1
|
Astrain-Redin N, Sanmartin C, Sharma AK, Plano D. From Natural Sources to Synthetic Derivatives: The Allyl Motif as a Powerful Tool for Fragment-Based Design in Cancer Treatment. J Med Chem 2023; 66:3703-3731. [PMID: 36858050 PMCID: PMC10041541 DOI: 10.1021/acs.jmedchem.2c01406] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Since the beginning of history, natural products have been an abundant source of bioactive molecules for the treatment of different diseases, including cancer. Many allyl derivatives, which have shown anticancer activity both in vitro and in vivo in a large number of cancers, are bioactive molecules found in garlic, cinnamon, nutmeg, or mustard. In addition, synthetic products containing allyl fragments have been developed showing potent anticancer properties. Of particular note is the allyl derivative 17-AAG, which has been evaluated in Phase I and Phase II/III clinical trials for the treatment of multiple myeloma, metastatic melanoma, renal cancer, and breast cancer. In this Perspective, we compile extensive literature evidence with descriptions and discussions of the most recent advances in different natural and synthetic allyl derivatives that could generate cancer drug candidates in the near future.
Collapse
Affiliation(s)
- Nora Astrain-Redin
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, E-31008 Pamplona, Spain
| | - Carmen Sanmartin
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, E-31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, E-31008 Pamplona, Spain
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, 500 University Drive, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
2
|
Kumar S, Basu M, Ghosh MK. Chaperone-assisted E3 ligase CHIP: A double agent in cancer. Genes Dis 2022; 9:1521-1555. [PMID: 36157498 PMCID: PMC9485218 DOI: 10.1016/j.gendis.2021.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
The carboxy-terminus of Hsp70-interacting protein (CHIP) is a ubiquitin ligase and co-chaperone belonging to Ubox family that plays a crucial role in the maintenance of cellular homeostasis by switching the equilibrium of the folding-refolding mechanism towards the proteasomal or lysosomal degradation pathway. It links molecular chaperones viz. HSC70, HSP70 and HSP90 with ubiquitin proteasome system (UPS), acting as a quality control system. CHIP contains charged domain in between N-terminal tetratricopeptide repeat (TPR) and C-terminal Ubox domain. TPR domain interacts with the aberrant client proteins via chaperones while Ubox domain facilitates the ubiquitin transfer to the client proteins for ubiquitination. Thus, CHIP is a classic molecule that executes ubiquitination for degradation of client proteins. Further, CHIP has been found to be indulged in cellular differentiation, proliferation, metastasis and tumorigenesis. Additionally, CHIP can play its dual role as a tumor suppressor as well as an oncogene in numerous malignancies, thus acting as a double agent. Here, in this review, we have reported almost all substrates of CHIP established till date and classified them according to the hallmarks of cancer. In addition, we discussed about its architectural alignment, tissue specific expression, sub-cellular localization, folding-refolding mechanisms of client proteins, E4 ligase activity, normal physiological roles, as well as involvement in various diseases and tumor biology. Further, we aim to discuss its importance in HSP90 inhibitors mediated cancer therapy. Thus, this report concludes that CHIP may be a promising and worthy drug target towards pharmaceutical industry for drug development.
Collapse
Affiliation(s)
- Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Paraganas, West Bengal 743372, India
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
3
|
Serrano-Del Valle A, Reina-Ortiz C, Benedi A, Anel A, Naval J, Marzo I. Future prospects for mitosis-targeted antitumor therapies. Biochem Pharmacol 2021; 190:114655. [PMID: 34129859 DOI: 10.1016/j.bcp.2021.114655] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/17/2022]
Abstract
Dysregulation of cell cycle progression is a hallmark of cancer cells. In recent years, efforts have been devoted to the development of new therapies that target proteins involved in cell cycle regulation and mitosis. Novel targeted antimitotic drugs include inhibitors of aurora kinase family, polo-like kinase 1, Mps1, Eg5, CENP-5 and the APC/cyclosome complex. While certain new inhibitors reached the clinical trial stage, most were discontinued due to negative results. However, these therapies should not be readily dismissed. Based on recent advances concerning their mechanisms of action, new strategies could be devised to increase their efficacy and promote further clinical trials. Here we discuss three main lines of action to empower these therapeutic approaches: increasing cell death signals during mitotic arrest, targeting senescent cells and facilitating antitumor immune response through immunogenic cell death (ICD).
Collapse
Affiliation(s)
| | - Chantal Reina-Ortiz
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Andrea Benedi
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Alberto Anel
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Javier Naval
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain
| | - Isabel Marzo
- Dept. Biochemistry, Molecular and Cell Biology, University of Zaragoza and IIS Aragón, Spain.
| |
Collapse
|
4
|
Talaei S, Mellatyar H, Asadi A, Akbarzadeh A, Sheervalilou R, Zarghami N. Spotlight on 17-AAG as an Hsp90 inhibitor for molecular targeted cancer treatment. Chem Biol Drug Des 2019; 93:760-786. [PMID: 30697932 DOI: 10.1111/cbdd.13486] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/31/2018] [Accepted: 01/06/2019] [Indexed: 12/11/2022]
Abstract
Hsp90 is a ubiquitous chaperone with important roles in the organization and maturation of client proteins that are involved in the progression and survival of cancer cells. Multiple oncogenic pathways can be affected by inhibition of Hsp90 function through degradation of its client proteins. That makes Hsp90 a therapeutic target for cancer treatment. 17-allylamino-17-demethoxy-geldanamycin (17-AAG) is a potent Hsp90 inhibitor that binds to Hsp90 and inhibits its chaperoning function, which results in the degradation of Hsp90's client proteins. There have been several preclinical studies of 17-AAG as a single agent or in combination with other anticancer agents for a wide range of human cancers. Data from various phases of clinical trials show that 17-AAG can be given safely at biologically active dosages with mild toxicity. Even though 17-AAG has suitable pharmacological potency, its low water solubility and high hepatotoxicity could significantly restrict its clinical use. Nanomaterials-based drug delivery carriers may overcome these drawbacks. In this paper, we review preclinical and clinical research on 17-AAG as a single agent and in combination with other anticancer agents. In addition, we highlight the potential of using nanocarriers and nanocombination therapy to improve therapeutic effects of 17-AAG.
Collapse
Affiliation(s)
- Sona Talaei
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Mellatyar
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asadollah Asadi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roghayeh Sheervalilou
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
5
|
Shevtsov M, Multhoff G. Therapeutic Implications of Heat Shock Proteins in Cancer. HEAT SHOCK PROTEINS 2019. [DOI: 10.1007/978-3-030-02254-9_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
6
|
Jana N, Nanda S. Resorcylic acid lactones (RALs) and their structural congeners: recent advances in their biosynthesis, chemical synthesis and biology. NEW J CHEM 2018. [DOI: 10.1039/c8nj02534g] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Resorcylic acid lactones (RALs) are naturally occurring 14-membered macrolactones that constitute a class of polyketides derived from fungal metabolites and that possess significant and promising biological activity.
Collapse
Affiliation(s)
- Nandan Jana
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur
- India
| | - Samik Nanda
- Department of Chemistry
- Indian Institute of Technology Kharagpur
- Kharagpur
- India
| |
Collapse
|
7
|
Kong F, Zhang H, Qu X, Zhang X, Chen D, Ding R, Mäkilä E, Salonen J, Santos HA, Hai M. Gold Nanorods, DNA Origami, and Porous Silicon Nanoparticle-functionalized Biocompatible Double Emulsion for Versatile Targeted Therapeutics and Antibody Combination Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:10195-10203. [PMID: 27689681 DOI: 10.1002/adma.201602763] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/21/2016] [Indexed: 05/28/2023]
Abstract
Gold nanorods, DNA origami, and porous silicon nanoparticle-functionalized biocompatible double emulsion are developed for versatile molecular targeted therapeutics and antibody combination therapy. This advanced photothermal responsive all-in-one biocompatible platform can be easily formed with great therapeutics loading capacity for different cancer treatments with synergism and multidrug resistance inhibition, which has great potential in advancing biomedical applications.
Collapse
Affiliation(s)
- Feng Kong
- Beijing Key Laboratory of Function Materials for Molecule and Structure Construction, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
| | - Hongbo Zhang
- Beijing Key Laboratory of Function Materials for Molecule and Structure Construction, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Xiangmeng Qu
- Beijing Key Laboratory of Function Materials for Molecule and Structure Construction, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, P. R. China
| | - Xu Zhang
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Cape Breton University, 1250 Grand Lake Road, Sydney, NS, B1P 6L2, Canada
| | - Dong Chen
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Ruihua Ding
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Ermei Mäkilä
- Laboratory of Industrial Physics, Department of Physics, University of Turku, FI-20014, Turku, Finland
| | - Jarno Salonen
- Laboratory of Industrial Physics, Department of Physics, University of Turku, FI-20014, Turku, Finland
| | - Hélder A Santos
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Mingtan Hai
- Beijing Key Laboratory of Function Materials for Molecule and Structure Construction, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, 100083, P. R. China
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
8
|
Cho H, Gao J, Kwon GS. PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels for drug delivery. J Control Release 2016; 240:191-201. [PMID: 26699425 PMCID: PMC4909590 DOI: 10.1016/j.jconrel.2015.12.015] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/07/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Poly(ethylene glycol)-block-poly(D,L-lactic acid) (PEG-b-PLA) micelles and poly(D,L-lactic-co-glycolic acid)-block-polyethylene glycol)-block-poly(D,L-lactic-co-glycolic acid) (PLGA-b-PEG-b-PLGA) sol-gels have been extensively researched for systemic and localized drug delivery applications, respectively, and they have both progressed into humans for paclitaxel, an important yet poorly water-soluble chemotherapeutic agent. In this review article, preclinical and clinical research on PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels that has focused on paclitaxel will be updated, and recent research on other poorly water-soluble anticancer agents and delivery of drug combinations (i.e. multi-drug delivery) that seeks synergistic anticancer efficacy will be summarized. PEG-b-PLA micelles are a first-generation platform for the systemic multi-delivery of poorly water soluble anticancer agents. PLGA-b-PEG-b-PLGA sol-gels are a first-generation platform for the localized multi-drug delivery of water-soluble and/or poorly water-soluble anticancer agents. In summary, PEG-b-PLA micelles and PLGA-b-PEG-b-PLGA sol-gels may safely enable pre-clinical evaluation and clinical translation of poorly water-soluble anticancer agents, especially for promising, rapidly emerging anticancer combinations.
Collapse
Affiliation(s)
- Hyunah Cho
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, 4588 Parkview Place, St. Louis, MO 63110, United States
| | - Jieming Gao
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States; Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul 136-791, Republic of Korea.
| |
Collapse
|
9
|
|
10
|
Pradhan R, Ramasamy T, Choi JY, Kim JH, Poudel BK, Tak JW, Nukolova N, Choi HG, Yong CS, Kim JO. Hyaluronic acid-decorated poly(lactic-co-glycolic acid) nanoparticles for combined delivery of docetaxel and tanespimycin. Carbohydr Polym 2015; 123:313-23. [DOI: 10.1016/j.carbpol.2015.01.064] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 01/27/2015] [Accepted: 01/28/2015] [Indexed: 11/28/2022]
|
11
|
Wang B, Chen L, Ni Z, Dai X, Qin L, Wu Y, Li X, Xu L, Lian J, He F. Hsp90 inhibitor 17-AAG sensitizes Bcl-2 inhibitor (-)-gossypol by suppressing ERK-mediated protective autophagy and Mcl-1 accumulation in hepatocellular carcinoma cells. Exp Cell Res 2014; 328:379-87. [DOI: 10.1016/j.yexcr.2014.08.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/19/2014] [Accepted: 08/24/2014] [Indexed: 01/20/2023]
|
12
|
Chambers JE, Marciniak SJ. Cellular mechanisms of endoplasmic reticulum stress signaling in health and disease. 2. Protein misfolding and ER stress. Am J Physiol Cell Physiol 2014; 307:C657-70. [PMID: 24944205 DOI: 10.1152/ajpcell.00183.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The endoplasmic reticulum (ER) is a major site of protein synthesis, most strikingly in the specialized secretory cells of metazoans, which can produce their own weight in proteins daily. Cells possess a diverse machinery to ensure correct folding, assembly, and secretion of proteins from the ER. When this machinery is overwhelmed, the cell is said to experience ER stress, a result of the accumulation of unfolded or misfolded proteins in the lumen of the organelle. Here we discuss the causes of ER stress and the mechanisms by which cells elicit a response, with an emphasis on recent discoveries.
Collapse
Affiliation(s)
- Joseph E Chambers
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, United Kingdom
| | - Stefan J Marciniak
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, United Kingdom
| |
Collapse
|
13
|
Jhaveri K, Ochiana SO, Dunphy MPS, Gerecitano JF, Corben AD, Peter RI, Janjigian YY, Gomes-DaGama EM, Koren J, Modi S, Chiosis G. Heat shock protein 90 inhibitors in the treatment of cancer: current status and future directions. Expert Opin Investig Drugs 2014; 23:611-28. [PMID: 24669860 PMCID: PMC4161020 DOI: 10.1517/13543784.2014.902442] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Heat shock protein 90 (HSP90) serves as a critical facilitator for oncogene addiction. There has been augmenting enthusiasm in pursuing HSP90 as an anticancer strategy. In fact, since the initial serendipitous discovery that geldanamycin (GM) inhibits HSP90, the field has rapidly moved from proof-of-concept clinical studies with GM derivatives to novel second-generation inhibitors. AREAS COVERED The authors highlight the current status of the second-generation HSP90 inhibitors in clinical development. Herein, the authors note the lessons learned from the completed clinical trials of first- and second-generation inhibitors and describe various assays attempting to serve for a more rational implementation of these agents to cancer treatment. Finally, the authors discuss the future perspectives for this promising class of agents. EXPERT OPINION The knowledge gained thus far provides perhaps only a glimpse at the potential of HSP90 for which there is still much work to be done. Lessons from the clinical trials suggest that HSP90 therapy would advance at a faster pace if patient selection and tumor pharmacokinetics of these drugs were better understood and applied to their clinical development. It is also evident that combining HSP90 inhibitors with other potent anticancer therapies holds great promise not only due to synergistic antitumor activity but also due to the potential of prolonging or preventing the development of drug resistance.
Collapse
Affiliation(s)
- Komal Jhaveri
- New York University Cancer Institute, NYU Clinical Cancer Center, Division of Hematology/Medical Oncology, NY, USA
| | - Stefan O Ochiana
- Sloan-Kettering Institute, Molecular Pharmacology and Chemistry Program, NY, USA
| | - Mark PS Dunphy
- Memorial Sloan-Kettering Cancer Center, Department of Radiology, NY, USA
| | - John F Gerecitano
- Memorial Sloan-Kettering Cancer Center, Lymphoma Medicine Service, NY, USA
| | - Adriana D Corben
- Memorial Sloan-Kettering Cancer Center, Breast Cancer Medicine Service, NY, USA
| | - Radu I Peter
- Technical University of Cluj-Napoca, Department of Mathematics, Cluj-Napoca, Romania
| | - Yelena Y Janjigian
- Memorial Sloan-Kettering Cancer Center, Gastrointestinal Oncology Service, NY, USA
| | - Erica M Gomes-DaGama
- Sloan-Kettering Institute, Molecular Pharmacology and Chemistry Program, NY, USA
| | - John Koren
- Sloan-Kettering Institute, Molecular Pharmacology and Chemistry Program, NY, USA
| | - Shanu Modi
- Memorial Sloan-Kettering Cancer Center, Breast Cancer Medicine Service, NY, USA
| | - Gabriela Chiosis
- Sloan-Kettering Institute, Molecular Pharmacology and Chemistry Program, NY, USA
- Memorial Sloan-Kettering Cancer Center, Breast Cancer Medicine Service, NY, USA
- Molecular Pharmacology & Chemistry, Sloan-Kettering Institute, Department of Medicine, Breast Cancer Service, Memorial Hospital, Memorial Sloan-Kettering Cancer Center, and Weill Graduate School of Medical Sciences, NY, USA
| |
Collapse
|
14
|
Austreid E, Lonning PE, Eikesdal HP. The emergence of targeted drugs in breast cancer to prevent resistance to endocrine treatment and chemotherapy. Expert Opin Pharmacother 2014; 15:681-700. [PMID: 24579888 DOI: 10.1517/14656566.2014.885952] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Deregulated signaling pathways are associated with resistance to chemotherapy and endocrine treatment, providing a rationale for the implementation of novel targeted therapies in breast cancer therapy. Key molecules targeted therapeutically in ongoing clinical breast cancer trials are phosphoinositide 3-kinase-Akt-mammalian target of rapamycin (mTOR), Src, insulin-like growth factor 1 receptor, heat shock protein-90, histone deacetylases, cyclin-dependent kinases (CDKs), Notch and human epidermal growth factor receptors (HERs). AREAS COVERED This review provides an overview of novel targeted agents currently explored in clinical breast cancer trials and registered in ClinicalTrials.gov. The main focus will be on their ability to prevent or reverse endocrine resistance and chemoresistance in breast cancer. EXPERT OPINION HER2 targeted agents have extended survival substantially, both in the adjuvant and metastatic setting, pointing to a crucial dependency on this pathway in HER2-amplified breast cancer, including drug resistance reversal. While data on mTOR inhibitors are encouraging and preliminary results on CDK4/6 and Src inhibitors exciting, so far other targeted agents have been of limited benefit when added in concert with conventional therapies. Future clinical trials should systematically explore biomarkers and defects in functional gene cascades to identify relevant biological mechanisms to be targeted therapeutically in breast cancer.
Collapse
Affiliation(s)
- Eilin Austreid
- University of Bergen, Department of Clinical Science, Section of Oncology , Bergen , Norway
| | | | | |
Collapse
|
15
|
Jhaveri K, Chandarlapaty S, Lake D, Gilewski T, Robson M, Goldfarb S, Drullinsky P, Sugarman S, Wasserheit-Leiblich C, Fasano J, Moynahan ME, D'Andrea G, Lim K, Reddington L, Haque S, Patil S, Bauman L, Vukovic V, El-Hariry I, Hudis C, Modi S. A phase II open-label study of ganetespib, a novel heat shock protein 90 inhibitor for patients with metastatic breast cancer. Clin Breast Cancer 2013; 14:154-60. [PMID: 24512858 DOI: 10.1016/j.clbc.2013.12.012] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 12/24/2013] [Accepted: 12/26/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND Ganetespib is a small molecule, nongeldanamycin HSP90 inhibitor with potent inhibitory effects on HSP90-dependent oncoproteins of relevance to breast cancer pathogenesis. We therefore tested ganetespib in an unselected cohort of patients with MBC. PATIENTS AND METHODS Patients were treated with single agent ganetespib at 200 mg/m(2) once weekly for 3 weeks, on a 28-day cycle. Therapy was continued until disease progression. The primary end point was ORR using Reponse Evaluation Criteria in Solid Tumors version 1.1. RESULTS Twenty-two patients were enrolled with a median age of 51(range, 38-70) years and a median Eastern Cooperative Oncology Group performance status of 0 (range, 0-1). Most patients had at least 2 previous lines of chemotherapy in the metastatic setting. Most common toxicities, largely grade 1/2, were diarrhea, fatigue, nausea, and hypersensitivity reaction. The ORR in this unselected population was 9%, with all responses coming from the subset of patients with HER2-positive MBC (2/13; 15%). One patient with TNBC had objective tumor regression in the lung metastases. The clinical benefit rate (complete response + partial response + stable disease > 6 months) was 9%, median progression-free survival was 7 weeks (95% confidence interval [CI], 7-19), and median overall survival was 46 weeks (95% CI, 27-not applicable). CONCLUSION The study did not meet the prespecified criteria for ORR in the first stage of the Simon 2-stage model in this heavily pretreated unselected population of MBC. However, activity was observed in trastuzumab-refractory HER2-positive and TNBC. Ganetespib was well tolerated and responses in more targeted populations harboring specific HSP90-dependent oncoproteins justifies its further study, particularly as part of rational combinations.
Collapse
Affiliation(s)
- Komal Jhaveri
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sarat Chandarlapaty
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Diana Lake
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Teresa Gilewski
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Mark Robson
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Shari Goldfarb
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | | | - Steven Sugarman
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | | | - Julie Fasano
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Mary Ellen Moynahan
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Gabriella D'Andrea
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Kristina Lim
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | | | - Sofia Haque
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sujata Patil
- Memorial Sloan-Kettering Cancer Center, New York, NY
| | | | | | | | - Clifford Hudis
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College
| | - Shanu Modi
- Memorial Sloan-Kettering Cancer Center, New York, NY; Weil Cornell Medical College.
| |
Collapse
|
16
|
Schenk E, Hendrickson AEW, Northfelt D, Toft DO, Ames MM, Menefee M, Satele D, Qin R, Erlichman C. Phase I study of tanespimycin in combination with bortezomib in patients with advanced solid malignancies. Invest New Drugs 2013; 31:1251-6. [PMID: 23543109 PMCID: PMC3929968 DOI: 10.1007/s10637-013-9946-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/01/2013] [Indexed: 10/27/2022]
Abstract
PURPOSE To determine the maximum tolerated dose (MTD) and characterize the dose-limiting toxicities (DLT) of tanespimycin when given in combination with bortezomib. EXPERIMENTAL DESIGN Phase I dose-escalating trial using a standard cohort "3+3" design performed in patients with advanced solid tumors. Patients were given tanespimycin and bortezomib twice weekly for 2 weeks in a 3 week cycle (days 1, 4, 8, 11 every 21 days). RESULTS Seventeen patients were enrolled in this study, fifteen were evaluable for toxicity, and nine patients were evaluable for tumor response. The MTD was 250 mg/m(2) of tanespimycin and 1.0 mg/m(2) of bortezomib when used in combination. DLTs of abdominal pain (13 %), complete atrioventricular block (7 %), fatigue (7 %), encephalopathy (7 %), anorexia (7 %), hyponatremia (7 %), hypoxia (7 %), and acidosis (7 %) were observed. There were no objective responses. One patient had stable disease. CONCLUSIONS The recommended phase II dose for twice weekly 17-AAG and PS341 are 250 mg/m(2) and 1.0 mg/m(2), respectively, on days 1, 4, 8 and 11 of a 21 day cycle.
Collapse
Affiliation(s)
- Erin Schenk
- Division of Medical Oncology, Mayo Clinic College of Medicine, Mayo Clinic, Gonda 19, 200 First Street, S.W, Rochester, MN, 55905, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
A rat retinal damage model predicts for potential clinical visual disturbances induced by Hsp90 inhibitors. Toxicol Appl Pharmacol 2013; 273:401-9. [PMID: 24090817 DOI: 10.1016/j.taap.2013.09.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/20/2013] [Accepted: 09/21/2013] [Indexed: 11/22/2022]
Abstract
In human trials certain heat shock protein 90 (Hsp90) inhibitors, including 17-DMAG and NVP-AUY922, have caused visual disorders indicative of retinal dysfunction; others such as 17-AAG and ganetespib have not. To understand these safety profile differences we evaluated histopathological changes and exposure profiles of four Hsp90 inhibitors, with or without clinical reports of adverse ocular effects, using a rat retinal model. Retinal morphology, Hsp70 expression (a surrogate marker of Hsp90 inhibition), apoptotic induction and pharmacokinetic drug exposure analysis were examined in rats treated with the ansamycins 17-DMAG and 17-AAG, or with the second-generation compounds NVP-AUY922 and ganetespib. Both 17-DMAG and NVP-AUY922 induced strong yet restricted retinal Hsp70 up-regulation and promoted marked photoreceptor cell death 24h after the final dose. In contrast, neither 17-AAG nor ganetespib elicited photoreceptor injury. When the relationship between drug distribution and photoreceptor degeneration was examined, 17-DMAG and NVP-AUY922 showed substantial retinal accumulation, with high retina/plasma (R/P) ratios and slow elimination rates, such that 51% of 17-DMAG and 65% of NVP-AUY922 present at 30 min post-injection were retained in the retina 6h post-dose. For 17-AAG and ganetespib, retinal elimination was rapid (90% and 70% of drugs eliminated from the retina at 6h, respectively) which correlated with lower R/P ratios. These findings indicate that prolonged inhibition of Hsp90 activity in the eye results in photoreceptor cell death. Moreover, the results suggest that the retina/plasma exposure ratio and retinal elimination rate profiles of Hsp90 inhibitors, irrespective of their chemical class, may predict for ocular toxicity potential.
Collapse
|
18
|
Lee JC, Chou LC, Lien JC, Wu JC, Huang CH, Chung CH, Lee FY, Huang LJ, Kuo SC, Way TD. CLC604 preferentially inhibits the growth of HER2-overexpressing cancer cells and sensitizes these cells to the inhibitory effect of Taxol in vitro and in vivo. Oncol Rep 2013; 30:1762-72. [PMID: 23900492 DOI: 10.3892/or.2013.2634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/25/2013] [Indexed: 11/06/2022] Open
Abstract
HER2 has become a solicitous therapeutic target in metastatic and clinical drug-resistant cancer. Here, we evaluated whether or not 1-benzyl-3-(5-hydroxymethyl-2-furyl)indazole (YC-1) and its furopyrazole and thienopyrazole analogues repress the expression of the HER2 protein. Among the test compounds, (1-benzyl-3-(p-hydroxymethylphenyl)-5-methylfuro[3,2-c]pyrazol) (CLC604), an isosteric analogue of YC-1, significantly suppressed the expression of HER2, and preferentially inhibited cell proliferation and induced apoptosis in HER2-overexpressing cancer cells. Our results revealed that CLC604 reduced HER2 expression through a post-transcriptional mechanism and involvement of proteasomal activity. CLC604 disrupted the association of 90-kDa heat shock protein (Hsp90) with HER2 resulting from the inhibition of Hsp90 ATPase activity. Moreover, we found that CLC604 significantly enhanced the antitumor efficacy of clinical drugs against HER2-overexpressing tumors and efficiently reduced HER2-induced drug resistance in vitro and in vivo. These findings suggest that CLC604 should be developed further as a novel antitumor drug candidate for the treatment of drug-resistant cancer.
Collapse
Affiliation(s)
- Jang-Chang Lee
- Graduate Institute of Pharmaceutical Chemistry, College of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Zagouri F, Sergentanis TN, Chrysikos D, Papadimitriou CA, Dimopoulos MA, Psaltopoulou T. Hsp90 inhibitors in breast cancer: a systematic review. Breast 2013; 22:569-78. [PMID: 23870456 DOI: 10.1016/j.breast.2013.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 05/26/2013] [Accepted: 06/07/2013] [Indexed: 10/26/2022] Open
Abstract
PURPOSE Pharmacological inhibition of Hsp90 shows great promise in breast cancer treatment. This is the first systematic review to synthesize all available data and to evaluate the efficacy and safety of Hsp90 inhibitors in breast cancer. METHODS This study was performed in accordance with the PRISMA guidelines. Eligible articles were identified by a search of MEDLINE and ClinicalTrials.gov databases, using a predefined combination of the terms "breast", "cancer", "Hsp90", "inhibitors". RESULTS Overall, 19 articles (190 patients) were eligible. The greatest clinical activity has been observed on the field of HER2-positive metastatic breast cancer. However, accumulating data suggest that Hsp90 inhibitors may play a significant role in the treatment of triple negative and aromatase inhibitor-resistant breast cancer. CONCLUSION In the last decade, the development of Hsp90 inhibitors has moved forward rapidly; however, no phase III trials have been conducted and none agent has been approved for use in the clinical practice.
Collapse
Affiliation(s)
- Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, Medical School, University of Athens, Vas Sofias Ave & Lourou Str, Athens 11521, Greece.
| | | | | | | | | | | |
Collapse
|
20
|
Krishnamoorthy GP, Guida T, Alfano L, Avilla E, Santoro M, Carlomagno F, Melillo RM. Molecular mechanism of 17-allylamino-17-demethoxygeldanamycin (17-AAG)-induced AXL receptor tyrosine kinase degradation. J Biol Chem 2013; 288:17481-94. [PMID: 23629654 PMCID: PMC3682548 DOI: 10.1074/jbc.m112.439422] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The receptor tyrosine kinase AXL is overexpressed in many cancer types including thyroid carcinomas and has well established roles in tumor formation and progression. Proper folding, maturation, and activity of several oncogenic receptor tyrosine kinases require HSP90 chaperoning. HSP90 inhibition by the antibiotic geldanamycin or its derivative 17-allylamino-17-demethoxygeldanamycin (17-AAG) causes destabilization of its client proteins. Here we show that AXL is a novel client protein of HSP90. 17-AAG induced a time- and dose-dependent down-regulation of endogenous or ectopically expressed AXL protein, thereby inhibiting AXL-mediated signaling and biological activity. 17-AAG-induced AXL down-regulation specifically affected fully glycosylated mature receptor present on cell membrane. By using biotin and [(35)S]methionine labeling, we showed that 17-AAG caused depletion of membrane-localized AXL by mediating its degradation in the intracellular compartment, thus restricting its exposure on the cell surface. 17-AAG induced AXL polyubiquitination and subsequent proteasomal degradation; under basal conditions, AXL co-immunoprecipitated with HSP90. Upon 17-AAG treatment, AXL associated with the co-chaperone HSP70 and the ubiquitin E3 ligase carboxyl terminus of HSC70-interacting protein (CHIP). Overexpression of CHIP, but not of the inactive mutant CHIP K30A, induced accumulation of AXL polyubiquitinated species upon 17-AAG treatment. The sensitivity of AXL to 17-AAG required its intracellular domain because an AXL intracellular domain-deleted mutant was insensitive to the compound. Active AXL and kinase-dead AXL were similarly sensitive to 17-AAG, implying that 17-AAG sensitivity does not require receptor phosphorylation. Overall our data elucidate the molecular basis of AXL down-regulation by HSP90 inhibitors and suggest that HSP90 inhibition in anticancer therapy can exert its effect through inhibition of multiple kinases including AXL.
Collapse
Affiliation(s)
| | - Teresa Guida
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and
| | - Luigi Alfano
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and
| | - Elvira Avilla
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and
| | - Massimo Santoro
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and ,Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche “G. Salvatore,” 80131 Naples, Italy
| | - Francesca Carlomagno
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and ,Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche “G. Salvatore,” 80131 Naples, Italy, To whom correspondence may be addressed: Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Istituto di Endocrinologia ed Oncologia Sperimentale, Via S. Pansini 5, 80131 Naples, Italy. Tel.: 39-0817463603; Fax: 39-0817463603; E-mail:
| | - Rosa Marina Melillo
- From the Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and ,Istituto di Endocrinologia ed Oncologia Sperimentale del Consiglio Nazionale delle Ricerche “G. Salvatore,” 80131 Naples, Italy, To whom correspondence may be addressed: Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Istituto di Endocrinologia ed Oncologia Sperimentale, Via S. Pansini 5, 80131 Naples, Italy. Tel.: 39-0817463603; Fax: 39-0817463603; E-mail:
| |
Collapse
|
21
|
Garon EB, Finn RS, Hamidi H, Dering J, Pitts S, Kamranpour N, Desai AJ, Hosmer W, Ide S, Avsar E, Jensen MR, Quadt C, Liu M, Dubinett SM, Slamon DJ. The HSP90 inhibitor NVP-AUY922 potently inhibits non-small cell lung cancer growth. Mol Cancer Ther 2013; 12:890-900. [PMID: 23493311 PMCID: PMC3681857 DOI: 10.1158/1535-7163.mct-12-0998] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Heat shock protein 90 (HSP90) is involved in protein folding and functions as a chaperone for numerous client proteins, many of which are important in non-small cell lung cancer (NSCLC) pathogenesis. We sought to define preclinical effects of the HSP90 inhibitor NVP-AUY922 and identify predictors of response. We assessed in vitro effects of NVP-AUY922 on proliferation and protein expression in NSCLC cell lines. We evaluated gene expression changes induced by NVP-AUY922 exposure. Xenograft models were evaluated for tumor control and biological effects. NVP-AUY922 potently inhibited in vitro growth in all 41 NSCLC cell lines evaluated with IC50 < 100 nmol/L. IC100 (complete inhibition of proliferation) < 40 nmol/L was seen in 36 of 41 lines. Consistent gene expression changes after NVP-AUY922 exposure involved a wide range of cellular functions, including consistently decreased dihydrofolate reductase after exposure. NVP-AUY922 slowed growth of A549 (KRAS-mutant) xenografts and achieved tumor stability and decreased EGF receptor (EGFR) protein expression in H1975 xenografts, a model harboring a sensitizing and a resistance mutation for EGFR-tyrosine kinase inhibitors in the EGFR gene. These data will help inform the evaluation of correlative data from a recently completed phase II NSCLC trial and a planned phase IB trial of NVP-AUY922 in combination with pemetrexed in NSCLCs.
Collapse
Affiliation(s)
- Edward B Garon
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Katragadda U, Fan W, Wang Y, Teng Q, Tan C. Combined delivery of paclitaxel and tanespimycin via micellar nanocarriers: pharmacokinetics, efficacy and metabolomic analysis. PLoS One 2013; 8:e58619. [PMID: 23505544 PMCID: PMC3591361 DOI: 10.1371/journal.pone.0058619] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/05/2013] [Indexed: 01/13/2023] Open
Abstract
Background Despite the promising anticancer efficacy observed in preclinical studies, paclitaxel and tanespimycin (17-AAG) combination therapy has yielded meager responses in a phase I clinical trial. One serious problem associated with paclitaxel/17-AAG combination therapy is the employment of large quantities of toxic organic surfactants and solvents for drug solubilization. The goal of this study was to evaluate a micellar formulation for the concurrent delivery of paclitaxel and 17-AAG in vivo. Methodology/Principal Findings Paclitaxel/17-AAG-loaded micelles were assessed in mice bearing human ovarian tumor xenografts. Compared with the free drugs at equivalent doses, intravenous administration of paclitaxel/17-AAG-loaded micelles led to 3.5- and 1.7-fold increase in the tumor concentrations of paclitaxel and 17-AAG, respectively, without significant altering drug levels in normal organs. The enhanced tumor accumulation of the micellar drugs was further confirmed by the whole-body near infrared imaging using indocyanine green-labeled micelles. Subsequently, the anticancer efficacy of paclitaxel/17-AAG-loaded micelles was examined in comparison with the free drugs (weekly 20 mg/kg paclitaxel, twice-weekly 37.5 mg/kg 17-AAG). We found that paclitaxel/17-AAG-loaded micelles caused near-complete arrest of tumor growth, whereas the free drug-treated tumors experienced rapid growth shortly after the 3-week treatment period ended. Furthermore, comparative metabolomic profiling by proton nuclear magnetic resonance revealed significant decrease in glucose, lactate and alanine with simultaneous increase in glutamine, glutamate, aspartate, choline, creatine and acetate levels in the tumors of mice treated with paclitaxel/17-AAG-loaded micelles. Conclusions/Significance We have demonstrated in the current wok a safe and efficacious nano-sized formulation for the combined delivery of paclitaxel and 17-AAG, and uncovered unique metabolomic signatures in the tumor that correlate with the favorable therapeutic response to paclitaxel/17-AAG combination therapy.
Collapse
Affiliation(s)
- Usha Katragadda
- Cancer Nanomedicine Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Mercer University, Atlanta, Georgia, United States of America
| | - Wei Fan
- Cancer Nanomedicine Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Mercer University, Atlanta, Georgia, United States of America
| | - Yingzhe Wang
- Cancer Nanomedicine Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Mercer University, Atlanta, Georgia, United States of America
| | - Quincy Teng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States of America
| | - Chalet Tan
- Cancer Nanomedicine Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Mercer University, Atlanta, Georgia, United States of America
- * E-mail: .
| |
Collapse
|
23
|
Abstract
Combination chemotherapies have been a mainstay in the treatment of disseminated malignancies for almost 60 y, yet even successful regimens fail to cure many patients. Although their single-drug components are well studied, the mechanisms by which drugs work together in clinical combination regimens are poorly understood. Here, we combine RNAi-based functional signatures with complementary informatics tools to examine drug combinations. This approach seeks to bring to combination therapy what the knowledge of biochemical targets has brought to single-drug therapy and creates a statistical and experimental definition of "combination drug mechanisms of action." We show that certain synergistic drug combinations may act as a more potent version of a single drug. Conversely, unlike these highly synergistic combinations, most drugs average extant single-drug variations in therapeutic response. When combined to form multidrug regimens, averaging combinations form averaging regimens that homogenize genetic variation in mouse models of cancer and in clinical genomics datasets. We suggest surprisingly simple and predictable combination mechanisms of action that are independent of biochemical mechanism and have implications for biomarker discovery as well as for the development of regimens with defined genetic dependencies.
Collapse
|
24
|
Jhaveri K, Taldone T, Modi S, Chiosis G. Advances in the clinical development of heat shock protein 90 (Hsp90) inhibitors in cancers. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1823:742-55. [PMID: 22062686 PMCID: PMC3288123 DOI: 10.1016/j.bbamcr.2011.10.008] [Citation(s) in RCA: 345] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 10/21/2011] [Accepted: 10/23/2011] [Indexed: 12/11/2022]
Abstract
Hsp90 is an ATP dependent molecular chaperone protein which integrates multiple oncogenic pathways. As such, Hsp90 inhibition is a promising anti-cancer strategy. Several inhibitors that act on Hsp90 by binding to its N-terminal ATP pocket have entered clinical evaluation. Robust pre-clinical data suggested anti-tumor activity in multiple cancer types. Clinically, encouraging results have been demonstrated in melanoma, acute myeloid leukemia, castrate refractory prostate cancer, non-small cell lung carcinoma and multiple myeloma. In breast cancer, proof-of-concept was demonstrated by first generation Hsp90 inhibitors in combination with trastuzumab mainly in human epidermal growth factor receptor 2 (HER2)+metastatic breast cancer. There are a multitude of second generation Hsp90 inhibitors currently under investigation. To date, however, there is no FDA approved Hsp90 inhibitor nor standardized assay to ascertain Hsp90 inhibition. This review summarizes the current status of both first and second generation Hsp90 inhibitors based on their chemical classification and stage of clinical development. It also discusses the pharmacodynamic assays currently implemented in clinic as well as other novel strategies aimed at enhancing the effectiveness of Hsp90 inhibitors. Ultimately, these efforts will aid in maximizing the full potential of this class of agents. This article is part of a Special Issue entitled: Heat Shock Protein 90 (HSP90).
Collapse
Affiliation(s)
- Komal Jhaveri
- Department of Medicine, Breast Cancer Service, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Tony Taldone
- Department of Molecular Pharmacology and Chemistry, Sloan-Kettering Institute, New York, NY
| | - Shanu Modi
- Department of Medicine, Breast Cancer Service, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Gabriela Chiosis
- Department of Medicine, Breast Cancer Service, Memorial Sloan-Kettering Cancer Center, New York, NY
- Department of Molecular Pharmacology and Chemistry, Sloan-Kettering Institute, New York, NY
| |
Collapse
|
25
|
Kwon GS. Polymeric Micelles for Multiple-Drug Delivery. NANOSTRUCTURE SCIENCE AND TECHNOLOGY 2012. [DOI: 10.1007/978-1-4614-2305-8_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
26
|
Doubrovin M, Che JT, Serganova I, Moroz E, Solit DB, Ageyeva L, Kochetkova T, Pillarsetti N, Finn R, Rosen N, Blasberg RG. Monitoring the Induction of Heat Shock Factor 1/Heat Shock Protein 70 Expression following 17-Allylamino-Demethoxygeldanamycin Treatment by Positron Emission Tomography and Optical Reporter Gene Imaging. Mol Imaging 2012. [DOI: 10.2310/7290.2011.00028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Mikhail Doubrovin
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jian T. Che
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Inna Serganova
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ekaterina Moroz
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - David B. Solit
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Lyudmila Ageyeva
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Tatiana Kochetkova
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Nagavarakishore Pillarsetti
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ronald Finn
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Neal Rosen
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ronald G. Blasberg
- From the Departments of Neurology and Radiology, Memorial Hospital; and Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
27
|
HSP90 Inhibitors for Cancer Therapy and Overcoming Drug Resistance. ADVANCES IN PHARMACOLOGY 2012; 65:471-517. [DOI: 10.1016/b978-0-12-397927-8.00015-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Krzykowska-Petitjean K, Małecki J, Bentke A, Ostrowska B, Laidler P. Tipifarnib and tanespimycin show synergic proapoptotic activity in U937 cells. J Cancer Res Clin Oncol 2011; 138:537-44. [PMID: 22209975 PMCID: PMC3278622 DOI: 10.1007/s00432-011-1131-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/20/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND Farnesyltransferase inhibitor tipifarnib (R115777) has been used for treatment of hematological malignancies; however, its observed anticancer effect was limited. This prompted us to search for inhibitors that would show synergic, proapoptotic effect when combined with R115777. We decided to study LY294002, which inhibits PI-3 kinase, and tanespimycin (17AAG), which inhibits Hsp90--a chaperone for a number of proteins, including Akt kinase. METHODS The effect of drugs, used alone or in combination, was tested in U937 cells (human leukemic monocyte lymphoma), which are often used as a model for liquid tumor. The number of viable cells was evaluated with trypan blue staining, while apoptosis was assessed by presence of active caspase-3 and terminal dUTP nick-end labeling of DNA (TUNEL). RESULTS At concentrations in which R115777, LY294002 and 17AAG were only slowing down the proliferation rate, when used separately, the combination of R115777 + LY294002 and R115777 + 17AAG significantly reduced the number of cells and induced cellular apoptosis. CONCLUSIONS Our results suggest that the combination of R115777 + 17AAG could be useful in treating some of the hematological malignancies.
Collapse
|
29
|
Dimopoulos MA, Mitsiades CS, Anderson KC, Richardson PG. Tanespimycin as antitumor therapy. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11:17-22. [PMID: 21454186 DOI: 10.3816/clml.2011.n.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND The 90 kDa heat shock protein (HSP90), which facilitates proper folding and stability of numerous signaling molecules involved in growth control, cell survival, and development, has been implicated in malignant processes. Like its parent compound geldanamycin, tanespimycin binds to HSP90 and causes antineoplastic effects in vitro and in vivo. MATERIALS AND METHODS All relevant published papers identified through searches of PubMed and abstracts from major recent hematology and oncology meetings were reviewed as of October 2009. RESULTS Different formulations and schedules of tanespimycin monotherapy and combination therapy have been tested in several phase I studies in patients with solid tumors or multiple myeloma (MM). No responses have been reported in studies of tanespimycin monotherapy in patients with metastatic melanoma. Tanespimycin given in combination with trastuzumab in patients with metastatic breast cancer induced a partial response in 24% of patients. Single-agent tanespimycin showed activity in MM and in combination with bortezomib, 27% of patients achieved minor response or better (48% bortezomib-naive patients, 22% bortezomib-pretreated patients, 13% bortezomib-refractory patients). CONCLUSION Tanespimycin represents a promising new agent for the treatment of relapsed/refractory MM. Results of ongoing and future trials will determine the role of tanespimycin both in MM and other malignancies, including breast cancer.
Collapse
|
30
|
Mahon KL, Henshall SM, Sutherland RL, Horvath LG. Pathways of chemotherapy resistance in castration-resistant prostate cancer. Endocr Relat Cancer 2011; 18:R103-23. [PMID: 21565970 DOI: 10.1530/erc-10-0343] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chemotherapy remains the major treatment option for castration-resistant prostate cancer (CRPC) and limited cytotoxic options are available. Inherent chemotherapy resistance occurs in half of all patients and inevitably develops even in those who initially respond. Docetaxel has been the mainstay of therapy for 6 years, providing a small survival benefit at the cost of significant toxicity. Cabazitaxel is a promising second-line agent; however, it is no less toxic, whereas mitoxantrone provides only symptomatic benefit. Multiple cellular pathways involving apoptosis, inflammation, angiogenesis, signalling intermediaries, drug efflux pumps and tubulin are implicated in the development of chemoresistance. A thorough understanding of these pathways is needed to identify biomarkers that predict chemotherapy resistance with the aim to avoid unwarranted toxicities in patients who will not benefit from treatment. Until recently, the search for predictive biomarkers has been disappointing; however, the recent discovery of macrophage inhibitory cytokine 1 as a marker of chemoresistance may herald a new era of biomarker discovery in CRPC. Understanding the interface between this complex array of chemoresistance pathways rather than their study in isolation will be required to effectively predict response and target the late stages of advanced disease. The pre-clinical evidence for these resistance pathways and their progress through clinical trials as therapeutic targets is reviewed in this study.
Collapse
Affiliation(s)
- Kate L Mahon
- Department of Medical Oncology, Sydney Cancer Centre, Missenden Road, Camperdown, New South Wales 2050, Australia.
| | | | | | | |
Collapse
|
31
|
Katragadda U, Teng Q, Rayaprolu BM, Chandran T, Tan C. Multi-drug delivery to tumor cells via micellar nanocarriers. Int J Pharm 2011; 419:281-6. [PMID: 21820041 DOI: 10.1016/j.ijpharm.2011.07.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/05/2011] [Accepted: 07/21/2011] [Indexed: 11/26/2022]
Abstract
The aim of this study was to develop micellar nanocarriers for concomitant delivery of paclitaxel and 17-allylamino-17-demethoxygeldanamycin (17-AAG) for cancer therapy. Paclitaxel and 17-AAG were simultaneously loaded into polymeric micelles by a solvent evaporation method. Two candidate nanocarrier constructs, polyethylene glycol-poly(D, L-lactic acid) (PEG-PLA) micelles and PEG-distearoylphosphatidylethanolamine/tocopheryl polyethylene glycol 1000 (PEG-DSPE/TPGS) mixed micelles, were assessed for the release kinetics of the loaded drugs. Compared to PEG-PLA micelles, entrapment of paclitaxel and 17-AAG into PEG-DSPE/TPGS mixed micelles resulted in significantly prolonged release half-lives. The simultaneous incorporation of paclitaxel and 17-AAG into PEG-DSPE/TPGS mixed micelles was confirmed by (1)H NMR analysis. Paclitaxel/17-AAG-loaded PEG-DSPE/TPGS mixed micelles were as effective in blocking the proliferation of human ovarian cancer SKOV-3 cells as the combined free drugs. PEG-DSPE/TPGS mixed micelles may provide a novel and advantageous delivery approach for paclitaxel/17-AAG combination therapy.
Collapse
Affiliation(s)
- Usha Katragadda
- Cancer Nanomedicine Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Mercer University, 3001 Mercer University Drive, Atlanta, GA 30341, USA
| | | | | | | | | |
Collapse
|
32
|
De Witt Hamer PC, Mir SE, Noske D, Van Noorden CJF, Würdinger T. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin Cancer Res 2011; 17:4200-7. [PMID: 21562035 DOI: 10.1158/1078-0432.ccr-10-2537] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
WEE1 kinase is a key molecule in maintaining G₂-cell-cycle checkpoint arrest for premitotic DNA repair. Whereas normal cells repair damaged DNA during G₁-arrest, cancer cells often have a deficient G₁-arrest and largely depend on G₂-arrest. The molecular switch for the G₂-M transition is held by WEE1 and is pushed forward by CDC25. WEE1 is overexpressed in various cancer types, including glioblastoma and breast cancer. Preclinical studies with cancer cell lines and animal models showed decreased cancer cell viability, reduced tumor burden, and improved survival after WEE1 inhibition by siRNA or small molecule inhibitors, which is enhanced by combination with conventional DNA-damaging therapy, such as radiotherapy and/or cytostatics. Mitotic catastrophe results from premature entry into mitosis with unrepaired lethal DNA damage. As such, cancer cells become sensitized to conventional therapy by WEE1 inhibition, in particular those with insufficient G₁-arrest due to deficient p53 signaling, like glioblastoma cells. One WEE1 inhibitor has now reached clinical phase I studies. Dose-limiting toxicity consisted of hematologic events, nausea and/or vomiting, and fatigue. The combination of DNA-damaging cancer therapy with WEE1 inhibition seems to be a rational approach to push cancer cells in mitotic catastrophe. Its safety and efficacy are being evaluated in clinical studies.
Collapse
|
33
|
Perez CA, Santos ES, Arango BA, Raez LE, Cohen EEW. Novel molecular targeted therapies for refractory thyroid cancer. Head Neck 2011; 34:736-45. [PMID: 21544895 DOI: 10.1002/hed.21755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2011] [Indexed: 02/06/2023] Open
Abstract
The incidence of thyroid cancer continues to increase and this neoplasia remains the most common endocrine malignancy. No effective systemic treatment currently exists for iodine-refractory differentiated or medullary thyroid carcinoma, but recent advances in the pathogenesis of these diseases have revealed key targets that are now being evaluated in the clinical setting. RET (rearranged during transfection)/PTC (papillary thyroid carcinoma) gene rearrangements, B-Raf gene mutations, and vascular endothelial growth factor receptor 2 (VEGFR-2) angiogenesis pathways are some of the known genetic alterations playing a crucial role in the development of thyroid cancer. Several novel agents have demonstrated promising responses. Of the treatments studied, multi-kinase inhibitors such as axitinib, sorafenib, motesanib, and XL-184 have shown to be the most effective by inducing clinical responses and stabilizing the disease process. Randomized clinical trials are currently evaluating these agents, results that may soon change the management of thyroid cancer.
Collapse
|
34
|
Allegra A, Sant'antonio E, Penna G, Alonci A, D'Angelo A, Russo S, Cannavò A, Gerace D, Musolino C. Novel therapeutic strategies in multiple myeloma: role of the heat shock protein inhibitors. Eur J Haematol 2010; 86:93-110. [PMID: 21114539 DOI: 10.1111/j.1600-0609.2010.01558.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite advances in understanding the molecular pathogenesis of multiple myeloma and promising new therapies, almost all patients eventually relapse with resistant disease. There is therefore a strong rationale for combining novel therapies that target intrinsic molecular pathways mediating multiple myeloma cell resistance. One such protein family is the heat shock proteins (HSP), especially the HSP90 family. Heat shock protein inhibitors have been identified as promising cancer treatments as, while they only inhibit a single biologic function, the chaperone-protein association, their effect is widespread as it results in the destruction of numerous client proteins. This article reviews the preclinical and clinical data, which support the testing of HSP90 inhibitors as cancer drugs and update the reader on the current status of the ongoing clinical trials of HSP90 inhibitors in multiple myeloma.
Collapse
|
35
|
Sun J, Liu L, Jiang X, Chen D, Huang Y. Therapeutic effects of radiolabeled 17-allylamino-17-demethoxygeldanamycin on human H460 nonsmall-cell lung carcinoma xenografts in mice. Cancer Biother Radiopharm 2010; 25:155-64. [PMID: 20423228 DOI: 10.1089/cbr.2009.0664] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Heat shock protein 90 (HSP90) is an exciting molecular target for cancer therapy because of this protein's key role in oncogenic signaling pathways. In this work, the binding property of 17-allylamino-17-demethoxygeldanamycin (17-AAG), an HSP90 inhibitor now in phase II clinical trials, was used by labeling with radioisotope iodine-131 ((131)I), to observe the potential therapeutic effects on nonsmall-cell lung carcinoma (NSCLC) xenografts. The compound (131)I-17-AAG and BALB/c nude mice bearing H460 human NSCLC xenografts were prepared. Intratumoral and intravenous administration routes were used. The potential effects of labeled 17-AAG were evaluated by biodistribution studies, in vivo imaging, cancer-treatment studies, and histological analysis. Specific tumor uptake of (131)I-17-AAG was achieved in the xenograft models. Compared to intravenous (i.v.) application, tumor uptake was significantly improved with intratumoral injection of (131)I-17-AAG and competitively reduced with preinjection of unlabeled 17-AAG. All treatment groups established tumor-growth inhibition compared with the control group (p < 0.05), with a certain dose-dependent relationship. The 16-day inhibition ratios via intratumoral delivery were better than those via i.v. delivery (p < 0.05). No significant treatment-induced abnormalities appeared in the mice. Compared with the control group, HSP90alpha + expression and Ki-67+ expression of tumor tissues declined after treatments (p < 0.05), which correlated positively with tumor inhibition. Thus, (131)I-17-AAG treatment undoubtedly had inhibitive effects on human NSCLC xenografts in mice, and the combination of radionuclides and HSP90 inhibitors may be considered for cancer therapy.
Collapse
Affiliation(s)
- Jin Sun
- Nuclear Medicine Technology Institute, Southeast University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | |
Collapse
|
36
|
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone involved in the trafficking of proteins in the cell. Under stressful conditions, Hsp90 stabilizes its client proteins and provides protection to the cell against cellular stressors such as in cancer cells. Disruption of Hsp90 leads to client protein degradation and often cell death. As Hsp90 has been found to be either overexpressed or constitutively more active in cancer cells, inhibitors of Hsp90 may have cancer cell selectivity. The N-terminal inhibitors, geldanamycin and radiciol, were the first two described inhibitors of Hsp90, but were not clinically useful. Subsequent analogues-17 allylamino-17demethoxygeldanamycin and 17 dimethylaminoethylamino-17-demethoxygeldanamycin-were found to be more clinically appropriate and have been studied in a number of clinical trials since 1999. In addition, to the N-terminal site of Hsp90, the C-terminal site appears to be another target for inhibition of Hsp90. More recently, inhibitors of the C terminus of Hsp90 have been developed and studied in vitro with promising results.
Collapse
Affiliation(s)
- Jeffrey M Holzbeierlein
- Department of Urology, University of Kansas Hospital, 3901 Rainbow Boulevard, Mail Stop 3016, Kansas, KS, 66160, USA.
| | | | | |
Collapse
|
37
|
Qin Z, DeFee M, Isaacs JS, Parsons C. Extracellular Hsp90 serves as a co-factor for MAPK activation and latent viral gene expression during de novo infection by KSHV. Virology 2010; 403:92-102. [PMID: 20451233 DOI: 10.1016/j.virol.2010.03.052] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 12/16/2009] [Accepted: 03/31/2010] [Indexed: 12/23/2022]
Abstract
The Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), an important cause of morbidity and mortality in immunocompromised patients. KSHV interaction with the cell membrane triggers activation of specific intracellular signal transduction pathways to facilitate virus entry, nuclear trafficking, and ultimately viral oncogene expression. Extracellular heat shock protein 90 localizes to the cell surface (csHsp90) and facilitates signal transduction in cancer cell lines, but whether csHsp90 assists in the coordination of KSHV gene expression through these or other mechanisms is unknown. Using a recently characterized non-permeable inhibitor specifically targeting csHsp90 and Hsp90-specific antibodies, we show that csHsp90 inhibition suppresses KSHV gene expression during de novo infection, and that this effect is mediated largely through the inhibition of mitogen-activated protein kinase (MAPK) activation by KSHV. Moreover, we show that targeting csHsp90 reduces constitutive MAPK expression and the release of infectious viral particles by patient-derived, KSHV-infected primary effusion lymphoma cells. These data suggest that csHsp90 serves as an important co-factor for KSHV-initiated MAPK activation and provide proof-of-concept for the potential benefit of targeting csHsp90 for the treatment or prevention of KSHV-associated illnesses.
Collapse
Affiliation(s)
- Zhiqiang Qin
- Department of Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
38
|
Abstract
Glioblastoma multiforme is the most common primary central nervous system tumor. The prognosis for these malignant brain tumors is poor, with a median survival of 14 months and a 5-year survival rate below 2%. Development of novel treatments is essential to improving survival and quality of life for these patients. Endogenous heat shock proteins have been implicated in mediation of both adaptive and innate immunity, and there is a rising interest in the use of this safe and multifaceted heat shock protein vaccine therapy as a promising treatment for human cancers, including glioblastoma multiforme.
Collapse
Affiliation(s)
- Isaac Yang
- Department of Neurological Surgery, University of California at San Francisco, 505 Parnassus Avenue, Room M779, Campus 0112, San Francisco, CA 94143, USA.
| | | | | |
Collapse
|
39
|
Heat shock proteins, cell survival and drug resistance: the mitochondrial chaperone TRAP1, a potential novel target for ovarian cancer therapy. Gynecol Oncol 2009; 117:177-82. [PMID: 19942270 DOI: 10.1016/j.ygyno.2009.10.078] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/16/2009] [Accepted: 10/02/2009] [Indexed: 12/18/2022]
Abstract
BACKGROUND Protein homeostasis is a highly complex network of molecular interactions governing the health and life span of the organism. Molecular chaperones, mainly heat shock proteins (HSP) and other stress-inducible proteins abundantly expressed in multiple compartments of the cell, are major modulators of protein homeostasis. TRAP1 is a mitochondrial HSP involved in protection against oxidant-induced DNA damage and apoptosis. It was recently described as a component of a mitochondrial pathway selectively up-regulated in tumor cells which antagonizes the proapoptotic activity of cyclophilin D, a mitochondrial permeability transition pore regulator, and is responsible for the maintenance of mitochondrial integrity, thus favoring cell survival. Interestingly, novel TRAP1 antagonists cause sudden collapse of mitochondrial function and selective tumor cell death, suggesting that this pathway may represent a novel molecular target to improve anticancer therapy. Preliminary data suggest that TRAP1 may be a valuable biomarker in ovarian cancers: in fact, TRAP1 levels are significantly higher in cisplatin-resistant ovarian tumors and ovarian carcinoma cell lines. CONCLUSIONS While major advances have been made in understanding the genetics and molecular biology of cancer, given the considerable heterogeneity of ovarian cancer, the introduction of novel targeted therapies and the consequent selection of treatments based on the molecular profile of each tumor may have a major impact on the management of this malignancy and might contribute to building a new era of personalized medicine.
Collapse
|
40
|
Tsaytler PA, Krijgsveld J, Goerdayal SS, Rüdiger S, Egmond MR. Novel Hsp90 partners discovered using complementary proteomic approaches. Cell Stress Chaperones 2009; 14:629-38. [PMID: 19396626 PMCID: PMC2866955 DOI: 10.1007/s12192-009-0115-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 03/27/2009] [Accepted: 04/07/2009] [Indexed: 01/05/2023] Open
Abstract
Hsp90 is an essential eukaryotic molecular chaperone that stabilizes a large set of client proteins, many of which are involved in various cellular signaling pathways. The current list of Hsp90 interactors comprises about 200 proteins and this number is growing steadily. In this paper, we report on the application of three complementary proteomic approaches directed towards identification of novel proteins that interact with Hsp90. These methods are coimmunoprecipitation, pull down with biotinylated geldanamycin, and immobilization of Hsp90beta on sepharose. In all, this study led to the identification of 42 proteins, including 18 proteins that had not been previously characterized as Hsp90 interactors. These novel Hsp90 partners not only represent abundant protein species, but several proteins were identified at low levels, among which signaling kinase Cdk3 and putative transcription factor tripartite motif-containing protein 29. Identification of tetratricopeptide-repeat-containing mitochondrial import receptor protein Tom34 suggests the involvement of Hsp90 in the early steps of translocation of mitochondrial preproteins. Taken together, our data expand the knowledge of the Hsp90 interactome and provide a further step in our understanding of the Hsp90 chaperone system.
Collapse
Affiliation(s)
- Pavel A Tsaytler
- Department of Membrane Enzymology, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3584 CH, The Netherlands.
| | | | | | | | | |
Collapse
|
41
|
Landriscina M, Maddalena F, Laudiero G, Esposito F. Adaptation to oxidative stress, chemoresistance, and cell survival. Antioxid Redox Signal 2009; 11:2701-16. [PMID: 19778285 DOI: 10.1089/ars.2009.2692] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The discovery of some additional properties and functions of reactive oxygen species (ROS), beyond their toxic effects, provides a novel scenario for the molecular basis and cell regulation of several pathophysiologic processes. ROS are generated by redox-sensitive, prosurvival signaling pathways and function as second messengers in the transduction of several extracellular signals. A complex intracellular redox buffering network has developed to adapt and protect cells against the dangerous effects of oxidative stress. However, pathways involved in ROS-adaptive response may also play a critical role in protecting cells against cytotoxic effects of anticancer agents, thus supporting the hypothesis of a correlation between adaptation/resistance to oxidative stress and resistance to anticancer drugs. This review summarizes the main systems involved in the adaptive responses: an overview on the pathophysiologic relevance of mitochondria on redox-sensitive transcription factors and genes and main antioxidant networks in tumor cells is provided. One of the major aims is to highlight the adaptive mechanisms and their interplay in the intricate connection between oncogenic signaling, oxidative stress, and chemoresistance. Clarification of these mechanisms has tremendous application potential, in terms of developing novel molecular-targeted anticancer therapies and innovative strategies for rational combination of these agents with chemotherapeutic or tumor-specific biologic drugs.
Collapse
Affiliation(s)
- Matteo Landriscina
- Clinical Oncology Unit, Department of Medical Sciences, University of Foggia, Foggia, Italy
| | | | | | | |
Collapse
|
42
|
Li Y, Zhang T, Schwartz SJ, Sun D. New developments in Hsp90 inhibitors as anti-cancer therapeutics: mechanisms, clinical perspective and more potential. Drug Resist Updat 2009; 12:17-27. [PMID: 19179103 DOI: 10.1016/j.drup.2008.12.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2008] [Revised: 11/26/2008] [Accepted: 12/04/2008] [Indexed: 12/20/2022]
Abstract
The molecular chaperone Hsp90 (heat shock protein 90) is a promising target in cancer therapy. Preclinical and clinical evaluations of a variety of Hsp90 inhibitors have shown anti-tumor effect as a single agent and in combination with chemotherapy. Current Hsp90 inhibitors are categorized into several classes based on distinct modes of inhibition, including (i) blockade of ATP binding, (ii) disruption of co-chaperone/Hsp90 interactions, (iii) antagonism of client/Hsp90 associations and (iv) interference with post-translational modifications of Hsp90. The different functions of Hsp90 isoforms and the isoform selectivity of drugs need further investigation. The correlation of cell surface Hsp90 with cancer metastasis and the emerging involvement of Hsp90 inhibition in cancer stem cells have become exciting areas that could be exploited. Therefore, the aim of this review is (1) to summarize the up-to-date knowledge of mechanistic studies and clinical prospect of currently available Hsp90 inhibitors, (2) to enhance our perspectives for designing and discovering novel Hsp90 inhibitors, and (3) to provide an insight into less-understood potential of Hsp90 inhibition in cancer therapy.
Collapse
Affiliation(s)
- Yanyan Li
- Department of Pharmaceutical Sciences, University of Michigan, MI 48109, USA
| | | | | | | |
Collapse
|
43
|
Schwock J, Dhani N, Cao MPJ, Zheng J, Clarkson R, Radulovich N, Navab R, Horn LC, Hedley DW. Targeting focal adhesion kinase with dominant-negative FRNK or Hsp90 inhibitor 17-DMAG suppresses tumor growth and metastasis of SiHa cervical xenografts. Cancer Res 2009; 69:4750-9. [PMID: 19458065 DOI: 10.1158/0008-5472.can-09-0454] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Focal adhesion kinase (FAK), a nonreceptor protein tyrosine kinase and key modulator of integrin signaling, is widely expressed in different tissues and cell types. Recent evidence indicates a central function of FAK in neoplasia where the kinase contributes to cell proliferation, resistance to apoptosis and anoikis, invasiveness, and metastasis. FAK, like other signaling kinases, is dependent on the chaperone heat shock protein 90 (Hsp90) for its stability and proper function. Thus, inhibition of Hsp90 might be a way of disrupting FAK signaling and, consequently, tumor progression. FAK is expressed in high-grade squamous intraepithelial lesions and metastatic cervical carcinomas but not in nonneoplastic cervical mucosa. In SiHa, a cervical cancer cell line with characteristics of epithelial-to-mesenchymal transition, the stable expression of dominant-negative FAK-related nonkinase decreases anchorage independence and delays xenograft growth. FAK-related nonkinase as well as the Hsp90 inhibitor 17-dimethylaminoethylamino-17-demethoxygeldanamycin both negatively interfere with FAK signaling and focal adhesion turnover. Short-term 17-dimethylaminoethylamino-17-demethoxygeldanamycin treatment prolongs survival in a SiHa lung metastasis model and chronic administration suppresses tumor growth as well as metastatic spread in orthotopic xenografts. Taken together, our data suggest that FAK is of importance for tumor progression in cervical cancer and that disruption of FAK signaling by Hsp90 inhibition might be an avenue to restrain tumor growth as well as metastatic spread.
Collapse
Affiliation(s)
- Joerg Schwock
- Ontario Cancer Institute, Departments of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Multi-drug loaded polymeric micelles for simultaneous delivery of poorly soluble anticancer drugs. J Control Release 2009; 140:294-300. [PMID: 19409432 DOI: 10.1016/j.jconrel.2009.04.024] [Citation(s) in RCA: 196] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 04/21/2009] [Accepted: 04/22/2009] [Indexed: 11/20/2022]
Abstract
Current clinical and preclinical anticancer formulations are limited by their use of toxic excipients and stability issues upon combining different drug formulations. We have found that poly(ethylene glycol)-block-poly(d,l lactic acid) (PEG-b-PLA) micelles can deliver multiple poorly water-soluble drugs at clinically relevant doses. Paclitaxel (PTX), etoposide (ETO), docetaxel (DCTX) and 17-allylamino-17-demethyoxygeldanamycin (17-AAG) were solubilized individually in PEG-b-PLA micelles. Combinations of PTX/17-AAG, ETO/17-AAG, DCTX/17-AAG and PTX/ETO/17-AAG were also solubilized in PEG-b-PLA micelles. PEG-b-PLA micelles were characterized in terms of drug loading, size, stability and drug release. All anticancer agents in all combinations were all solubilized at the level of mg/mL and were stable for 24 h in the 2- and 3-drug combination PEG-b-PLA micelles. The stability of the 2- and 3-drug combination PEG-b-PLA micelles was due to the presence of 17-AAG. In vitro, t(1/2) values for 2- and 3-drug combination PEG-b-PLA micelles spanned 1-5 h. PEG-b-PLA micelles offer a promising alternative for combination drug therapy without formulation related side effects.
Collapse
|
45
|
Kim YS, Alarcon SV, Lee S, Lee MJ, Giaccone G, Neckers L, Trepel JB. Update on Hsp90 inhibitors in clinical trial. Curr Top Med Chem 2009; 9:1479-92. [PMID: 19860730 PMCID: PMC7241864 DOI: 10.2174/156802609789895728] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Accepted: 09/28/2009] [Indexed: 11/22/2022]
Abstract
Twenty-five years ago the first small molecule inhibitors of Hsp90 were identified. In the intervening years there has been dramatic progress in basic scientific understanding of the Hsp90 chaperone machinery and in the role of Hsp90 in malignancy. The first-in-class Hsp90 inhibitor 17-AAG entered into Phase I clinical trials in 1999. There are now 13 Hsp90 inhibitors in clinical trial, representing multiple drug classes, and hundreds of patients have been treated in adult oncology and pediatric oncology trials. This review will provide an overview of the clinical trial results thus far. In addition, pivotal issues in further development of Hsp90 inhibitors as anticancer drugs will be discussed.
Collapse
Affiliation(s)
- Y. S. Kim
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - S. V. Alarcon
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - S. Lee
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - M.-J. Lee
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - G. Giaccone
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - L. Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - J. B. Trepel
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
46
|
Ordóñez JL, Martins AS, Osuna D, Madoz–Gúrpide J, de Alava E. Targeting sarcomas: therapeutic targets and their rational. Semin Diagn Pathol 2008; 25:304-16. [DOI: 10.1053/j.semdp.2008.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|