarcoma: not just the usual suspects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:47-66. [PMID: 24924168 DOI: 10.1007/978-3-319-04843-7_3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Despite aggressive surgical and chemotherapy protocols, survival rates for osteosarcoma patients have not improved over the last 30 years. Therefore, novel therapeutic agents are needed. Receptor tyrosine kinases have emerged as targets for the development of new cancer therapies since their activation leads to enhanced proliferation, survival, and metastasis. In fact, aberrant expression and activation of RTKs have been associated with the progression of many cancers. Studies from our lab using phosphoproteomic screening identified RTKs that are activated and thus may contribute to the signaling within metastatic human osteosarcoma cells. Functional genomic screening using siRNA was performed to distinguish which of the activated RTKs contribute to in vitro phenotypes associated with metastatic potential (motility, invasion, colony formation, and cell growth). The resulting RTK hits were then validated using independent validation experiments. From these results, we identified four RTKs (Axl, EphB2, FGFR2, and Ret) that have not been previously studied in osteosarcoma and provide targets for the development of novel therapeutics.
Collapse
Affiliation(s)
- Ashley N Rettew
- Department of Orthopaedics, Case Medical Center, Case Western Reserve University, Cleveland, OH, USA,
| | | | | |
Collapse
|
122
|
Upponi JR, Torchilin VP. Passive vs. Active Targeting: An Update of the EPR Role in Drug Delivery to Tumors. NANO-ONCOLOGICALS 2014. [DOI: 10.1007/978-3-319-08084-0_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
123
|
DeNardo BD, Holloway MP, Ji Q, Nguyen KT, Cheng Y, Valentine MB, Salomon A, Altura RA. Quantitative phosphoproteomic analysis identifies activation of the RET and IGF-1R/IR signaling pathways in neuroblastoma. PLoS One 2013; 8:e82513. [PMID: 24349301 PMCID: PMC3859635 DOI: 10.1371/journal.pone.0082513] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 10/24/2013] [Indexed: 12/20/2022] Open
Abstract
Neuroblastoma is an embryonal tumor of childhood with a heterogenous clinical presentation that reflects differences in activation of complex biological signaling pathways. Protein phosphorylation is a key component of cellular signal transduction and plays a critical role in processes that control cancer cell growth and survival. We used shotgun LC/MS to compare phosphorylation between a human MYCN amplified neuroblastoma cell line (NB10), modeling a resistant tumor, and a human neural precursor cell line (NPC), modeling a normal baseline neural crest cell. 2181 unique phosphorylation sites representing 1171 proteins and 2598 phosphopeptides were found. Protein kinases accounted for 6% of the proteome, with a predominance of tyrosine kinases, supporting their prominent role in oncogenic signaling pathways. Highly abundant receptor tyrosine kinase (RTK) phosphopeptides in the NB10 cell line relative to the NPC cell line included RET, insulin-like growth factor 1 receptor/insulin receptor (IGF-1R/IR), and fibroblast growth factor receptor 1 (FGFR1). Multiple phosphorylated peptides from downstream mediators of the PI3K/AKT/mTOR and RAS pathways were also highly abundant in NB10 relative to NPC. Our analysis highlights the importance of RET, IGF-1R/IR and FGFR1 as RTKs in neuroblastoma and suggests a methodology that can be used to identify potential novel biological therapeutic targets. Furthermore, application of this previously unexploited technology in the clinic opens the possibility of providing a new wide-scale molecular signature to assess disease progression and prognosis.
Collapse
Affiliation(s)
- Bradley D. DeNardo
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Michael P. Holloway
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Qinqin Ji
- Department of Chemistry, Brown University, Providence, Rhode Island, United States of America
| | - Kevin T. Nguyen
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Yan Cheng
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| | - Marcus B. Valentine
- St. Jude Comprehensive Cancer Center Cytogenetic Shared Resource, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Arthur Salomon
- Department of Molecular and Cellular Biochemistry, Brown University, Providence, Rhode Island, United States of America
| | - Rachel A. Altura
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, The Warren Albert School of Medicine at Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
124
|
Stehle F, Schulz K, Seliger B. Towards defining biomarkers indicating resistances to targeted therapies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1844:909-16. [PMID: 24269379 DOI: 10.1016/j.bbapap.2013.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 10/17/2013] [Accepted: 11/13/2013] [Indexed: 12/20/2022]
Abstract
An impressive, but often short objective response was obtained in many tumor patients treated with different targeted therapies, but most of the patients develop resistances against these drugs. So far, a number of distinct mechanisms leading to intrinsic as well as acquired resistances have been identified in tumors of distinct origin. These can arise from genetic alterations, like mutations, truncations, and amplifications or due to deregulated expression of various proteins and signal transduction pathways, but also from cellular heterogeneity within tumors after an initial response. Therefore, biomarkers are urgently needed for cancer prognosis and personalized cancer medicine. The application of "ome"-based technologies including cancer (epi)genomics, next generation sequencing, cDNA microarrays and proteomics might led to the predictive or prognostic stratification of patients to categorize resistance mechanisms and to postulate combinations of treatment strategies. This review discusses the implementation of proteome-based analysis to identify markers of pathway (in)activation in tumors and the resistance mechanisms, which represent major clinical problems as a tool to optimize individually tailored therapies based on targeted drugs. This article is part of a Special Issue entitled: Biomarkers: A Proteomic Challenge.
Collapse
Affiliation(s)
- Franziska Stehle
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany
| | - Kristin Schulz
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany
| | - Barbara Seliger
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology, Magdeburger Str. 2, D-06112 Halle, Saale, Germany.
| |
Collapse
|
125
|
Gong H, Kovar JL, Cheung L, Rosenthal EL, Olive DM. A comparative study of affibody, panitumumab, and EGF for near-infrared fluorescence imaging of EGFR- and EGFRvIII-expressing tumors. Cancer Biol Ther 2013; 15:185-93. [PMID: 24100437 DOI: 10.4161/cbt.26719] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Aberrant overexpression and/or activation of epidermal growth factor receptor (EGFR) is associated with many types of cancers. EGFR variant III (EGFRvIII) is a common in-frame deletion mutant, which lacks a large part of the extracellular portion (exons 2-7), including components of the ligand-binding domain. Although EGFR has been extensively studied as a molecular imaging target, information about EGFRvIII-targeted molecular imaging is lacking. In this study, the EGFR-specific affibody, therapeutic antibody panitumumab, and ligand EGF were labeled with IRDye 800CW (Ex/Em: 774/789 nm), yielding Aff800, Pan800, and EGF800, respectively. The binding affinities of the labeled agents were compared in cell-based assays using a rat glioma cell line F98 parental (F98-p) lacking EGFR expression, and 2 F98-derived transgenic cell lines expressing EGFR or EGFRvIII (designated as F98-EGFR and F98-vIII, respectively). Results showed that all agents could bind to F98-EGFR, with Pan800 having the highest binding affinity, followed by Aff800 and EGF800. Pan800 and Aff800, but not EGF800, also bound to F98-vIII. In vivo animal imaging demonstrated that compared with F98-p tumors, F98-EGFR tumors generated higher signals with all three agents. However, in the case of F98-vIII, only Pan800 and Aff800 signals were higher. Analysis of tissue lysates showed that a large portion of Pan800 was degraded into small fragments in F98-EGFR and F98-vIII tumors, possibly due to proteolytic digestion after its specific binding and internalization. In conclusion, Pan800 and Aff800 could be used as imaging agents for both wild-type EGFR and EGFRvIII, whereas EGF800 only targets wild-type EGFR.
Collapse
Affiliation(s)
| | | | | | - Eben L Rosenthal
- Division of Otolaryngology; Head and Neck Surgery; University of Alabama at Birmingham; Birmingham, AL USA
| | | |
Collapse
|
126
|
Abstract
PURPOSE OF REVIEW This review provides a concise summary of significant research progress on SIRT1 deacetylase in leukemia in the past year. SIRT1 is a multifunctional protein and recent studies demonstrate that SIRT1 plays a crucial role in myeloid leukemogenesis and drug resistance. RECENT FINDINGS SIRT1 expression is typically low in normal adult hematopoietic stem/progenitor cells, but is increased in the leukemic stem/progenitor cells of chronic myeloid leukemia (CML). SIRT1 activation is mediated in both BCR-ABL tyrosine kinase-dependent and independent manners. SIRT1 activation promotes resistance of CML stem cells to tyrosine kinase inhibitors and acquisition of BCR-ABL mutations for acquired resistance. SUMMARY On the basis of current findings, SIRT1 inhibition in combination with BCR-ABL tyrosine kinase inhibitors can be explored as a novel approach to eradicate leukemic stem cells and residual disease in chronic phase CML. SIRT1 inhibition may also help prevent acquired resistance through genetic mutations of advanced phases of CML, and extend remission.
Collapse
|
127
|
Differences in the binding affinities of ErbB family: heterogeneity in the prediction of resistance mutants. PLoS One 2013; 8:e77054. [PMID: 24194858 PMCID: PMC3806757 DOI: 10.1371/journal.pone.0077054] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 08/24/2013] [Indexed: 11/25/2022] Open
Abstract
The pressure exerted by drugs targeted to a protein in any therapy inevitably leads to the emergence of drug resistance. One major mechanism of resistance involves the mutation of key residues in the target protein. Drugs that competitively replace a natural substrate are often made ineffective by mutations that reduce the drug’s affinity relative to that of the natural substrate. Hence atomic level understanding of the mechanisms that underlie this behavior is of utmost importance in efforts to design new drugs that can target such mutant proteins. Methods that can predict these mutations before they appear in clinic would be a major advance in the selection of the appropriate treatment strategy in patients. The present computational approach aims to model this emergence in EGFR and ErbB2 after treatment with the drug lapatinib, by investigating the structural, dynamic and energetic effects on these kinases when bound to the natural substrate ATP and to lapatinib. The study reveals binding modes and subpopulations that are presumably normally cryptic and these have been analyzed extensively here with respect to sites that are predicted to be hotspots for resisting mutations. These positions are compared in the context of currently available data from laboratory-based experiments and mechanistic details, at the atomistic level, of the origin of resistance are developed. The prediction of novel mutations, if validated by their emergence in the clinic, will make these methods as a powerful predictive tool which can be used in the design of new kinase inhibitors.
Collapse
|
128
|
Shiraishi K, Mimura K, Izawa S, Inoue A, Shiba S, Maruyama T, Watanabe M, Kawaguchi Y, Inoue M, Fujii H, Kono K. Lapatinib acts on gastric cancer through both antiproliferative function and augmentation of trastuzumab-mediated antibody-dependent cellular cytotoxicity. Gastric Cancer 2013. [PMID: 23187882 DOI: 10.1007/s10120-012-0219-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Trastuzumab has been recently approved for clinical use to treat HER2-expressing advanced gastric cancer, and anti-HER2-targeting therapy has become a promising option for gastric cancer. Lapatinib is a dual tyrosine kinase inhibitor targeting EGFR and HER2. The aim of the present study was to explore the utility of lapatinib for gastric cancer, with a particular focus on trastuzumab-mediated antibody-dependent cellular cytotoxicity (ADCC). METHODS Nine gastric cancer cell lines were evaluated for the effects of lapatinib on the cell-surface accumulation of HER2 and analyzed for their additional effects on trastuzumab-mediated ADCC. Also, HER2 signaling with Western blot, proliferative function with the MTT assay, and apoptosis-inducing activity with 7ADD/Annexin-V were investigated when a panel of gastric cancer cell lines was treated with lapatinib. RESULTS Lapatinib inhibited HER2 signaling and cell proliferation in the panel of gastric cancer cell lines. Lapatinib also induced the accumulation of HER2 on the cell surface, resulting in the enhancement of trastuzumab-mediated ADCC of gastric cancer. CONCLUSIONS Lapatinib exhibits inhibitory activity in gastric cancer cells, and the combination of lapatinib with trastuzumab may be a promising treatment strategy for gastric cancer patients.
Collapse
Affiliation(s)
- Kensuke Shiraishi
- First Department of Surgery, University of Yamanashi, Yamanashi, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Abstract
Innate resistance to various therapeutic interventions is a hallmark of cancer. In recent years, acquired resistance has emerged as a daunting challenge to targeted cancer therapy, which abolishes the efficacy of otherwise successful targeting drugs. Cancer cells gain the resistance property through a variety of mechanisms in primary and metastatic cancers, involving cellular intrinsic and extrinsic factors. Increasing evidence suggests that the mammalian stress response gene sirtuin 1 (SIRT1) plays a critical role in multiple aspects of cancer drug resistance. SIRT1 decreases drug penetration, confers proliferation and antiapoptotic survival advantages to cancer cells, facilitates acquired resistance through genetic mutations, promotes the survival of cancer stem cells, and changes the tumor microenvironment for resistance in cell-autonomous and -nonautonomous manners. This article provides an overview of research advances in the roles of SIRT1 in cancer drug resistance and highlights the prospect of targeting SIRT1 as a new strategy to overcome cancer drug resistance and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Cancer Biology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | | |
Collapse
|
130
|
Abstract
Cancer is a major devastating disease, and is a leading cause of death worldwide. Despite the progress in cancer treatment, cancer mortality rate remains high. Therefore, the discovery and development of improved anticancer drugs to treat cancer are needed. 4H-chromenes have strong cytotoxicity against a panel of human cancer cell lines involving pathways that include microtubule depolarization and tumor vasculature disruption. A chromene analog, Crolibulin™ (EPC2407) is currently in Phase I/II clinical trials for the treatment of advanced solid tumors. This article reviews the general synthesis, biological activities and structure–activity relatinships of different classes of chromenes.
Collapse
|
131
|
Eifert C, Wang X, Kokabee L, Kourtidis A, Jain R, Gerdes MJ, Conklin DS. A novel isoform of the B cell tyrosine kinase BTK protects breast cancer cells from apoptosis. Genes Chromosomes Cancer 2013; 52:961-75. [PMID: 23913792 DOI: 10.1002/gcc.22091] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 02/03/2023] Open
Abstract
Tyrosine kinases orchestrate key cellular signaling pathways and their dysregulation is often associated with cellular transformation. Several recent cases in which inhibitors of tyrosine kinases have been successfully used as anticancer agents have underscored the importance of this class of proteins in the development of targeted cancer therapies. We have carried out a large-scale loss-of-function analysis of the human tyrosine kinases using RNA interference to identify novel survival factors for breast cancer cells. In addition to kinases with known roles in breast and other cancers, we identified several kinases that were previously unknown to be required for breast cancer cell survival. The most surprising of these was the cytosolic, nonreceptor tyrosine kinase, Bruton's tyrosine kinase (BTK), which has been extensively studied in B cell development. Down regulation of this protein with RNAi or inhibition with pharmacological inhibitors causes apoptosis; overexpression inhibits apoptosis induced by Doxorubicin in breast cancer cells. Our results surprisingly show that BTK is expressed in several breast cancer cell lines and tumors. The predominant form of BTK found in tumor cells is transcribed from an alternative promoter and results in a protein with an amino-terminal extension. This alternate form of BTK is expressed at significantly higher levels in tumorigenic breast cells than in normal breast cells. Since this protein is a survival factor for these cells, it represents both a potential marker and novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Cheryl Eifert
- Department of Biomedical Sciences, Gen*NY*Sis Center for Excellence in Cancer Genomics, University at Albany, Rensselaer, NY
| | | | | | | | | | | | | |
Collapse
|
132
|
Malenfant S, Margaillan G, Loehr JE, Bonnet S, Provencher S. The emergence of new therapeutic targets in pulmonary arterial hypertension: from now to the near future. Expert Rev Respir Med 2013; 7:43-55. [PMID: 23362814 DOI: 10.1586/ers.12.83] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease that pathologically increases pulmonary vascular resistance. Ultimately, this leads to right ventricular failure and premature death. Current therapeutic strategies are mainly designed to induce relaxation of the pulmonary arteries, but are not directly aimed to improve vascular remodeling that characterize PAH. Although these treatments modestly improve patient symptoms, pulmonary hemodynamics and survival, none of them are curative and approximately 15% of patients die within 1 year of medical follow-up despite treatment. Within the last 5 years, tremendous advances in our understanding of the PAH pathophysiology have arisen. These advances have a high potential for the development of better patient care by providing novel therapeutic targets. The goal of this report is to review the current PAH treatments, as well as novel therapies that will pave the future in this devastating disease.
Collapse
Affiliation(s)
- Simon Malenfant
- Pulmonary Hypertension Research Group, Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec G1V 4G5, Canada
| | | | | | | | | |
Collapse
|
133
|
do Carmo A, Balça-Silva J, Matias D, Lopes MC. PKC signaling in glioblastoma. Cancer Biol Ther 2013; 14:287-94. [PMID: 23358475 PMCID: PMC3667867 DOI: 10.4161/cbt.23615] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 01/11/2023] Open
Abstract
Glioblastoma Multiforme (GBM) is the most aggressive brain tumor characterized by intratumoral heterogeneity at cytopathological, genomic and transcriptional levels. Despite the efforts to develop new therapeutic strategies the median survival of GBM patients is 12-14 months. Results from large-scale gene expression profile studies confirmed that the genetic alterations in GBM affect pathways controlling cell cycle progression, cellular proliferation and survival and invasion ability, which may explain the difficulty to treat GBM patients. One of the signaling pathways that contribute to the aggressive behavior of glioma cells is the protein kinase C (PKC) pathway. PKC is a family of serine/threonine-specific protein kinases organized into three groups according the activating domains. Due to the variability of actions controlled by PKC isoforms, its contribution to the development of GBM is poorly understood. This review intends to highlight the contribution of PKC isoforms to proliferation, survival and invasive ability of glioma cells.
Collapse
Affiliation(s)
- Anália do Carmo
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | | | | | | |
Collapse
|
134
|
A survey of marine natural compounds and their derivatives with anti-cancer activity reported in 2011. Molecules 2013; 18:3641-73. [PMID: 23529027 PMCID: PMC6270579 DOI: 10.3390/molecules18043641] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 12/13/2022] Open
Abstract
Cancer continues to be a major public health problem despite the efforts that have been made in the search for novel drugs and treatments. The current sources sought for the discovery of new molecules are plants, animals and minerals. During the past decade, the search for anticancer agents of marine origin to fight chemo-resistance has increased greatly. Each year, several novel anticancer molecules are isolated from marine organisms and represent a renewed hope for cancer therapy. The study of structure-function relationships has allowed synthesis of analogues with increased efficacy and less toxicity. In this report, we aim to review 42 compounds of marine origin and their derivatives that were published in 2011 as promising anticancer compounds.
Collapse
|
135
|
Wainberg ZA, Anghel A, Rogers AM, Desai AJ, Kalous O, Conklin D, Ayala R, O'Brien NA, Quadt C, Akimov M, Slamon DJ, Finn RS. Inhibition of HSP90 with AUY922 induces synergy in HER2-amplified trastuzumab-resistant breast and gastric cancer. Mol Cancer Ther 2013; 12:509-19. [PMID: 23395886 DOI: 10.1158/1535-7163.mct-12-0507] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HSP90 enables the activation of many client proteins of which the most clinically validated is HER2. NVP-AUY922, a potent HSP90 inhibitor, is currently in phase II clinical trials. To explore its potential clinical use in HER2-amplified breast and gastric cancers, we evaluated the effect of AUY922 alone and in combination with trastuzumab in both trastuzumab-sensitive and -resistant models. A panel of 16 human gastric and 45 breast cancer cell lines, including 16 HER2-amplified (3 and 13, respectively) cells, was treated with AUY922 over various concentrations. In both breast and gastric cancer, we used cell lines and xenograft models with conditioned trastuzumab-resistance to investigate the efficacy of AUY922 alongside trastuzumab. Effects of this combination on downstream markers were analyzed via Western blot analysis. AUY922 exhibited potent antiproliferative activity in the low nanomolar range (<40 nmol/L) for 59 of 61 cell lines. In both histologies, HER2-amplified cells expressed greater sensitivity to AUY than HER2-negative cells. In conditioned trastuzumab-resistant models, AUY922 showed a synergistic effect with trastuzumab. In vitro, the combination induced greater decreases in HER2, a G2 cell-cycle arrest, and increased apoptosis. In a trastuzumab-resistant gastric cancer xenograft model, the combination of AUY922 and trastuzumab showed greater antitumor efficacy than either drug alone. These data suggest that AUY922 in combination with trastuzumab has unique efficacy in trastuzumab-resistant models. The combination of HSP90 inhibition and direct HER2 blockade represents a novel approach to the treatment of HER2-amplified cancers and clinical trials based on the above data are ongoing.
Collapse
Affiliation(s)
- Zev A Wainberg
- University of California Geffen School of Medicine, Department of Medicine, Division of Hematology/Oncology, Santa Monica, CA 90404, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, Goel N, Aronson AB, Li S, Ma CX, Ding L, Mardis ER, Ellis MJ. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov 2013; 3:224-37. [PMID: 23220880 PMCID: PMC3570596 DOI: 10.1158/2159-8290.cd-12-0349] [Citation(s) in RCA: 651] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
UNLABELLED Data from 8 breast cancer genome-sequencing projects identified 25 patients with HER2 somatic mutations in cancers lacking HER2 gene amplification. To determine the phenotype of these mutations, we functionally characterized 13 HER2 mutations using in vitro kinase assays, protein structure analysis, cell culture, and xenograft experiments. Seven of these mutations are activating mutations, including G309A, D769H, D769Y, V777L, P780ins, V842I, and R896C. HER2 in-frame deletion 755-759, which is homologous to EGF receptor (EGFR) exon 19 in-frame deletions, had a neomorphic phenotype with increased phosphorylation of EGFR or HER3. L755S produced lapatinib resistance, but was not an activating mutation in our experimental systems. All of these mutations were sensitive to the irreversible kinase inhibitor, neratinib. These findings show that HER2 somatic mutation is an alternative mechanism to activate HER2 in breast cancer and they validate HER2 somatic mutations as drug targets for breast cancer treatment. SIGNIFICANCE We show that the majority of HER2 somatic mutations in breast cancer patients are activating mutations that likely drive tumorigenesis. Several patients had mutations that are resistant to the reversible HER2 inhibitor lapatinib, but are sensitive to the irreversible HER2 inhibitor, neratinib. Our results suggest that patients with HER2 mutation–positive breast cancers could benefit from existing HER2-targeted drugs.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antineoplastic Agents/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Amplification
- Gene Expression
- Humans
- Lapatinib
- MCF-7 Cells
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Nude
- Models, Molecular
- Molecular Sequence Data
- Mutation
- NIH 3T3 Cells
- Protein Structure, Tertiary
- Quinazolines/pharmacology
- Quinolines/pharmacology
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Amino Acid
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Ron Bose
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Hacker SM, Hardt N, Buntru A, Pagliarini D, Möckel M, Mayer TU, Scheffner M, Hauck CR, Marx A. Fingerprinting differential active site constraints of ATPases. Chem Sci 2013. [DOI: 10.1039/c3sc21916j] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
138
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
139
|
Beltran L, Casado P, Rodríguez-Prados JC, Cutillas PR. Global profiling of protein kinase activities in cancer cells by mass spectrometry. J Proteomics 2012; 77:492-503. [DOI: 10.1016/j.jprot.2012.09.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/03/2012] [Accepted: 09/24/2012] [Indexed: 01/15/2023]
|
140
|
Multiple receptor tyrosine kinases promote the in vitro phenotype of metastatic human osteosarcoma cell lines. Oncogenesis 2012; 1:e34. [PMID: 23552467 PMCID: PMC3511679 DOI: 10.1038/oncsis.2012.34] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The survival rate for osteosarcoma patients with localized disease is 70% and only 25% for patients with metastases. Therefore, novel therapeutic and prognostic tools are needed. In this study, extensive screening and validation strategies identified Axl, EphB2, FGFR2, IGF-1R and Ret as specific receptor tyrosine kinases (RTKs) that are activated and promote the in vitro phenotype of two genetically different metastatic osteosarcoma cell lines. Initial phosphoproteomic screening identified twelve RTKs that were phosphorylated in 143B and/or LM7 metastatic human osteosarcoma cells. A small interfering RNA (siRNA) screen demonstrated that siRNA pools targeting ten of the twelve RTKS inhibited the in vitro phenotype of one or both cell lines. To validate the results, we individually tested the four siRNA duplexes that comprised each of the effective siRNA pools from the initial screen. The pattern of phenotype inhibition replicated the pattern of mRNA knockdown by the individual duplexes for seven of the ten RTKs, indicating the effects are consistent with on-target silencing. Five of those seven RTKs were further validated using independent approaches including neutralizing antibodies (IGF-1R), antisense-mediated knockdown (EphB2, FGFR2, and Ret) or small molecule inhibitors (Axl), indicating that those specific RTKs promote the in vitro behavior of metastatic osteosarcoma cell lines and are potential therapeutic targets for osteosarcoma. Immunohistochemistry demonstrated that Axl is frequently activated in osteosarcoma patient biopsy samples, further supporting our screening and validation methods to identify RTKs that may be valuable targets for novel therapies for osteosarcoma patients.
Collapse
|
141
|
Björkelund H, Gedda L, Malmqvist M, Andersson K. Resolving the EGF-EGFR interaction characteristics through a multiple-temperature, multiple-inhibitor, real-time interaction analysis approach. Mol Clin Oncol 2012; 1:343-352. [PMID: 24649173 DOI: 10.3892/mco.2012.37] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/19/2012] [Indexed: 01/15/2023] Open
Abstract
Overexpression and aberrant activity of the epidermal growth factor (EGF) have been observed in various cancer types, rendering it an important target in oncology research. The interaction between EGF and its receptor (EGFR), as well as subsequent internalization, is complex and may be affected by various factors including tyrosine kinase inhibitors (TKIs). By combining real-time binding curves produced in LigandTracer® with internalization assays conducted at different temperatures and with different TKIs, the processes of ligand binding, internalization and excretion was visualized. SKOV3 cells had a slower excretion rate compared to A431 and U343 cells, and the tested TKIs (gefitinib, lapatinib, AG1478 and erlotinib) reduced the degree of internalization. The kinetic analysis of the binding curves further demonstrated TKI-dependent balances of EGFR monomer and dimer populations, where lapatinib promoted the monomeric form, while the other TKIs induced dimers. The dimer levels were found to be associated with the apparent affinity of the EGF-EGFR interaction, with EGF binding stronger to EGFR dimers compared to monomers. This study analyzed how real-time molecular interaction analysis may be utilized in combination with perturbations in order to understand the kinetics of a ligand-receptor interaction, as well as some of its associated intracellular processes. Our multiple-temperature and -inhibitor assay setup renders it possible to follow the EGFR monomer, dimer and internalized populations in a detailed manner, allowing for a new perspective of the EGFR biology.
Collapse
Affiliation(s)
- Hanna Björkelund
- Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Sciences, Rudbeck Laboratory, Uppsala University; ; Ridgeview Instruments AB, Uppsala
| | - Lars Gedda
- Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Sciences, Rudbeck Laboratory, Uppsala University; ; Swedish Radiation Safety Authority, Stockholm
| | - Magnus Malmqvist
- Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Sciences, Rudbeck Laboratory, Uppsala University; ; Ridgeview Instruments AB, Uppsala; ; Ridgeview Diagnostics AB c/o Bioventia AB, Uppsala, Sweden
| | - Karl Andersson
- Biomedical Radiation Sciences, Department of Radiology, Oncology and Radiation Sciences, Rudbeck Laboratory, Uppsala University; ; Ridgeview Instruments AB, Uppsala; ; Ridgeview Diagnostics AB c/o Bioventia AB, Uppsala, Sweden
| |
Collapse
|
142
|
Davoodi-Semiromi A, Wasserfall CH, Hassanzadeh A, Cooper-DeHoff RM, Wabitsch M, Atkinson M. Influence of Tyrphostin AG490 on the expression of diabetes-associated markers in human adipocytes. Immunogenetics 2012; 65:83-90. [PMID: 23081744 DOI: 10.1007/s00251-012-0659-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/08/2012] [Indexed: 11/25/2022]
Abstract
Tyrosine kinase inhibitors (TKi) hold promise as a treatment for a variety of disorders ranging from those in oncology to diseases thought as immune mediated. Tyrphostin AG490 is a potent Jak-Stat TKi shown effective in the prevention of allograft transplant rejection, experimental autoimmune disease, as well as the treatment of cancer. However, given its ability to modulate this important but pleiotropic intracellular pathway, we thought that it is important to examine its effects on glucose metabolism and expression of major transcription factors and adipokines associated with insulin insensitivity and diabetes. We investigated the metabolic effects of AG490 on glucose levels in vivo using an animal model of diabetes, nonobese diabetic (NOD) mice, and transcription factor expression through assessment of human adipocytes. AG490 treatment of young nondiabetic NOD mice significantly reduced blood glucose levels (p = 0.002). In vitro, treatment of adipocytes with rosiglitazone, an insulin sensitizer that binds to peroxisome proliferator-activated receptor (PPAR) receptors and increases the adipocyte response to insulin, significantly increased the expression of the antidiabetic adipokine adiponectin. Importantly, the combination of rosiglitazone plus Tyrphostin AG490 further increased this effect and was specifically associated with significant upregulation of C-enhanced binding protein (C/EBP) (p < 0.0001). In terms of the mechanism underlying this action, regulatory regions of the PPARγ, ADIPOQ, and C/EBP contain the Stat5 DNA-binding sequences and were demonstrated, by gel shift experiments in vitro. These data suggest that blocking Jak-Stat signaling with AG490 reduces blood glucose levels and modulates the expression of transcription factors previously associated with diabetes, thereby supporting its potential as a therapy for this disease.
Collapse
Affiliation(s)
- Abdoreza Davoodi-Semiromi
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 33136, USA.
| | | | | | | | | | | |
Collapse
|
143
|
Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res Treat 2012; 136:331-45. [PMID: 23073759 DOI: 10.1007/s10549-012-2289-9] [Citation(s) in RCA: 517] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 10/03/2012] [Indexed: 02/07/2023]
Abstract
Decades of research in molecular oncology have brought about promising new therapies which are designed to target specific molecules which promote tumor growth and survival. The epidermal growth factor receptor (EGFR) is one of the first identified important targets of these novel antitumor agents. Approximately half of cases of triple-negative breast cancer (TNBC) and inflammatory breast cancer (IBC) overexpress EGFR. Thus, EGFR inhibitors for treatment of breast cancer have been evaluated in several studies. However, results so far have been disappointing. One of the reasons for these unexpected results is the lack of biomarkers for predicting which patients are most likely to respond to EGFR inhibitors. Recent studies have shown that EGFR and its downstream pathway regulate epithelial-mesenchymal transition, migration, and tumor invasion and that high EGFR expression is an independent predictor of poor prognosis in IBC. Further, recent studies have shown that targeting EGFR enhances the chemosensitivity of TNBC cells by rewiring apoptotic signaling networks in TNBC. These studies indicate that EGFR-targeted therapy might have a promising role in TNBC and IBC. Further studies of the role of EGFR in TNBC and IBC are needed to better understand the best way to use EGFR-targeted therapy-e.g., as a chemosensitizer or to prevent metastases-to treat these aggressive diseases.
Collapse
|
144
|
|
145
|
Kaneko T, Huang H, Cao X, Li X, Li C, Voss C, Sidhu SS, Li SSC. Superbinder SH2 Domains Act as Antagonists of Cell Signaling. Sci Signal 2012; 5:ra68. [DOI: 10.1126/scisignal.2003021] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
146
|
Galvani E, Peters GJ, Giovannetti E. EGF receptor-targeted therapy in non-small-cell lung cancer: role of germline polymorphisms in outcome and toxicity. Future Oncol 2012; 8:1015-1029. [PMID: 22894673 DOI: 10.2217/fon.12.89] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Conventional chemotherapeutic regimens have limited impact against most solid tumors and deal with significant toxicity. Over the last 10 years, novel anticancer treatments targeting specific molecules or genes involved in cancer progression have been developed to improve outcome and reduce side effects. In particular, the tyrosine kinase inhibitors gefitinib and erlotinib have been approved for the treatment of non-small-cell lung cancer. Their clinical activity has been related to different clinical and biological parameters, such as EGFR-activating mutations. However, not all clinical outcomes, including tolerability, are explained, and the identification/validation of novel biomarkers is a viable area of research. Germline polymorphisms can be easily assessed in blood samples, and candidate polymorphisms in EGFR and ABCG2 have been correlated with outcome and toxicity in non-small-cell lung cancer patients treated with gefitinib or erlotinib. However, differences in study population and design resulted in several controversial findings, while the prognostic versus predictive role of the polymorphisms still needs to be validated within larger prospective studies. More studies on the relationship of the genotype with drug pharmacokinetics and mechanism of action are also warranted. These future studies, as well as further development and application of novel technologies to decipher genetic alterations, might contribute to the validation of selected polymorphisms as molecular markers predictive of drug activity and help in the selection of tyrosine kinase inhibitors best suited to the individual patient.
Collapse
Affiliation(s)
- Elena Galvani
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | | | | |
Collapse
|
147
|
Fok KL, Chung CM, Yi SQ, Jiang X, Sun X, Chen H, Chen YC, Kung HF, Tao Q, Diao R, Chan H, Zhang XH, Chung YW, Cai Z, Chang Chan H. STK31 maintains the undifferentiated state of colon cancer cells. Carcinogenesis 2012; 33:2044-53. [PMID: 22828137 DOI: 10.1093/carcin/bgs246] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The expression of serine/threonine kinase (STK) family is frequently altered in human cancers. However, the functions of these kinases in cancer development remain elusive. Here, we report that STK31 is robustly and heterogeneously expressed in colon cancer tissues and plays a critical role in determining the differentiation state of colon cancer cells. Knockdown or overexpression of STK31 induced or inhibited differentiation of colon cancer cells, respectively. Deletion of the STK domain abolished the inhibiting effect of STK31. Associated with differentiation, knockdown of STK31 resulted in significant suppression of tumorigenicity both in vitro and in vivo. Genome microarray analysis showed that knockdown of STK31 altered the expression profile of genes that are known to be involved in germ cell and cancer differentiation. Taken together, these results suggest that STK31 is able to control the differentiation state of colon cancer cells, which critically depends on its STK domain. The present findings may shed light on the new therapeutic approach against cancer by targeting STK31 and cancer differentiation.
Collapse
Affiliation(s)
- Kin Lam Fok
- Epithelial Cell Biology Research Centre The Chinese University of Hong Kong Hong Kong SAR
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Napolitano C, Palwai VR, Eriksson LA, Murphy PV. Synthesis, kinase activity and molecular modeling of a resorcylic acid lactone incorporating an amide and a trans-enone in the macrocycle. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.04.082] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
149
|
Carrión-Salip D, Panosa C, Menendez JA, Puig T, Oliveras G, Pandiella A, De Llorens R, Massaguer A. Androgen-independent prostate cancer cells circumvent EGFR inhibition by overexpression of alternative HER receptors and ligands. Int J Oncol 2012; 41:1128-38. [PMID: 22684500 DOI: 10.3892/ijo.2012.1509] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 05/04/2012] [Indexed: 11/05/2022] Open
Abstract
The deregulation of the epidermal growth factor receptor (EGFR) pathway plays a major role in the pathogenesis of prostate cancer (PCa). However, therapies targeting EGFR have demonstrated limited effectiveness in PCa. A potential mechanism to overcome EGFR blockade in cancer cells is the autocrine activation of alternative receptors of the human EGFR (HER) family through the overexpression of the HER receptors and ligands. In the present study, we were interested in analyzing if this intrinsic resistance mechanism might contribute to the inefficacy of EGFR inhibitors in PCa. To this end, we selected two androgen-independent human prostate carcinoma cell lines (DU145 and PC3) and established DU145 erlotinib-resistant cells (DUErR). Cells were treated with three EGFR inhibitors (cetuximab, gefinitib and erlotinib) and the sensitivity to each treatment was assessed. The gene expression of the four EGFR/HER receptors and seven ligands of the HER family was analyzed by real-time PCR prior to and after each treatment. The receptors expression and activation were further characterized by flow cytometry and western blot analysis. EGFR inhibition rapidly induced enhanced gene expression of the EGF, betacellulin and neuregulin-1 ligands along with HER2, HER3 and HER4 receptors in the DU145 cells. In contrast, slight changes were observed in the PC3 cells, which are defective in the phosphatase and tensin homolog (PTEN) tumor suppressor gene. In the erlotinib-resistant DUErR cells, the expression of HER2 and HER3 was increased at mRNA and protein levels together with neuregulin-1, leading to enhanced HER3 phosphorylation and the activation of the downstream PI3K/Akt survival pathway. HER3 blockage by a monoclonal antibody restored the cytostatic activity of erlotinib in DUErR cells. Our results confirm that the overexpression and autocrine activation of HER3 play a key role in mediating the resistance to EGFR inhibitors in androgen-independent PCa cells.
Collapse
Affiliation(s)
- Dolors Carrión-Salip
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona 17071, Spain
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Davoodi-Semiromi A, Hassanzadeh A, Wasserfall CH, Droney A, Atkinson M. Tyrphostin AG490 agent modestly but significantly prevents onset of type 1 in NOD mouse; implication of immunologic and metabolic effects of a Jak-Stat pathway inhibitor. J Clin Immunol 2012; 32:1038-47. [PMID: 22661285 DOI: 10.1007/s10875-012-9707-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 05/11/2012] [Indexed: 01/06/2023]
Abstract
Previously, we have reported that the Jak-Stat signaling pathway is defective in NOD mice. In this study, prediabetic female NOD mice (4 weeks) were treated by intraperitoneal injection either with AG490 or DMSO three times per week for 4 consecutive weeks, followed by once a week for an additional 6 weeks. The onset of diabetes was attenuated in NOD mice treated with AG490 relative to DMSO treated control mice (p < 0.02). From an immunological standpoint, AG490 induced the expression of Foxp3 in CD4(+)CD25(-) T-cells and down-regulated expression of co-stimulatory molecules in dendritic cells (DC) both in vitro and in vivo. AG490 treated CD4+CD25- T-cells and DC in vitro, acquired regulatory functions; namely, the ability to suppress proliferation of a responding cell population in vitro. AG490 treatment resulted in significant reduction of blood glucose values and increased expression of PPARγ in splenocytes and markedly increased expression PPARγ2 but not PPARγ1 in adipocyte in vitro. Presence of multiple Stat5 DNA binding consensus sequences within the promoter region of the PPARγ gene in human and in mouse suggests that PPARγ is downstream to the Jak-Stat signaling pathway. This study highlights a critical role of the Jak-Stat signaling pathway in the pathogenesis of T1D and suggests that blocking the Jak-Stat signaling pathway by AG490 as a tyrosine kinase inhibitor may provide an effective means for preventing autoimmune T1D via both immunological and metabolic effects.
Collapse
Affiliation(s)
- Abdoreza Davoodi-Semiromi
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, Gainesville, FL, USA.
| | | | | | | | | |
Collapse
|