1
|
Kohlmann P, Krylov SN, Marchand P, Jose J. FRET Assays for the Identification of C. albicans HSP90-Sba1 and Human HSP90α-p23 Binding Inhibitors. Pharmaceuticals (Basel) 2024; 17:516. [PMID: 38675476 PMCID: PMC11053944 DOI: 10.3390/ph17040516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Heat shock protein 90 (HSP90) is a critical target for anticancer and anti-fungal-infection therapies due to its central role as a molecular chaperone involved in protein folding and activation. In this study, we developed in vitro Förster Resonance Energy Transfer (FRET) assays to characterize the binding of C. albicans HSP90 to its co-chaperone Sba1, as well as that of the homologous human HSP90α to p23. The assay for human HSP90α binding to p23 enables selectivity assessment for compounds aimed to inhibit the binding of C. albicans HSP90 to Sba1 without affecting the physiological activity of human HSP90α. The combination of the two assays is important for antifungal drug development, while the assay for human HSP90α can potentially be used on its own for anticancer drug discovery. Since ATP binding of HSP90 is a prerequisite for HSP90-Sba1/p23 binding, ATP-competitive inhibitors can be identified with the assays. The specificity of binding of fusion protein constructs-HSP90-mNeonGreen (donor) and Sba1-mScarlet-I (acceptor)-to each other in our assay was confirmed via competitive inhibition by both non-labeled Sba1 and known ATP-competitive inhibitors. We utilized the developed assays to characterize the stability of both HSP90-Sba1 and HSP90α-p23 affinity complexes quantitatively. Kd values were determined and assessed for their precision and accuracy using the 95.5% confidence level. For HSP90-Sba1, the precision confidence interval (PCI) was found to be 70-120 (100 ± 20) nM while the accuracy confidence interval (ACI) was 100-130 nM. For HSP90α-p23, PCI was 180-260 (220 ± 40) nM and ACI was 200-270 nM. The developed assays were used to screen a nucleoside-mimetics library of 320 compounds for inhibitory activity against both C. albicans HSP90-Sba1 and human HSP90α-p23 binding. No novel active compounds were identified. Overall, the developed assays exhibited low data variability and robust signal separation, achieving Z factors > 0.5.
Collapse
Affiliation(s)
- Philip Kohlmann
- Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, University of Münster, 48149 Münster, Germany;
| | - Sergey N. Krylov
- Department of Chemistry, York University, Toronto, ON M3J 1P3, Canada;
- Centre for Research on Biomolecular Interactions, York University, Toronto, ON M3J 1P3, Canada
| | - Pascal Marchand
- Cibles et Médicaments des Infections et de l’Immunité, IICiMed, Nantes Université, UR 1155, F-44000 Nantes, France;
| | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, Pharmacampus, University of Münster, 48149 Münster, Germany;
| |
Collapse
|
2
|
Carlson DL, Kowalewski M, Bodoor K, Lietzan AD, Hughes PF, Gooden D, Loiselle DR, Alcorta D, Dingman Z, Mueller EA, Irnov I, Modla S, Chaya T, Caplan J, Embers M, Miller JC, Jacobs-Wagner C, Redinbo MR, Spector N, Haystead TAJ. Targeting Borrelia burgdorferi HtpG with a berserker molecule, a strategy for anti-microbial development. Cell Chem Biol 2024; 31:465-476.e12. [PMID: 37918401 DOI: 10.1016/j.chembiol.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 08/14/2023] [Accepted: 10/06/2023] [Indexed: 11/04/2023]
Abstract
Conventional antimicrobial discovery relies on targeting essential enzymes in pathogenic organisms, contributing to a paucity of new antibiotics to address resistant strains. Here, by targeting a non-essential enzyme, Borrelia burgdorferi HtpG, to deliver lethal payloads, we expand what can be considered druggable within any pathogen. We synthesized HS-291, an HtpG inhibitor tethered to the photoactive toxin verteporfin. Reactive oxygen species, generated by light, enables HS-291 to sterilize Borrelia cultures by causing oxidation of HtpG, and a discrete subset of proteins in proximity to the chaperone. This caused irreversible nucleoid collapse and membrane blebbing. Tethering verteporfin to the HtpG inhibitor was essential, since free verteporfin was not retained by Borrelia in contrast to HS-291. For this reason, we liken HS-291 to a berserker, wreaking havoc upon the pathogen's biology once selectively absorbed and activated. This strategy expands the druggable pathogenic genome and offsets antibiotic resistance by targeting non-essential proteins.
Collapse
Affiliation(s)
- Dave L Carlson
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - Mark Kowalewski
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, 3(rd) Floor, Genetic Medicine Building, Chapel Hill, NC 27599, USA
| | - Khaldon Bodoor
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - Adam D Lietzan
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, The University of North Carolina at Chapel Hill, 385 South Columbia Street, Chapel Hill, NC 27599, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - David Gooden
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - David R Loiselle
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - David Alcorta
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - Zoey Dingman
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - Elizabeth A Mueller
- Sarafan ChEM-H Institute, Stanford University, 290 Jane Stanford Way, Stanford, CA 94035, USA
| | - Irnov Irnov
- Sarafan ChEM-H Institute, Stanford University, 290 Jane Stanford Way, Stanford, CA 94035, USA
| | - Shannon Modla
- Delaware Biotechnology Institute, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Tim Chaya
- Delaware Biotechnology Institute, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Jeffrey Caplan
- Delaware Biotechnology Institute, University of Delaware, 590 Avenue 1743, Newark, DE 19713, USA
| | - Monica Embers
- Department of Microbiology and Immunology, 18703 Three Rivers Road, Covington, LA 70433, USA
| | - Jennifer C Miller
- Galaxy Diagnostics, Inc, P.O. Box 14346 7020 Kit Creek Road, Ste 130, Research Triangle Park, Raliegh, NC 27709, USA
| | - Christine Jacobs-Wagner
- Sarafan ChEM-H Institute, Stanford University, 290 Jane Stanford Way, Stanford, CA 94035, USA; Biology Department, Stanford University, 290 Jane Stanford Way, Stanford, CA 94035, USA; Howard Hughes Medical Institute, Stanford University, 290 Jane Stanford Way, Stanford, CA 94035, USA
| | - Matthew R Redinbo
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, 120 Mason Farm Road, 3(rd) Floor, Genetic Medicine Building, Chapel Hill, NC 27599, USA; Department of Chemistry, University of North Carolina at Chapel Hill, 4350 Genome Sciences Building, 250 Bell Tower Drive, Chapel Hill, NC 27599-3290, USA.
| | - Neil Spector
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University, C119 LSRC, Research Drive, Durham NC 27701, USA.
| |
Collapse
|
3
|
Nisar N, Mir SA, Kareem O, Pottoo FH. Proteomics approaches in the identification of cancer biomarkers and drug discovery. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
4
|
Rezvani S, Ebadi A, Razzaghi-Asl N. In silico identification of potential Hsp90 inhibitors via ensemble docking, DFT and molecular dynamics simulations. J Biomol Struct Dyn 2022; 40:10665-10676. [PMID: 34286666 DOI: 10.1080/07391102.2021.1947383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) has emerged as one of the most exciting targets for anticancer drug development and Hsp90 inhibitors are potentially useful chemotherapeutic agents in cancer. Within the current study, Hsp90 inhibitors that entered different phases of clinical trials were subjected to Zinc15 structure query to find similar compounds (≥ 78%). Obtained small molecules (1-29) with defined similarity cut-off were docked into ensemble of Hsp90-α NTDs. Docked complexes were ranked on the basis of binding modes and Gibbs free energies as Hsp90 binders (cut-off point; ΔGb ≤ -12 kcal/mol). Top-ranked compounds were subjected to energy decomposition analysis per residue of binding pocket via density functional theory (DFT) calculations in B3LYP level of theory. Subsequent MD simulations of the top-ranked complexes were performed for 100 ns to explore the stable binding modes during a reasonable period in explicit water. Results of molecular docking and intermolecular binding analysis indicated that H-bond, hydrophobic and salt bridge interactions were determinant forces in complex formation. Compounds 19 and 20 were well accommodated in binding pocket of Hsp90 via relatively varied conformations. It was revealed that Asn51 and Phe138 were key residues that interacted stably to 19 and 20. Although primary mechanism of action for proposed molecules are unknown and yet to be explored, results of the present study revealed key structural features for future structure-guided optimization toward potent inhibitors of Hsp90-α NTD. HighlightsHsp90 inhibitors that entered different phases of clinical trials were subjected to Zinc15 based structure query to afford potential enzyme inhibitors 19 and 20.Quantum chemical calculations confirmed docking results and verified pivotal role of a conserved residues (Asn51, Leu103, Phe138 and Tyr139) in making effective hydrogen bonds.MD simulations of top-ranked docked derivatives revealed the achievement of stable binding modes with less conformational variation of 20 than 19 in the active site of Hsp90-α NTD.H-bond, hydrophobic contacts and salt bridge interactions were determinant forces in binding interactions of in silico hits.Resorcinol and isoxazole were important structural motifs of in silico hits in binding to the active site of Hsp90-α NTD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saba Rezvani
- Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ahmad Ebadi
- Department of Medicinal Chemistry, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nima Razzaghi-Asl
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
5
|
Fu X, Liu J, Yan X, DiSanto ME, Zhang X. Heat Shock Protein 70 and 90 Family in Prostate Cancer. Life (Basel) 2022; 12:1489. [PMID: 36294924 PMCID: PMC9605364 DOI: 10.3390/life12101489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer (PCa) is the second most frequent cancer that affects aging men worldwide. However, its exact pathogenesis has not been fully elucidated. The heat shock protein (HSP) family has cell-protective properties that may promote tumor growth and protect cancer cells from death. On a cellular level, HSP molecules have a strong relationship with multiple important biological processes, such as cell differentiation, epithelial-mesenchymal transition (EMT), and fibrosis. Because of the facilitation of HSP family molecules on tumorigenesis, a number of agents and inhibitors are being developed with potent antitumor effects whose target site is the critical structure of HSP molecules. Among all target molecules, HSP70 family and HSP90 are two groups that have been well studied, and therefore, the development of their inhibitors makes great progress. Only a small number of agents, however, have been clinically tested in recruited patients. As a result, more clinical studies are warranted for the establishment of the relationship between the HSP70 family, alongside the HSP90 molecule, and prostate cancer treatment.
Collapse
Affiliation(s)
- Xun Fu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Jiang Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Xin Yan
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Michael E. DiSanto
- Department of Surgery and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08028, USA
| | - Xinhua Zhang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| |
Collapse
|
6
|
Hsp90 in Human Diseases: Molecular Mechanisms to Therapeutic Approaches. Cells 2022; 11:cells11060976. [PMID: 35326427 PMCID: PMC8946885 DOI: 10.3390/cells11060976] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
The maturation of hemeprotein dictates that they incorporate heme and become active, but knowledge of this essential cellular process remains incomplete. Studies on chaperon Hsp90 has revealed that it drives functional heme maturation of inducible nitric oxide synthase (iNOS), soluble guanylate cyclase (sGC) hemoglobin (Hb) and myoglobin (Mb) along with other proteins including GAPDH, while globin heme maturations also need an active sGC. In all these cases, Hsp90 interacts with the heme-free or apo-protein and then drives the heme maturation by an ATP dependent process before dissociating from the heme-replete proteins, suggesting that it is a key player in such heme-insertion processes. As the studies on globin maturation also need an active sGC, it connects the globin maturation to the NO-sGC (Nitric oxide-sGC) signal pathway, thereby constituting a novel NO-sGC-Globin axis. Since many aggressive cancer cells make Hbβ/Mb to survive, the dependence of the globin maturation of cancer cells places the NO-sGC signal pathway in a new light for therapeutic intervention. Given the ATPase function of Hsp90 in heme-maturation of client hemeproteins, Hsp90 inhibitors often cause serious side effects and this can encourage the alternate use of sGC activators/stimulators in combination with specific Hsp90 inhibitors for better therapeutic intervention.
Collapse
|
7
|
Otvagin VF, Kuzmina NS, Kudriashova ES, Nyuchev AV, Gavryushin AE, Fedorov AY. Conjugates of Porphyrinoid-Based Photosensitizers with Cytotoxic Drugs: Current Progress and Future Directions toward Selective Photodynamic Therapy. J Med Chem 2022; 65:1695-1734. [DOI: 10.1021/acs.jmedchem.1c01953] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vasilii F. Otvagin
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Natalia S. Kuzmina
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Ekaterina S. Kudriashova
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Alexander V. Nyuchev
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | | | - Alexey Yu. Fedorov
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
8
|
Goswami R, Russell VS, Tu JJ, Thomas C, Hughes P, Kelly F, Langel SN, Steppe J, Palmer SM, Haystead T, Blasi M, Permar SR. Oral Hsp90 inhibitor SNX-5422 attenuates SARS-CoV-2 replication and dampens inflammation in airway cells. iScience 2021; 24:103412. [PMID: 34786537 PMCID: PMC8579697 DOI: 10.1016/j.isci.2021.103412] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/03/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Currently available SARS-CoV-2 therapeutics are targeted toward moderately to severely ill patients and require intravenous infusions, with limited options for exposed or infected patients with no or mild symptoms. Although vaccines have demonstrated protective efficacy, vaccine hesitancy and logistical distribution challenges will delay their ability to end the pandemic. Hence, there is a need for rapidly translatable, easy-to-administer-therapeutics that can prevent SARS-CoV-2 disease progression, when administered in the early stages of infection. We demonstrate that an orally bioavailable Hsp90 inhibitor, SNX-5422, currently in clinical trials as an anti-cancer therapeutic, inhibits SARS-CoV-2 replication in vitro at a high selectivity index. SNX-5422 treatment of human primary airway epithelial cells dampened expression of inflammatory pathways previously associated with poor SARS-CoV-2 disease outcomes. In addition, SNX-5422 interrupted expression of host factors demonstrated to be crucial for SARS-CoV-2 replication. Development of SNX-5422 as SARS-CoV-2-early-therapy will dampen disease severity, resulting in better clinical outcomes and reduced hospitalizations.
Collapse
Affiliation(s)
- Ria Goswami
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Veronica S. Russell
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Joshua J. Tu
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Charlene Thomas
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York 10065, USA
| | - Philip Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Francine Kelly
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephanie N. Langel
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Justin Steppe
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Scott M. Palmer
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
9
|
Bonanni D, Citarella A, Moi D, Pinzi L, Bergamini E, Rastelli G. Dual Targeting Strategies On Histone Deacetylase 6 (HDAC6) And Heat Shock Protein 90 (Hsp90). Curr Med Chem 2021; 29:1474-1502. [PMID: 34477503 DOI: 10.2174/0929867328666210902145102] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/08/2021] [Accepted: 07/23/2021] [Indexed: 11/22/2022]
Abstract
The design of multi-target drugs acting simultaneously on multiple signaling pathways is a growing field in medicinal chemistry, especially for the treatment of complex diseases such as cancer. Histone deacetylase 6 (HDAC6) is an established anticancer drug target involved in tumor cells transformation. Being an epigenetic enzyme at the interplay of many biological processes, HDAC6 has become an attractive target for polypharmacology studies aimed at improving therapeutic efficacy of anticancer drugs. For example, the molecular chaperone Heat shock protein 90 (Hsp90) is a substrate of HDAC6 deacetylation, and several lines of evidence demonstrate that simultaneous inhibition of HDAC6 and Hsp90 promote synergistic antitumor effects on different cancer cell lines, highlighting the potential benefits of developing a single molecule endowed with multi-target activity. This review will summarize the complex interplay between HDAC6 and Hsp90, providing also useful hints for multi-target drug design and discovery approaches in this field. To this end, crystallographic structures of HDAC6 and Hsp90 complexes will be extensively reviewed in the light of discussing binding pockets features and pharmacophore requirements and providing useful guidelines for the design of dual inhibitors. The few examples of multi-target inhibitors obtained so far, mostly based on chimeric approaches, will be summarized and put into context. Finally, the main features of HDAC6 and Hsp90 inhibitors will be compared, and ligand- and structure-based strategies potentially useful for the development of small molecular weight dual inhibitors will be proposed and discussed.
Collapse
Affiliation(s)
- Davide Bonanni
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| | - Andrea Citarella
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| | - Davide Moi
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| | - Elisa Bergamini
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia Via Campi 183, 41125 Modena, Italy
| |
Collapse
|
10
|
Abstract
In vivo diagnostic imaging of bacterial infections is currently reliant on targeting their metabolic pathways, an ineffective method to identify microbial species with low metabolic activity. Here, we establish HS-198 as a small-molecule fluorescent conjugate that selectively targets the highly conserved bacterial protein HtpG (high-temperature protein G), within Borrelia burgdorferi, the bacterium responsible for Lyme disease. We describe the use of HS-198 to target morphologic forms of B. burgdorferi in both the logarithmic growth phase and the metabolically dormant stationary phase as well as in inactivated spirochetes. Furthermore, in a murine infection model, systemically injected HS-198 identified B. burgdorferi as revealed by imaging in postnecropsy tissue sections. These findings demonstrate how small-molecule probes directed at conserved bacterial protein targets can function to identify the microbe using noninvasive imaging and potentially as scaffolds to deliver antimicrobial agents to the pathogen.
Collapse
|
11
|
Yoshimura C, Nagatoishi S, Kuroda D, Kodama Y, Uno T, Kitade M, Chong-Takata K, Oshiumi H, Muraoka H, Yamashita S, Kawai Y, Ohkubo S, Tsumoto K. Thermodynamic Dissection of Potency and Selectivity of Cytosolic Hsp90 Inhibitors. J Med Chem 2021; 64:2669-2677. [PMID: 33621080 DOI: 10.1021/acs.jmedchem.0c01715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The cytosolic Hsp90-selective inhibitor TAS-116 has an acceptable safety profile and promising antitumor activity in clinical trials. We examined the binding characteristics of TAS-116 and its analogs to determine the impact of the ligand binding mode on selectivity for cytosolic Hsp90. Analyses of the co-crystal structure of Hsp90 and inhibitor TAS-116 suggest that TAS-116 interacts with the ATP-binding pocket, the ATP lid region, and the hydrophobic pocket. A competitive isothermal titration calorimetry analysis confirmed that a small fragment of TAS-116 (THS-510) docks into the lid region and hydrophobic pockets without binding to the ATP-binding pocket. THS-510 exhibited enthalpy-driven binding to Hsp90α and selectively inhibited cytosolic Hsp90 activity. The heat capacity change of THS-510 binding was positive, likely due to the induced conformational rearrangement of Hsp90. Thus, we concluded that interactions with the hydrophobic pocket of Hsp90 determine potency and selectivity of TAS-116 and derivatives for the cytosolic Hsp90 isoform.
Collapse
Affiliation(s)
- Chihoko Yoshimura
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Daisuke Kuroda
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Yasuo Kodama
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Takao Uno
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Makoto Kitade
- Chemical Technology Laboratory, CMC Division, Taiho Pharmaceutical Co. Ltd., Kamikawamachi, Kodama-gun, Saitama 367-0241, Japan
| | - Khoontee Chong-Takata
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Hiromi Oshiumi
- Formulation Research, CMC Division, Taiho Pharmaceutical Co. Ltd., Kawauchi-cho, Tokushima 771-0194, Japan
| | - Hiromi Muraoka
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Satoshi Yamashita
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Yuichi Kawai
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division, Taiho Pharmaceutical Co. Ltd., Tsukuba, Ibaraki 300-2611, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.,Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
12
|
Sanchez J, Carter TR, Cohen MS, Blagg BSJ. Old and New Approaches to Target the Hsp90 Chaperone. Curr Cancer Drug Targets 2020; 20:253-270. [PMID: 31793427 DOI: 10.2174/1568009619666191202101330] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/30/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
The 90-kDa heat shock protein (Hsp90) is a molecular chaperone that ensures cellular proteostasis by maintaining the folding, stabilization, activation, and degradation of over 400 client proteins. Hsp90 is not only critical for routine protein maintenance in healthy cells, but also during states of cellular stress, such as cancer and neurodegenerative diseases. Due to its ability to affect phosphorylation of numerous client proteins, inhibition of Hsp90 has been an attractive anticancer approach since the early 1990's, when researchers identified a druggable target on the amino terminus of Hsp90 for a variety of cancers. Since then, 17 Hsp90 inhibitors that target the chaperone's Nterminal domain, have entered clinical trials. None, however, have been approved thus far by the FDA as a cancer monotherapy. In these trials, a major limitation observed with Hsp90 inhibition at the N-terminal domain was dose-limiting toxicities and relatively poor pharmacokinetic profiles. Despite this, preclinical and clinical research continues to show that Hsp90 inhibitors effectively target cancer cell death and decrease tumor progression supporting the rationale for the development of novel Hsp90 inhibitors. Here, we present an in-depth overview of the Hsp90 inhibitors used in clinical trials. Finally, we present current shifts in the field related to targeting the carboxy-terminal domain of Hsp90 as well as to the development of isoform-selective inhibitors as a means to bypass the pitfalls of current Hsp90 inhibitors and improve clinical trial outcomes.
Collapse
Affiliation(s)
- Jackee Sanchez
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trever R Carter
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Mark S Cohen
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States.,Department of Surgery, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
13
|
Steel TR, Hartinger CG. Metalloproteomics for molecular target identification of protein-binding anticancer metallodrugs. Metallomics 2020; 12:1627-1636. [PMID: 33063808 DOI: 10.1039/d0mt00196a] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Proteomics has played an important role in elucidating the fundamental processes occuring in living cells. Translating these methods to metallodrug research ('metalloproteomics') has provided a means for molecular target identification of metal-based anticancer agents which should signifcantly advance the research field. In combination with biological assays, these techniques have enabled the mechanisms of action of metallodrugs to be linked to their interactions with molecular targets and aid understanding of their biological properties. Such investigations have profoundly increased our knowledge of the complex and dynamic nature of metallodrug-biomolecule interactions and have provided, at least for some compound types, a more detailed picture on their specific protein-binding patterns. This perspective highlights the progression of metallodrug proteomics research for the identification of non-DNA targets from standard analytical techniques to powerful metallodrug pull-down methods.
Collapse
Affiliation(s)
- Tasha R Steel
- School of Chemical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | | |
Collapse
|
14
|
Nazar A, Abbas G, Azam SS. Deciphering the Inhibition Mechanism of under Trial Hsp90 Inhibitors and Their Analogues: A Comparative Molecular Dynamics Simulation. J Chem Inf Model 2020; 60:3812-3830. [PMID: 32659088 DOI: 10.1021/acs.jcim.9b01134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Heat shock protein 90 (Hsp90) performs functions in cellular activities together with other signaling pathways. Hsp90 is evolutionarily conserved and universally articulated as a human cancer-causing agent involved in lung cancer and breast cancer followed by colon and rectum cancers. It has emerged as an effective drug candidate, and inhibition may affect several signaling pathways associated with cancer spread. Therefore, in-silico approaches, molecular docking, molecular dynamics simulation, and binding free energy calculations were applied to create insights into the inhibition mechanism against Hsp90 to identify new cancer therapeutic drugs. Top-docked Hsp90-inhibitor complexes with their analogues were selected as the best complexes based on the GOLD fitness score and orientation. The significant interaction of Hsp90 inhibitors and their analogues were observed to be bound with active site residues as well as residing within the same cavity region. System stability factors RMSD, RMSF, beta-factor, and radius of gyration were analyzed for top-docked complexes and ensure strong binding interaction between inhibitors and the Hsp90 cavity. Cavity bound inhibitors were found to retain consistent hydrogen bonding during the simulation. The radial distribution function (RDF) illustrated that interacting active site residues drive the binding and stability of the inhibitors. Similarly, the axial frequency distribution, which is an indigenously developed analytical tool, produced noteworthy knowledge of the hydrogen-bonding pattern. Results yielded new insights into the design of cancer therapeutic drugs against Hsp90. This finding suggests that under trial Hsp90 inhibitors MPC-3100 could be a potential starting point into the development of potential anticancer agents with the possibility of future directions for the improvement of early existing Hsp90 inhibitors CNF-2024 and SNX-5422 as an anticancer agent.
Collapse
Affiliation(s)
- Asma Nazar
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Ghulam Abbas
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Syed Sikander Azam
- Computational Biology Lab, National Center for Bioinformatics (NCB), Quaid-i-Azam University, Islamabad 45320, Pakistan
| |
Collapse
|
15
|
Babbitt GA, Fokoue EP, Evans JR, Diller KI, Adams LE. DROIDS 3.0-Detecting Genetic and Drug Class Variant Impact on Conserved Protein Binding Dynamics. Biophys J 2019; 118:541-551. [PMID: 31928763 PMCID: PMC7002913 DOI: 10.1016/j.bpj.2019.12.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023] Open
Abstract
The application of statistical methods to comparatively framed questions about the molecular dynamics (MD) of proteins can potentially enable investigations of biomolecular function beyond the current sequence and structural methods in bioinformatics. However, the chaotic behavior in single MD trajectories requires statistical inference that is derived from large ensembles of simulations representing the comparative functional states of a protein under investigation. Meaningful interpretation of such complex forms of big data poses serious challenges to users of MD. Here, we announce Detecting Relative Outlier Impacts from Molecular Dynamic Simulation (DROIDS) 3.0, a method and software package for comparative protein dynamics that includes maxDemon 1.0, a multimethod machine learning application that trains on large ensemble comparisons of concerted protein motions in opposing functional states generated by DROIDS and deploys learned classifications of these states onto newly generated MD simulations. Local canonical correlations in learning patterns generated from independent, yet identically prepared, MD validation runs are used to identify regions of functionally conserved protein dynamics. The subsequent impacts of genetic and/or drug class variants on conserved dynamics can also be analyzed by deploying the classifiers on variant MD simulations and quantifying how often these altered protein systems display opposing functional states. Here, we present several case studies of complex changes in functional protein dynamics caused by temperature, genetic mutation, and binding interactions with nucleic acids and small molecules. We demonstrate that our machine learning algorithm can properly identify regions of functionally conserved dynamics in ubiquitin and TATA-binding protein (TBP). We quantify the impact of genetic variation in TBP and drug class variation targeting the ATP-binding region of Hsp90 on conserved dynamics. We identify regions of conserved dynamics in Hsp90 that connect the ATP binding pocket to other functional regions. We also demonstrate that dynamic impacts of various Hsp90 inhibitors rank accordingly with how closely they mimic natural ATP binding.
Collapse
Affiliation(s)
- Gregory A Babbitt
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, New York.
| | - Ernest P Fokoue
- School of Mathematical Sciences, Rochester Institute of Technology, Rochester, New York
| | - Joshua R Evans
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, New York
| | - Kyle I Diller
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, New York; Golisano College for Computing and Information Science, Rochester, New York
| | - Lily E Adams
- Thomas H. Gosnell School of Life Sciences, Rochester Institute of Technology, Rochester, New York
| |
Collapse
|
16
|
Kadasi S, Yerroju R, Gaddam S, Pullanagiri N, Chary M, Pingili D, Raj S, Raghavendra NM. Discovery of N-pyridoyl-Δ 2 -pyrazolines as Hsp90 inhibitors. Arch Pharm (Weinheim) 2019; 353:e1900192. [PMID: 31808979 DOI: 10.1002/ardp.201900192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/02/2019] [Accepted: 11/17/2019] [Indexed: 01/14/2023]
Abstract
Hsp90, as a key molecular chaperone, plays an important role in modulating the activity of many cell signaling proteins and is an attractive target for anticancer therapeutics. Herein, we report the discovery of N-pyridoyl-Δ2 -pyrazoline analogs as novel Hsp90 inhibitors by integrated approaches of drug design, organic synthesis, cell biology, and qualitative proteomic analysis. Novel chemical compounds were designed and optimized in the adenosine triphosphate-binding site of Hsp90; lead optimized compounds were found to have significant interactions with Asp93 and other amino acids crucial for Hsp90 inhibition. The designed compounds were synthesized by a two-step procedure; different aromatic aldehydes were reacted with various acetophenones to form substituted 1,3-diphenyl-prop-2-enones (Ic-Io), which upon reaction with isonicotinic acid hydrazide in the presence of glacial acetic acid form N-pyridoyl-Δ2 -pyrazoline compounds (PY1-PY13). Compounds PY3, PY2, and PY1 were identified as potential leads amongst the series, with promising anticancer activity against human breast cancer and melanoma cells, and the ability to inhibit Hsp90 similar to radicicol by drug-affinity responsive target stability proteomic analysis in a whole-cell assay.
Collapse
Affiliation(s)
- Sundeep Kadasi
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India.,Department of Chemistry, Osmania University, Hyderabad, India
| | - Ravali Yerroju
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Swetha Gaddam
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Nikhila Pullanagiri
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Meghana Chary
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India
| | - Divya Pingili
- Sri Venkateshwara College of Pharmacy, Osmania University, Hyderabad, India
| | - Shiva Raj
- Department of Chemistry, Osmania University, Hyderabad, India
| | - Nulgumnalli Manjunathaiah Raghavendra
- Department of Pharmaceutical Chemistry, Gokaraju Rangaraju College of Pharmacy, Osmania University, Hyderabad, India.,Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, India
| |
Collapse
|
17
|
Application of immobilized ATP to the study of NLRP inflammasomes. Arch Biochem Biophys 2019; 670:104-115. [PMID: 30641048 DOI: 10.1016/j.abb.2018.12.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/01/2018] [Accepted: 12/17/2018] [Indexed: 01/15/2023]
Abstract
The NLRP proteins are a subfamily of the NOD-like receptor (NLR) innate immune sensors that possess an ATP-binding NACHT domain. As the most well studied member, NLRP3 can initiate the assembly process of a multiprotein complex, termed the inflammasome, upon detection of a wide range of microbial products and endogenous danger signals and results in the activation of pro-caspase-1, a cysteine protease that regulates multiple host defense pathways including cytokine maturation. Dysregulated NLRP3 activation contributes to inflammation and the pathogenesis of several chronic diseases, and the ATP-binding properties of NLRPs are thought to be critical for inflammasome activation. In light of this, we examined the utility of immobilized ATP matrices in the study of NLRP inflammasomes. Using NLRP3 as the prototypical member of the family, P-linked ATP Sepharose was determined to be a highly-effective capture agent. In subsequent examinations, P-linked ATP Sepharose was used as an enrichment tool to enable the effective profiling of NLRP3-biomarker signatures with selected reaction monitoring-mass spectrometry (SRM-MS). Finally, ATP Sepharose was used in combination with a fluorescence-linked enzyme chemoproteomic strategy (FLECS) screen to identify potential competitive inhibitors of NLRP3. The identification of a novel benzo[d]imidazol-2-one inhibitor that specifically targets the ATP-binding and hydrolysis properties of the NLRP3 protein implies that ATP Sepharose and FLECS could be applied other NLRPs as well.
Collapse
|
18
|
Ghosh A, Stuehr DJ. Hsp90 and Its Role in Heme-Maturation of Client Proteins: Implications for Human Diseases. HEAT SHOCK PROTEINS 2019. [DOI: 10.1007/978-3-030-23158-3_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
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: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
20
|
Uno T, Kawai Y, Yamashita S, Oshiumi H, Yoshimura C, Mizutani T, Suzuki T, Chong KT, Shigeno K, Ohkubo M, Kodama Y, Muraoka H, Funabashi K, Takahashi K, Ohkubo S, Kitade M. Discovery of 3-Ethyl-4-(3-isopropyl-4-(4-(1-methyl-1 H-pyrazol-4-yl)-1 H-imidazol-1-yl)-1 H-pyrazolo[3,4- b]pyridin-1-yl)benzamide (TAS-116) as a Potent, Selective, and Orally Available HSP90 Inhibitor. J Med Chem 2018; 62:531-551. [PMID: 30525599 DOI: 10.1021/acs.jmedchem.8b01085] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The molecular chaperone heat shock protein 90 (HSP90) is a promising target for cancer therapy, as it assists in the stabilization of cancer-related proteins, promoting cancer cell growth, and survival. A novel series of HSP90 inhibitors were discovered by structure-activity relationship (SAR)-based optimization of an initial hit compound 11a having a 4-(4-(quinolin-3-yl)-1 H-indol-1-yl)benzamide structure. The pyrazolo[3,4- b]pyridine derivative, 16e (TAS-116), is a selective inhibitor of HSP90α and HSP90β among the HSP90 family proteins and exhibits oral availability in mice. The X-ray cocrystal structure of the 16e analogue 16d demonstrated a unique binding mode at the N-terminal ATP binding site. Oral administration of 16e demonstrated potent antitumor effects in an NCI-H1975 xenograft mouse model without significant body weight loss.
Collapse
Affiliation(s)
- Takao Uno
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Yuichi Kawai
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Satoshi Yamashita
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Hiromi Oshiumi
- Formulation Research, CMC Division , Taiho Pharmaceutical Co. Ltd. , Kawauchi-cho, Tokushima 771-0194 , Japan
| | - Chihoko Yoshimura
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Takashi Mizutani
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Tatsuya Suzuki
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Khoon Tee Chong
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Kazuhiko Shigeno
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Mitsuru Ohkubo
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Yasuo Kodama
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Hiromi Muraoka
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Kaoru Funabashi
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Koichi Takahashi
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Shuichi Ohkubo
- Discovery and Preclinical Research Division , Taiho Pharmaceutical Co. Ltd. , Tsukuba , Ibaraki 300-2611 , Japan
| | - Makoto Kitade
- Chemical Technology Laboratory, CMC Division , Taiho Pharmaceutical Co. Ltd. , Kamikawa-machi, Kodama-gun, Saitama 367-0241 , Japan
| |
Collapse
|
21
|
Neckers L, Blagg B, Haystead T, Trepel JB, Whitesell L, Picard D. Methods to validate Hsp90 inhibitor specificity, to identify off-target effects, and to rethink approaches for further clinical development. Cell Stress Chaperones 2018; 23:467-482. [PMID: 29392504 PMCID: PMC6045531 DOI: 10.1007/s12192-018-0877-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/16/2018] [Accepted: 01/17/2018] [Indexed: 12/12/2022] Open
Abstract
The molecular chaperone Hsp90 is one component of a highly complex and interactive cellular proteostasis network (PN) that participates in protein folding, directs misfolded and damaged proteins for destruction, and participates in regulating cellular transcriptional responses to environmental stress, thus promoting cell and organismal survival. Over the last 20 years, it has become clear that various disease states, including cancer, neurodegeneration, metabolic disorders, and infection by diverse microbes, impact the PN. Among PN components, Hsp90 was among the first to be pharmacologically targeted with small molecules. While the number of Hsp90 inhibitors described in the literature has dramatically increased since the first such small molecule was described in 1994, it has become increasingly apparent that not all of these agents have been sufficiently validated for specificity, mechanism of action, and lack of off-target effects. Given the less than expected activity of Hsp90 inhibitors in cancer-related human clinical trials, a re-evaluation of potentially confounding off-target effects, as well as confidence in target specificity and mechanism of action, is warranted. In this commentary, we provide feasible approaches to achieve these goals and we discuss additional considerations to improve the clinical efficacy of Hsp90 inhibitors in treating cancer and other diseases.
Collapse
Affiliation(s)
- Len Neckers
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, 20892, USA.
| | - Brian Blagg
- Warren Family Research Center for Drug Discovery and Development, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27710, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Luke Whitesell
- Whitehead Institute, Cambridge, MA, 02142, USA
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5G 1M1, Canada
| | - Didier Picard
- Département de Biologie Cellulaire, Université de Genève, 1211, Geneva 4, Switzerland.
| |
Collapse
|
22
|
Abstract
Activation of the heat shock response, and in particular upregulation of stress-inducible Hsp70, herein referred to as Hsp70i, in newly transformed cells, appears to protect against protein damaging stimuli, induction of premature oncogene-induced terminal senescence (OIS), and apoptosis, thereby enabling tumor initiation and progression to an aggressive phenotype. Expressed at very low or undetectable levels in normal tissue, the cytoprotective effects of Hsp70i appear to be mediated through its activity as a molecular chaperone allowing proper folding of mutated proteins, and by blocking cell signaling pathways that regulate OIS and apoptosis. Identification of small-molecule inhibitors selective for Hsp70i could provide new therapeutic tools for cancer treatment. However, identification of selective inhibitors of Hsp70i has proven challenging largely because of the affinity of the protein for ATP. Additionally, its chaperone functions do not lend the protein amenable to traditional enzymatic high-throughput screens. Here, we describe the use of fluorescence-linked enzyme chemoproteomic strategy (FLECS) to identify Hsp70i inhibitors. The FLECS assay is a simple binding assay that enables proteins tagged with fluorophors to be rapidly and quantitative screened against small-molecule libraries. We show several case history examples of the methodology that led to the discovery of the Fatty acid synthase inhibitor, FASNALL, the DAPK3 inhibitor HS38, and HS72, an allosteric inhibitor selective for Hsp70i.
Collapse
|
23
|
Osada T, Kaneko K, Gwin WR, Morse MA, Hobeika A, Pogue BW, Hartman ZC, Hughes PF, Haystead T, Lyerly HK. In Vivo Detection of HSP90 Identifies Breast Cancers with Aggressive Behavior. Clin Cancer Res 2017; 23:7531-7542. [DOI: 10.1158/1078-0432.ccr-17-1453] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/29/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022]
|
24
|
Kulkarni MM, Ratcliff AN, Bhat M, Alwarawrah Y, Hughes P, Arcos J, Loiselle D, Torrelles JB, Funderburg NT, Haystead TA, Kwiek JJ. Cellular fatty acid synthase is required for late stages of HIV-1 replication. Retrovirology 2017; 14:45. [PMID: 28962653 PMCID: PMC5622536 DOI: 10.1186/s12977-017-0368-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 09/14/2017] [Indexed: 11/21/2022] Open
Abstract
Background
Like all viruses, HIV-1 relies on host systems to replicate. The human purinome consists of approximately two thousand proteins that bind and use purines such as ATP, NADH, and NADPH. By virtue of their purine binding pockets, purinome proteins are highly druggable, and many existing drugs target purine-using enzymes. Leveraging a protein affinity media that uses the purine-binding pocket to capture the entire purinome, we sought to define purine-binding proteins regulated by HIV-1 infection. Results Using purinome capture media, we observed that HIV-1 infection increases intracellular levels of fatty acid synthase (FASN), a NADPH-using enzyme critical to the synthesis of de novo fatty acids. siRNA mediated knockdown of FASN reduced HIV-1 particle production by 80%, and treatment of tissue culture cells or primary PBMCs with Fasnall, a newly described selective FASN inhibitor, reduced HIV-1 virion production by 90% (EC50 = 213 nM). Despite the requirement of FASN for nascent virion production, FASN activity was not required for intracellular Gag protein production, indicating that FASN dependent de novo fatty acid biosynthesis contributes to a late step of HIV-1 replication. Conclusions Here we show that HIV-1 replication both increases FASN levels and requires host FASN activity. We also report that Fasnall, a novel FASN inhibitor that demonstrates anti-tumor activity in vivo, is a potent and efficacious antiviral, blocking HIV-1 replication in both tissue culture and primary cell models of HIV-1 replication. In adults, most fatty acids are obtained exogenously from the diet, thus making FASN a plausible candidate for pharmacological intervention. In conclusion, we hypothesize that FASN is a novel host dependency factor and that inhibition of FASN activity has the potential to be exploited as an antiretroviral strategy.
Collapse
Affiliation(s)
- Manjusha M Kulkarni
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Annette N Ratcliff
- Department of Microbiology, Center for Retrovirus Research, The Ohio State University, 476 Biological Sciences Building, 484 W. 12th Avenue, Columbus, OH, 43210, USA.,Promega Corporation, 2800 Woods Hollow Rd, Madison, WI, 53711-5399, USA
| | - Menakshi Bhat
- Department of Microbiology, Center for Retrovirus Research, The Ohio State University, 476 Biological Sciences Building, 484 W. 12th Avenue, Columbus, OH, 43210, USA
| | - Yazan Alwarawrah
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C118 LSRC, Box 3813, Durham, NC, 27710, USA
| | - Philip Hughes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C118 LSRC, Box 3813, Durham, NC, 27710, USA
| | - Jesus Arcos
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - David Loiselle
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C118 LSRC, Box 3813, Durham, NC, 27710, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA.,Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH, USA
| | - Timothy A Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C118 LSRC, Box 3813, Durham, NC, 27710, USA.
| | - Jesse J Kwiek
- Department of Microbiology, Center for Retrovirus Research, The Ohio State University, 476 Biological Sciences Building, 484 W. 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
25
|
Targeting Heat Shock Proteins in Cancer: A Promising Therapeutic Approach. Int J Mol Sci 2017; 18:ijms18091978. [PMID: 28914774 PMCID: PMC5618627 DOI: 10.3390/ijms18091978] [Citation(s) in RCA: 312] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022] Open
Abstract
Heat shock proteins (HSPs) are a large family of chaperones that are involved in protein folding and maturation of a variety of "client" proteins protecting them from degradation, oxidative stress, hypoxia, and thermal stress. Hence, they are significant regulators of cellular proliferation, differentiation and strongly implicated in the molecular orchestration of cancer development and progression as many of their clients are well established oncoproteins in multiple tumor types. Interestingly, tumor cells are more HSP chaperonage-dependent than normal cells for proliferation and survival because the oncoproteins in cancer cells are often misfolded and require augmented chaperonage activity for correction. This led to the development of several inhibitors of HSP90 and other HSPs that have shown promise both preclinically and clinically in the treatment of cancer. In this article, we comprehensively review the roles of some of the important HSPs in cancer, and how targeting them could be efficacious, especially when traditional cancer therapies fail.
Collapse
|
26
|
Jhaveri K, Wang R, Teplinsky E, Chandarlapaty S, Solit D, Cadoo K, Speyer J, D'Andrea G, Adams S, Patil S, Haque S, O'Neill T, Friedman K, Esteva FJ, Hudis C, Modi S. A phase I trial of ganetespib in combination with paclitaxel and trastuzumab in patients with human epidermal growth factor receptor-2 (HER2)-positive metastatic breast cancer. Breast Cancer Res 2017; 19:89. [PMID: 28764748 PMCID: PMC5540198 DOI: 10.1186/s13058-017-0879-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/07/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Targeted therapies in HER2-positive metastatic breast cancer significantly improve outcomes but efficacy is limited by therapeutic resistance. HER2 is an acutely sensitive Heat Shock Protein 90 (HSP90) client and HSP90 inhibition can overcome trastuzumab resistance. Preclinical data suggest that HSP90 inhibition is synergistic with taxanes with the potential for significant clinical activity. We therefore tested ganetespib, a HSP90 inhibitor, in combination with paclitaxel and trastuzumab in patients with trastuzumab-refractory HER2-positive metastatic breast cancer. METHODS In this phase I dose-escalation study, patients with trastuzumab-resistant HER2-positive metastatic breast cancer received weekly trastuzumab (2 mg/kg) and paclitaxel (80 mg/m2) on days 1, 8, 15, and 22 of a 28-day cycle with escalating doses of ganetespib (100 mg/m2, 150 mg/m2, and a third cohort of 125 mg/m2 if needed) on days 1, 8, and 15. Therapy was continued until disease progression or toxicity. The primary objective was to establish the safety and maximum tolerated dose and/or recommended phase II dose (RP2D) of this therapy. The secondary objectives included evaluation of the effects of ganetespib on the pharmacokinetics of paclitaxel, and to make a preliminary assessment of the efficacy of the combination therapy. RESULTS Dose escalation was completed for the two main cohorts without any observed dose-limiting toxicities. Nine patients received treatment. The median prior lines of anti-HER2 therapy numbered three (range 2-4), including prior pertuzumab in 9/9 patients and ado-trastuzumab emtansine (T-DM1) in 8/9 patients. The most common grade 1/2 adverse events (AEs) were diarrhea, fatigue, anemia, and rash. There were no grade 4 AEs related to ganetespib. The overall response rate was 22% (2/9 patients had partial response) and stable disease was seen in 56% (5/9 patients). The clinical benefit rate was 44% (4/9 patients). The median progression-free survival was 20 weeks (range 8-55). CONCLUSION The RP2D of ganetespib is 150 mg/m2 in combination with weekly paclitaxel plus trastuzumab. The combination was safe and well tolerated. Despite prior taxanes, pertuzumab, and T-DM1, clinical activity of this triplet regimen in this heavily pretreated cohort is promising and warrants further study in HER2-positive metastatic breast cancer. TRIAL REGISTRATION ClinicalTrials.gov NCT02060253 . Registered 30 January 2014.
Collapse
Affiliation(s)
- Komal Jhaveri
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Rui Wang
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | | - David Solit
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Karen Cadoo
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - James Speyer
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, NY, USA
| | | | - Sylvia Adams
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, NY, USA
| | - Sujata Patil
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Sofia Haque
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Tara O'Neill
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Kent Friedman
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, NY, USA
| | - Francisco J Esteva
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Medical Center, New York, NY, USA
| | - Clifford Hudis
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Shanu Modi
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
27
|
Gajate P, Martínez-Sáez O, Alonso-Gordoa T, Grande E. Emerging use of everolimus in the treatment of neuroendocrine tumors. Cancer Manag Res 2017; 9:215-224. [PMID: 28684922 PMCID: PMC5484559 DOI: 10.2147/cmar.s113382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neuroendocrine tumors (NETs) consist of a diverse family of malignancies, which are derived from neuroendocrine cells, most commonly originating from the gastroenteropancreatic (GEP) tract or the bronchopulmonary system. In general, NETs are more indolent than epithelial tumors, with median survival rates of longer than 30 months. The upregulation of mTOR pathway has been shown to play a pivotal role in NET pathogenesis. Inhibition of mTOR protein with everolimus represents a progress in the treatment of advanced NETs. Everolimus has shown a significant improvement in progression-free survival (PFS) among patients with pancreatic NETs (pNETs) and nonfunctional GEP and lung NETs in the Phase III RAD001 in Advanced Neuroendocrine Tumors (RADIANT)-3 and RADIANT-4 studies, respectively. In addition, the combination of everolimus with octreotide showed a clinically significant improvement versus octreotide alone in functional NETs in the RADIANT-2 trial. These studies led to the US Food and Drug Administration (FDA) and European Medical Agency (EMA) approval of everolimus. Safety profile of everolimus is generally acceptable. The most common adverse events are stomatitis, diarrhea, rash and fatigue. There is a growing range of novel treatment options in the setting of NETs, but there are no data comparing the activity of different treatment strategies. Thus, treatment decisions are based on different aspects, such as clinical course, patient symptomatology, primary tumor site, tumor functionality, rate of progression and burden of disease. Further research is required to clarify the treatment sequencing to achieve the maximum prolongation in survival and maintenance of quality of life. Future research should concentrate on identification of predictive biomarkers for benefit from different therapies, and studies should also include quality of life as an important measurement in this disease.
Collapse
Affiliation(s)
- Pablo Gajate
- Medical Oncology Department, Ramon y Cajal University Hospital, Madrid, Spain
| | - Olga Martínez-Sáez
- Medical Oncology Department, Ramon y Cajal University Hospital, Madrid, Spain
| | | | - Enrique Grande
- Medical Oncology Department, Ramon y Cajal University Hospital, Madrid, Spain
| |
Collapse
|
28
|
Crowe LB, Hughes PF, Alcorta DA, Osada T, Smith AP, Totzke J, Loiselle DR, Lutz ID, Gargesha M, Roy D, Roques J, Darr D, Lyerly HK, Spector NL, Haystead TA. A Fluorescent Hsp90 Probe Demonstrates the Unique Association between Extracellular Hsp90 and Malignancy in Vivo. ACS Chem Biol 2017; 12:1047-1055. [PMID: 28103010 DOI: 10.1021/acschembio.7b00006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular expression of heat shock protein 90 (eHsp90) by tumor cells is correlated with malignancy. Development of small molecule probes that can detect eHsp90 in vivo may therefore have utility in the early detection of malignancy. We synthesized a cell impermeable far-red fluorophore-tagged Hsp90 inhibitor to target eHsp90 in vivo. High resolution confocal and lattice light sheet microscopy show that probe-bound eHsp90 accumulates in punctate structures on the plasma membrane of breast tumor cells and is actively internalized. The extent of internalization correlates with tumor cell aggressiveness, and this process can be induced in benign cells by overexpressing p110HER2. Whole body cryoslicing, imaging, and histology of flank and spontaneous tumor-bearing mice strongly suggests that eHsp90 expression and internalization is a phenomenon unique to tumor cells in vivo and may provide an "Achilles heel" for the early diagnosis of metastatic disease and targeted drug delivery.
Collapse
Affiliation(s)
- Lauren B. Crowe
- Department of Cell
Biology, Duke University, Durham, North Carolina 27710, United States
| | - Philip F. Hughes
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - David A. Alcorta
- Department of Medicine, Duke University, Durham, North Carolina 27710, United States
| | - Takuya Osada
- Department of Surgery, Duke University, Durham, North Carolina 27710, United States
| | - Aaron P. Smith
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - Juliane Totzke
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - David R. Loiselle
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - Isaac D. Lutz
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | | | - Debasish Roy
- BioInVision, Inc., Mayfield Village, Ohio 44143, United States
| | - Jose Roques
- Lineberger Comprehensive
Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - David Darr
- Lineberger Comprehensive
Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - H. Kim Lyerly
- Department of Surgery, Duke University, Durham, North Carolina 27710, United States
| | - Neil L. Spector
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - Timothy A.J. Haystead
- Department
of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| |
Collapse
|
29
|
Howe MK, Speer BL, Hughes PF, Loiselle DR, Vasudevan S, Haystead TAJ. An inducible heat shock protein 70 small molecule inhibitor demonstrates anti-dengue virus activity, validating Hsp70 as a host antiviral target. Antiviral Res 2016; 130:81-92. [PMID: 27058774 DOI: 10.1016/j.antiviral.2016.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/31/2016] [Accepted: 03/31/2016] [Indexed: 12/27/2022]
Abstract
An estimated three billion people are at risk of Dengue virus (DENV) infection worldwide and there are currently no approved therapeutic interventions for DENV infection. Due to the relatively small size of the DENV genome, DENV is reliant on host factors throughout the viral life cycle. The inducible form of Heat Shock Protein 70 (Hsp70i) has been implicated as a host factor in DENV pathogenesis, however the complete role remains to be elucidated. Here we further illustrate the importance of Hsp70i in dengue virus pathogenesis and describe the antiviral activity of the allosteric small molecule inhibitor that is selective for Hsp70i, called HS-72. In monocytes, Hsp70i is expressed at low levels preceding DENV infection, but Hsp70i expression is induced upon DENV infection. Targeting Hsp70i with HS-72, results in a dose dependent reduction in DENV infected monocytes, while cell viability was maintained. HS-72 works to reduce DENV infection by inhibiting the entry stage of the viral life cycle, through disrupting the association of Hsp70i with the DENV receptor complex. This work highlights Hsp70i as an antiviral target and HS-72 as a potential anti-DENV therapeutic agent.
Collapse
Affiliation(s)
- Matthew K Howe
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Brittany L Speer
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - David R Loiselle
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Subhash Vasudevan
- Emerging Infectious Diseases Program, Duke-NUS Graduate and Medical School, Singapore
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
30
|
Wang M, Shen A, Zhang C, Song Z, Ai J, Liu H, Sun L, Ding J, Geng M, Zhang A. Development of Heat Shock Protein (Hsp90) Inhibitors To Combat Resistance to Tyrosine Kinase Inhibitors through Hsp90-Kinase Interactions. J Med Chem 2016; 59:5563-86. [PMID: 26844689 DOI: 10.1021/acs.jmedchem.5b01106] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heat shock protein 90 (Hsp90) is a ubiquitous chaperone of all of the oncogenic tyrosine kinases. Many Hsp90 inhibitors, alone or in combination, have shown significant antitumor efficacy against the kinase-positive naïve and mutant models. However, clinical trials of these inhibitors are unsuccessful due to insufficient clinical benefits and nonoptimal safety profiles. Recently, much progress has been reported on the Hsp90-cochaperone-client complex, which will undoubtedly assist in the understanding of the interactions between Hsp90 and its clients. Meanwhile, Hsp90 inhibitors have shown promise against patients' resistance caused by early generation tyrosine kinase inhibitors (TKIs), and at least 13 Hsp90 inhibitors are being reevaluated in the clinic. In this regard, the objectives of the current perspective are to summarize the structure and function of the Hsp90-cochaperone-client complex, to analyze the structural and functional insights into the Hsp90-client interactions to address several existing unresolved problems with Hsp90 inhibitors, and to highlight the preclinical and clinical studies of Hsp90 inhibitors as an effective treatment against resistance to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Meining Wang
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| | - Aijun Shen
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Chi Zhang
- Department of Medicinal Chemistry, China Pharmaceutical University , Nanjing 210009, China
| | - Zilan Song
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| | - Jing Ai
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Hongchun Liu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Liping Sun
- Department of Medicinal Chemistry, China Pharmaceutical University , Nanjing 210009, China
| | - Jian Ding
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Meiyu Geng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , Shanghai 201203, China
| | - Ao Zhang
- CAS Key Laboratory of Receptor Research, Synthetic Organic & Medicinal Chemistry Laboratory, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zuchongzhi Lu, Building 3, Room 426, Pudong, Shanghai 201203, China
| |
Collapse
|
31
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. The heat shock protein 90 inhibitor SNX5422 has a synergistic activity with histone deacetylase inhibitors in induction of death of anaplastic thyroid carcinoma cells. Endocrine 2016. [PMID: 26219406 DOI: 10.1007/s12020-015-0706-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The influence of the heat shock protein 90 (hsp90) inhibitor SNX5422 alone or in combination with the histone deacetylase (HDAC) inhibitors PXD101, suberoylanilide hydroxamic acid (SAHA), and trichostatin A (TSA) on survival of anaplastic thyroid carcinoma (ATC) cells was investigated. In 8505C and CAL62 cells, SNX5422 caused cell death with concomitant changes in the expression of hsp90 client proteins. After treatment of both SNX5422 and PXD101, SAHA and TSA, compared with treatment of SNX5422 alone, cell viability was diminished, whereas inhibition rate and cytotoxic activity were enhanced. All of the combination index values were lower than 1.0, suggesting the synergism between SNX5422 and PXD101, SAHA and TSA in induction of cell death. In cells treated with both SNX5422 and PXD101, SAHA and TSA, compared with cells treated with SNX5422 alone, the protein levels of Akt, phospho-4EBP1, phospho-S6 K, and survivin were diminished, while those of γH2AX, acetyl. histone H3, acetyl. histone H4, cleaved PARP, and cleaved caspase-3 were enhanced. In conclusion, these results demonstrate that SNX5422 has a cytotoxic activity in conjunction with alterations in the expression of hsp90 client proteins in ATC cells. Moreover, SNX5422 synergizes with HDAC inhibitors in induction of cytotoxicity accompanied by the suppression of PI3K/Akt/mTOR signaling and survivin, and the overexpression of DNA damage-related proteins in ATC cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jun Goo Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Chul Sik Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Sung-Hee Ihm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Moon Gi Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Hyung Joon Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Seong Jin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon, Republic of Korea.
| |
Collapse
|
32
|
Nanduri P, Hao R, Fitzpatrick T, Yao TP. Chaperone-mediated 26S proteasome remodeling facilitates free K63 ubiquitin chain production and aggresome clearance. J Biol Chem 2015; 290:9455-64. [PMID: 25713068 DOI: 10.1074/jbc.m114.627950] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Indexed: 11/06/2022] Open
Abstract
Efficient elimination of misfolded proteins by the proteasome system is critical for proteostasis. Inadequate proteasome capacity can lead to aberrant aggregation of misfolded proteins and inclusion body formation, a hallmark of neurodegenerative disease. The proteasome system cannot degrade aggregated proteins; however, it stimulates autophagy-dependent aggregate clearance by producing unanchored lysine (K)63-linked ubiquitin chains via the proteasomal deubiquitinating enzyme Poh1. The canonical function of Poh1, which removes ubiquitin chains en bloc from proteasomal substrates prior to their degradation, requires intact 26S proteasomes. Here we present evidence that during aggresome clearance, 20S proteasomes dissociate from protein aggregates, while Poh1 and selective subunits of 19S proteasomes are retained. The dissociation of 20S proteasome components requires the molecular chaperone Hsp90. Hsp90 inhibition suppresses 26S proteasome remodeling, unanchored ubiquitin chain production, and aggresome clearance. Our results suggest that 26S proteasomes undergo active remodeling to generate a Poh1-dependent K63-deubiquitinating enzyme to facilitate protein aggregate clearance.
Collapse
Affiliation(s)
- Priyaanka Nanduri
- From the Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| | - Rui Hao
- From the Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| | - Thomas Fitzpatrick
- From the Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| | - Tso-Pang Yao
- From the Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| |
Collapse
|
33
|
Laukens K, Naulaerts S, Berghe WV. Bioinformatics approaches for the functional interpretation of protein lists: from ontology term enrichment to network analysis. Proteomics 2015; 15:981-96. [PMID: 25430566 DOI: 10.1002/pmic.201400296] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/16/2014] [Accepted: 11/24/2014] [Indexed: 12/24/2022]
Abstract
The main result of a great deal of the published proteomics studies is a list of identified proteins, which then needs to be interpreted in relation to the research question and existing knowledge. In the early days of proteomics this interpretation was only based on expert insights, acquired by digesting a large amount of relevant literature. With the growing size and complexity of the experimental datasets, many computational techniques, databases, and tools have claimed a central role in this task. In this review we discuss commonly and less commonly used methods to functionally interpret experimental proteome lists and compare them with available knowledge. We first address several functional analysis and enrichment techniques based on ontologies and literature. Then we outline how various types of network and pathway information can be used. While the problem of functional interpretation of proteome data is to an extent equivalent to the interpretation of transcriptome or other ''omics'' data, this paper addresses some of the specific challenges and solutions of the proteomics field.
Collapse
Affiliation(s)
- Kris Laukens
- Department of Mathematics and Computer Science, University of Antwerp, Middelheimlaan, Antwerp, Belgium; Biomedical Informatics Research Center Antwerp (biomina), University of Antwerp / Antwerp University Hospital, Antwerp, Belgium
| | | | | |
Collapse
|
34
|
Babak MV, Meier SM, Huber KVM, Reynisson J, Legin AA, Jakupec MA, Roller A, Stukalov A, Gridling M, Bennett KL, Colinge J, Berger W, Dyson PJ, Superti-Furga G, Keppler BK, Hartinger CG. Target profiling of an antimetastatic RAPTA agent by chemical proteomics: relevance to the mode of action. Chem Sci 2015; 6:2449-2456. [PMID: 29308157 PMCID: PMC5647740 DOI: 10.1039/c4sc03905j] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/09/2015] [Indexed: 12/24/2022] Open
Abstract
The RAPTA pharmacophore was linked to beads to identify its biomolecular targets in cancer cells.
The clinical development of anticancer metallodrugs is often hindered by the elusive nature of their molecular targets. To identify the molecular targets of an antimetastatic ruthenium organometallic complex based on 1,3,5-triaza-7-phosphaadamantane (RAPTA), we employed a chemical proteomic approach. The approach combines the design of an affinity probe featuring the pharmacophore with mass-spectrometry-based analysis of interacting proteins found in cancer cell lysates. The comparison of data sets obtained for cell lysates from cancer cells before and after treatment with a competitive binder suggests that RAPTA interacts with a number of cancer-related proteins, which may be responsible for the antiangiogenic and antimetastatic activity of RAPTA complexes. Notably, the proteins identified include the cytokines midkine, pleiotrophin and fibroblast growth factor-binding protein 3. We also detected guanine nucleotide-binding protein-like 3 and FAM32A, which is in line with the hypothesis that the antiproliferative activity of RAPTA compounds is due to induction of a G2/M arrest and histone proteins identified earlier as potential targets.
Collapse
Affiliation(s)
- Maria V Babak
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand . .,Institute of Inorganic Chemistry , University of Vienna , Waehringer Str. 42 , A-1090 Vienna , Austria
| | - Samuel M Meier
- Institute of Analytical Chemistry , University of Vienna , Waehringer Str. 38 , A-1090 Vienna , Austria
| | - Kilian V M Huber
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Jóhannes Reynisson
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand .
| | - Anton A Legin
- Institute of Inorganic Chemistry , University of Vienna , Waehringer Str. 42 , A-1090 Vienna , Austria
| | - Michael A Jakupec
- Institute of Inorganic Chemistry , University of Vienna , Waehringer Str. 42 , A-1090 Vienna , Austria
| | - Alexander Roller
- Institute of Inorganic Chemistry , University of Vienna , Waehringer Str. 42 , A-1090 Vienna , Austria
| | - Alexey Stukalov
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Manuela Gridling
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Jacques Colinge
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Walter Berger
- Department of Medicine I , Institute of Cancer Research , Medical University Vienna , Borschkegasse 8a , A-1090 Vienna , Austria
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine , Lazarettgasse 14, AKH BT 25.3 , A-1090 Vienna , Austria
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry , University of Vienna , Waehringer Str. 42 , A-1090 Vienna , Austria
| | - Christian G Hartinger
- School of Chemical Sciences , University of Auckland , Private Bag 92019 , Auckland 1142 , New Zealand .
| |
Collapse
|
35
|
Vieira JM, Riley PR. Chemical genetics and its potential in cardiac stem cell therapy. Br J Pharmacol 2014; 169:318-27. [PMID: 22385148 DOI: 10.1111/j.1476-5381.2012.01928.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Over the last decade or so, intensive research in cardiac stem cell biology has led to significant discoveries towards a potential therapy for cardiovascular disease; the main cause of morbidity and mortality in humans. The major goal within the field of cardiovascular regenerative medicine is to replace lost or damaged cardiac muscle and coronaries following ischaemic disease. At present, de novo cardiomyocytes can be generated either in vitro, for cell transplantation or disease modelling using directed differentiation of embryonic stem cells or induced pluripotent stem cells, or in vivo via direct reprogramming of resident adult cardiac fibroblast or ectopic stimulation of resident cardiac stem or progenitor cells. A major bottleneck with all of these approaches is the low efficiency of cardiomyocyte differentiation alongside their relative functional immaturity. Chemical genetics, and the application of phenotypic screening with small molecule libraries, represent a means to enhance understanding of the molecular pathways controlling cardiovascular cell differentiation and, moreover, offer the potential for discovery of new drugs to invoke heart repair and regeneration. Here, we review the potential of chemical genetics in cardiac stem cell therapy, highlighting not only the major contributions to the field so far, but also the future challenges.
Collapse
Affiliation(s)
- Joaquim M Vieira
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | |
Collapse
|
36
|
Li M, Tou WI, Zhou H, Li F, Ren H, Chen CYC, Yang B. Developing hypothetical inhibition mechanism of novel urea transporter B inhibitor. Sci Rep 2014; 4:5775. [PMID: 25047372 PMCID: PMC5376056 DOI: 10.1038/srep05775] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/27/2014] [Indexed: 02/03/2023] Open
Abstract
Urea transporter B (UT-B) is a membrane channel protein that specifically transports urea. UT-B null mouse exhibited urea selective urine concentrating ability deficiency, which suggests the potential clinical applications of the UT-B inhibitors as novel diuretics. Primary high-throughput virtual screening (HTVS) of 50000 small-molecular drug-like compounds identified 2319 hit compounds. These 2319 compounds were screened by high-throughput screening using an erythrocyte osmotic lysis assay. Based on the pharmacological data, putative UT-B binding sites were identified by structure-based drug design and validated by ligand-based and QSAR model. Additionally, UT-B structural and functional characteristics under inhibitors treated and untreated conditions were simulated by molecular dynamics (MD). As the result, we identified four classes of compounds with UT-B inhibitory activity and predicted a human UT-B model, based on which computative binding sites were identified and validated. A novel potential mechanism of UT-B inhibitory activity was discovered by comparing UT-B from different species. Results suggest residue PHE198 in rat and mouse UT-B might block the inhibitor migration pathway. Inhibitory mechanisms of UT-B inhibitors and the functions of key residues in UT-B were proposed. The binding site analysis provides a structural basis for lead identification and optimization of UT-B inhibitors.
Collapse
Affiliation(s)
- Min Li
- The State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- These authors contributed equally to this work
| | - Weng Ieong Tou
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
- These authors contributed equally to this work
| | - Hong Zhou
- The State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Fei Li
- The State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- School of Pharmaceutical Sciences, Hubei University of Medicine, Shiyan, 442000, China
| | - Huiwen Ren
- The State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Calvin Yu-Chian Chen
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
- Human Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Department of Biomedical Informatics, Asia University, Taichung, 41354, Taiwan
- Research Center for Chinese Medicine & Acupuncture, China Medical University, Taichung 40402, Taiwan
| | - Baoxue Yang
- The State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
37
|
Huang W, Ye M, Zhang LR, Wu QD, Zhang M, Xu JH, Zheng W. FW-04-806 inhibits proliferation and induces apoptosis in human breast cancer cells by binding to N-terminus of Hsp90 and disrupting Hsp90-Cdc37 complex formation. Mol Cancer 2014; 13:150. [PMID: 24927996 PMCID: PMC4074137 DOI: 10.1186/1476-4598-13-150] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/05/2014] [Indexed: 11/29/2022] Open
Abstract
Background Heat shock protein 90 (Hsp90) is a promising therapeutic target and inhibition of Hsp90 will presumably result in suppression of multiple signaling pathways. FW-04-806, a bis-oxazolyl macrolide compound extracted from China-native Streptomyces FIM-04-806, was reported to be identical in structure to the polyketide Conglobatin. Methods We adopted the methods of chemproteomics, computational docking, immunoprecipitation, siRNA gene knock down, Quantitative Real-time PCR and xenograft models on the research of FW-04-806 antitumor mechanism, through the HER2-overexpressing breast cancer SKBR3 and HER2-underexpressing breast cancer MCF-7 cell line. Results We have verified the direct binding of FW-04-806 to the N-terminal domain of Hsp90 and found that FW-04-806 inhibits Hsp90/cell division cycle protein 37 (Cdc37) chaperone/co-chaperone interactions, but does not affect ATP-binding capability of Hsp90, thereby leading to the degradation of multiple Hsp90 client proteins via the proteasome pathway. In breast cancer cell lines, FW-04-806 inhibits cell proliferation, caused G2/M cell cycle arrest, induced apoptosis, and downregulated Hsp90 client proteins HER2, Akt, Raf-1 and their phosphorylated forms (p-HER2, p-Akt) in a dose and time-dependent manner. Importantly, FW-04-806 displays a better anti-tumor effect in HER2-overexpressed SKBR3 tumor xenograft model than in HER2-underexpressed MCF-7 model. The result is consistent with cell proliferation assay and in vitro apoptosis assay applied for SKBR-3 and MCF-7. Furthermore, FW-04-806 has a favorable toxicity profile. Conclusions As a novel Hsp90 inhibitor, FW-04-806 binds to the N-terminal of Hsp90 and inhibits Hsp90/Cdc37 interaction, resulting in the disassociation of Hsp90/Cdc37/client complexes and the degradation of Hsp90 client proteins. FW-04-806 displays promising antitumor activity against breast cancer cells both in vitro and in vivo, especially for HER2-overexpressed breast cancer cells.
Collapse
Affiliation(s)
| | - Min Ye
- School of Pharmacy, Fujian Medical University, Basic Medicine Building North 205, No,88 Jiaotong Road, Fuzhou, Fujian 350004, China.
| | | | | | | | | | | |
Collapse
|
38
|
Hsp90 inhibitors as new leads to target parasitic diarrheal diseases. Antimicrob Agents Chemother 2014; 58:4138-44. [PMID: 24820073 DOI: 10.1128/aac.02576-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Entamoeba histolytica and Giardia lamblia are anaerobic protozoan parasites that cause amebiasis and giardiasis, two of the most common diarrheal diseases worldwide. Current therapy relies on metronidazole, but resistance has been reported and the drug has significant adverse effects. Therefore, it is critical to search for effective, better-tolerated antiamebic and antigiardial drugs. We synthesized several examples of a recently reported class of Hsp90 inhibitors and evaluated these compounds as potential leads for antiparasitic chemotherapy. Several of these inhibitors showed strong in vitro activity against both E. histolytica and G. lamblia trophozoites. The inhibitors were rescreened to discriminate between amebicidal and giardicidal activity and general cytotoxicity toward a mammalian cell line. No mammalian cytotoxicity was found at >100 μM for 48 h for any of the inhibitors. To understand the mechanism of action, a competitive binding assay was performed using the fluorescent ATP analogue bis-ANS (4,4'-dianilino-1,1'-binaphthyl-5,5'-disulfonic acid dipotassium salt) and recombinant E. histolytica Hsp90 preincubated in both the presence and absence of Hsp90 inhibitors. There was significant reduction in fluorescence compared to the level in the control, suggesting that E. histolytica Hsp90 is a selective target. The in vivo efficacy and safety of one Hsp90 inhibitor in a mouse model of amebic colitis and giardiasis was demonstrated by significant inhibition of parasite growth at a single oral dose of 5 mg/kg of body weight/day for 7 days and 10 mg/kg/day for 3 days. Considering the results for in vitro activity and in vivo efficacy, Hsp90 inhibitors represent a promising therapeutic option for amebiasis and giardiasis.
Collapse
|
39
|
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: 12.6] [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
|
40
|
Buckley DL, Crews CM. Small-molecule control of intracellular protein levels through modulation of the ubiquitin proteasome system. Angew Chem Int Ed Engl 2014; 53:2312-30. [PMID: 24459094 PMCID: PMC4348030 DOI: 10.1002/anie.201307761] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Indexed: 12/25/2022]
Abstract
Traditionally, biological probes and drugs have targeted the activities of proteins (such as enzymes and receptors) that can be readily controlled by small molecules. The remaining majority of the proteome has been deemed "undruggable". By using small-molecule modulators of the ubiquitin proteasome, protein levels, rather than protein activity, can be targeted instead, thus increasing the number of druggable targets. Whereas targeting of the proteasome itself can lead to a global increase in protein levels, the targeting of other components of the UPS (e.g., the E3 ubiquitin ligases) can lead to an increase in protein levels in a more targeted fashion. Alternatively, multiple strategies for inducing protein degradation with small-molecule probes are emerging. With the ability to induce and inhibit the degradation of targeted proteins, small-molecule modulators of the UPS have the potential to significantly expand the druggable portion of the proteome beyond traditional targets, such as enzymes and receptors.
Collapse
Affiliation(s)
- Dennis L. Buckley
- Departments of Chemistry; Molecular, Cellular & Developmental, Biology; Pharmacology, Yale University, New Haven, Connecticut 06511, United States
| | - Craig M. Crews
- Departments of Chemistry; Molecular, Cellular & Developmental, Biology; Pharmacology, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
41
|
Buckley DL, Crews CM. Steuerung der intrazellulären Proteinmenge durch niedermolekulare Modulatoren des Ubiquitin-Proteasom-Systems. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201307761] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Rathore APS, Haystead T, Das PK, Merits A, Ng ML, Vasudevan SG. Chikungunya virus nsP3 & nsP4 interacts with HSP-90 to promote virus replication: HSP-90 inhibitors reduce CHIKV infection and inflammation in vivo. Antiviral Res 2013; 103:7-16. [PMID: 24388965 DOI: 10.1016/j.antiviral.2013.12.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
The global emergence of Chikungunya virus (CHIKV) infection is alarming and currently there is no licensed vaccine or antiviral treatment available to mitigate this disease. CHIKV infection typically results in high viral load with an outcome of high fever, skin rashes, muscle pain, and sequelae of prolonged arthritis, which occurs in >90% of the infected cases. In this study, using biochemical pull-downs, mass-spectrometry, and microscopic imaging techniques, we have identified novel interactions between CHIKV nsP3 or nsP4 proteins with the host stress-pathway chaperone HSP-90 protein. Indeed, silencing of HSP-90 transcripts using siRNA disrupts CHIKV replication in cultured cells. Furthermore, drugs targeting HSP-90, such as commercially available geldanamycin, as well as other specific HSP-90 inhibitor drugs that had been obtained from a purinome mining approach (HS-10 and SNX-2112) showed dramatic reduction in viral titers and reduced inflammation in a CHIKV mouse model of severe infection and musculopathy. The detailed study of the underlying molecular mechanism of these viral and host protein interactions may provide a platform to develop novel therapeutics against CHIKV infection.
Collapse
Affiliation(s)
- Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore; Department of Microbiology, National University of Singapore, Singapore 117597, Singapore
| | - Timothy Haystead
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, United States; Department of Pathology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Pratyush K Das
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Andres Merits
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Mah-Lee Ng
- Department of Microbiology, National University of Singapore, Singapore 117597, Singapore
| | - Subhash G Vasudevan
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore; Department of Microbiology, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
43
|
Carlson DA, Franke AS, Weitzel DH, Speer BL, Hughes PF, Hagerty L, Fortner CN, Veal JM, Barta TE, Zieba BJ, Somlyo AV, Sutherland C, Deng JT, Walsh MP, MacDonald JA, Haystead TAJ. Fluorescence linked enzyme chemoproteomic strategy for discovery of a potent and selective DAPK1 and ZIPK inhibitor. ACS Chem Biol 2013; 8:2715-23. [PMID: 24070067 DOI: 10.1021/cb400407c] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
DAPK1 and ZIPK (also called DAPK3) are closely related serine/threonine protein kinases that regulate programmed cell death and phosphorylation of non-muscle and smooth muscle myosin. We have developed a fluorescence linked enzyme chemoproteomic strategy (FLECS) for the rapid identification of inhibitors for any element of the purinome and identified a selective pyrazolo[3,4-d]pyrimidinone (HS38) that inhibits DAPK1 and ZIPK in an ATP-competitive manner at nanomolar concentrations. In cellular studies, HS38 decreased RLC20 phosphorylation. In ex vivo studies, HS38 decreased contractile force generated in mouse aorta, rabbit ileum, and calyculin A stimulated arterial muscle by decreasing RLC20 and MYPT1 phosphorylation. The inhibitor also promoted relaxation in Ca(2+)-sensitized vessels. A close structural analogue (HS43) with 5-fold lower affinity for ZIPK produced no effect on cells or tissues. These findings are consistent with a mechanism of action wherein HS38 specifically targets ZIPK in smooth muscle. The discovery of HS38 provides a lead scaffold for the development of therapeutic agents for smooth muscle related disorders and a chemical means to probe the function of DAPK1 and ZIPK across species.
Collapse
Affiliation(s)
- David A. Carlson
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Aaron S. Franke
- Department
of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Douglas H. Weitzel
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Brittany L. Speer
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Philip F. Hughes
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Laura Hagerty
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Christopher N. Fortner
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - James M. Veal
- Quanticel
Pharmaceuticals, San Francisco, California 94158, United States
| | - Thomas E. Barta
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Bartosz J. Zieba
- Department
of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Avril V. Somlyo
- Department
of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Cindy Sutherland
- Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta. Department of Biochemistry & Molecular Biology, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Jing Ti Deng
- Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta. Department of Biochemistry & Molecular Biology, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Michael P. Walsh
- Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta. Department of Biochemistry & Molecular Biology, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Justin A. MacDonald
- Smooth Muscle Research Group at the Libin Cardiovascular Institute of Alberta. Department of Biochemistry & Molecular Biology, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N 4Z6, Canada
| | - Timothy A. J. Haystead
- Department
of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| |
Collapse
|
44
|
Mayer C, Janin YL. Non-quinolone inhibitors of bacterial type IIA topoisomerases: a feat of bioisosterism. Chem Rev 2013; 114:2313-42. [PMID: 24313284 DOI: 10.1021/cr4003984] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Claudine Mayer
- Unité de Microbiologie Structurale, Département de Biologie Structurale et Chimie, Institut Pasteur , 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | | |
Collapse
|
45
|
Pizarro JC, Hills T, Senisterra G, Wernimont AK, Mackenzie C, Norcross NR, Ferguson MAJ, Wyatt PG, Gilbert IH, Hui R. Exploring the Trypanosoma brucei Hsp83 potential as a target for structure guided drug design. PLoS Negl Trop Dis 2013; 7:e2492. [PMID: 24147171 PMCID: PMC3798429 DOI: 10.1371/journal.pntd.0002492] [Citation(s) in RCA: 31] [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: 01/30/2013] [Accepted: 09/09/2013] [Indexed: 01/08/2023] Open
Abstract
Human African trypanosomiasis is a neglected parasitic disease that is fatal if untreated. The current drugs available to eliminate the causative agent Trypanosoma brucei have multiple liabilities, including toxicity, increasing problems due to treatment failure and limited efficacy. There are two approaches to discover novel antimicrobial drugs - whole-cell screening and target-based discovery. In the latter case, there is a need to identify and validate novel drug targets in Trypanosoma parasites. The heat shock proteins (Hsp), while best known as cancer targets with a number of drug candidates in clinical development, are a family of emerging targets for infectious diseases. In this paper, we report the exploration of T. brucei Hsp83 – a homolog of human Hsp90 – as a drug target using multiple biophysical and biochemical techniques. Our approach included the characterization of the chemical sensitivity of the parasitic chaperone against a library of known Hsp90 inhibitors by means of differential scanning fluorimetry (DSF). Several compounds identified by this screening procedure were further studied using isothermal titration calorimetry (ITC) and X-ray crystallography, as well as tested in parasite growth inhibitions assays. These experiments led us to the identification of a benzamide derivative compound capable of interacting with TbHsp83 more strongly than with its human homologs and structural rationalization of this selectivity. The results highlight the opportunities created by subtle structural differences to develop new series of compounds to selectively target the Trypanosoma brucei chaperone and effectively kill the sleeping sickness parasite. Sleeping sickness, or human African trypanosomiasis (HAT), is a deadly neglected disease for which new therapeutic options are badly needed. Current drugs have several liabilities including toxicity and route of administration limiting their efficacy to combat the disease. Our study aimed at validating a potential new drug target against Trypanosoma brucei, its heat shock protein 83 (Hsp83). The chaperone was screened against a repurposed library composed of inhibitors against the human Hsp90. The compounds were assayed in their ability to bind the T. brucei protein and to kill the parasite. Our work has identified selective and high-affinity chemical compounds targeting the parasitic Hsp83. Additionally, structural studies were conducted to explore the observed selectivity of selected inhibitors. Our work has validated T. brucei Hsp83 as a potential target for future drug discovery campaigns. It has also shown the strength of repurposing chemical libraries developed against human proteins, emphasizing the possibility to piggyback current and past drug discovery efforts for other diseases in the search for new drugs against neglected tropical diseases.
Collapse
Affiliation(s)
- Juan Carlos Pizarro
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| | - Tanya Hills
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Guillermo Senisterra
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Amy K. Wernimont
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| | - Claire Mackenzie
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Neil R. Norcross
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Michael A. J. Ferguson
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Paul G. Wyatt
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Ian H. Gilbert
- Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom
| | - Raymond Hui
- The Structural Genomics Consortium (SGC), University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Barrott JJ, Hughes PF, Osada T, Yang XY, Hartman ZC, Loiselle DR, Spector NL, Neckers L, Rajaram N, Hu F, Ramanujam N, Vaidyanathan G, Zalutsky MR, Lyerly HK, Haystead TA. Optical and radioiodinated tethered Hsp90 inhibitors reveal selective internalization of ectopic Hsp90 in malignant breast tumor cells. ACTA ACUST UNITED AC 2013; 20:1187-97. [PMID: 24035283 DOI: 10.1016/j.chembiol.2013.08.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 07/18/2013] [Accepted: 08/10/2013] [Indexed: 01/02/2023]
Abstract
Inhibitors of heat-shock protein 90 (Hsp90) have demonstrated an unusual selectivity for tumor cells despite its ubiquitous expression. This phenomenon has remained unexplained, but could be influenced by ectopically expressed Hsp90 in tumors. In this work, we synthesized Hsp90 inhibitors that can carry optical or radioiodinated probes via a polyethyleneglycol tether. We show that these tethered inhibitors selectively recognize cells expressing ectopic Hsp90 and become internalized. The internalization process is blocked by Hsp90 antibodies, suggesting that active cycling of the protein occurs at the plasma membrane. In mice, we observed exquisite accumulation of the fluor-tethered versions within breast tumors at very sensitive levels. Cell-based assays with the radiolabeled version showed picomolar detection in cells that express ectopic Hsp90. Our findings show that fluor-tethered or radiolabeled inhibitors that target ectopic Hsp90 can be used to detect breast cancer malignancies through noninvasive imaging.
Collapse
Affiliation(s)
- Jared J Barrott
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Friedman JA, Wise SC, Hu M, Gouveia C, Vander Broek R, Freudlsperger C, Kannabiran VR, Arun P, Mitchell JB, Chen Z, Van Waes C. HSP90 Inhibitor SNX5422/2112 Targets the Dysregulated Signal and Transcription Factor Network and Malignant Phenotype of Head and Neck Squamous Cell Carcinoma. Transl Oncol 2013; 6:429-41. [PMID: 23908686 PMCID: PMC3730018 DOI: 10.1593/tlo.13292] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/16/2013] [Accepted: 06/05/2013] [Indexed: 01/09/2023] Open
Abstract
Heat shock protein 90 (HSP90) is a chaperone protein that stabilizes proteins involved in oncogenic and therapeutic resistance pathways of epithelial cancers, including head and neck squamous cell carcinomas (HNSCCs). Here, we characterized the molecular, cellular, and preclinical activity of HSP90 inhibitor SNX5422/2112 in HNSCC overexpressing HSP90. SNX2112 inhibited proliferation, induced G2/M block, and enhanced cytotoxicity, chemosensitivity, and radiosensitivity between 25 and 250 nM in vitro. SNX2112 showed combinatorial activity with paclitaxel in wild-type (wt) TP53-deficient and cisplatin in mutant (mt) TP53 HNSCC lines. SNX2112 decreased expression or phosphorylation of epidermal growth factor receptor (EGFR), c-MET, v-akt murine thymoma viral oncogene homolog 1 (AKT), extracellular signal-regulated kinases (ERK) 1 and 2, inhibitor κB kinase, and signal transducer and transcription factor 3 (STAT3), corresponding downstream nuclear factor κB, activator protein-1, and STAT3 reporter genes, and target oncogenes and angiogenic cytokines. Furthermore, SNX2112 enhanced re-expression of TP53 and targets p21WAF1 and PUMA, while TP53 inhibitor Pifithrin or siRNA attenuated the antiproliferative activity of SNX2112 in wtTP53 HNSCC in vitro. Prodrug SNX5422 similarly down-modulated key signal targets, enhanced TP53 expression and apoptosis, and inhibited proliferation, angiogenesis, and tumorigenesis in a wtTP53-deficient HNSCC xenograft model. Thus, HSP90 inhibitor SNX5422/2112 broadly modulates multiple key nodes within the dysregulated signaling network, with corresponding effects upon the malignant phenotype. Our data support investigation of SNX5422/2112 in combination with paclitaxel, cisplatin, and radiotherapy in HNSCC with different TP53 status.
Collapse
Affiliation(s)
- Jay A Friedman
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Stephanie C Wise
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Michael Hu
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Chris Gouveia
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Robert Vander Broek
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Christian Freudlsperger
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
- Department of Oral and Maxillofacial Surgery, University Hospital, Heidelberg, Germany
| | - Vishnu R Kannabiran
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Pattatheyil Arun
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - James B Mitchell
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Zhong Chen
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Carter Van Waes
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Identification of an aminothiazole with antifungal activity against intracellular Histoplasma capsulatum. Antimicrob Agents Chemother 2013; 57:4349-59. [PMID: 23817367 DOI: 10.1128/aac.00459-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
As eukaryotes, fungi possess relatively few molecules sufficiently unique from mammalian cell components to be used as drug targets. Consequently, most current antifungals have significant host cell toxicity. Primary fungal pathogens (e.g., Histoplasma) are of particular concern, as few antifungals are effective in treating them. To identify additional antifungal candidates for the treatment of histoplasmosis, we developed a high-throughput platform for monitoring Histoplasma growth and employed it in a phenotypic screen of 3,600 commercially available compounds. Seven hit compounds that inhibited Histoplasma yeast growth were identified. Compound 41F5 has fungistatic activity against Histoplasma yeast at micromolar concentrations, with a 50% inhibitory concentration (IC50) of 0.87 μM, and has the greatest selectivity for yeast (at least 62-fold) relative to host cells. Structurally, 41F5 consists of an aminothiazole core with an alicyclic substituent at the 2-position and an aromatic substituent at the 5-position. 41F5 inhibits Histoplasma growth in liquid culture and similarly inhibits yeast cells within macrophages, the actual host environment of this fungal pathogen during infection. Importantly, 41F5 protects infected host cells from Histoplasma-induced macrophage death, making this aminothiazole hit compound an excellent candidate for development as an antifungal for Histoplasma infections.
Collapse
|
49
|
Synthesis of 19-substituted geldanamycins with altered conformations and their binding to heat shock protein Hsp90. Nat Chem 2013; 5:307-14. [PMID: 23511419 DOI: 10.1038/nchem.1596] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 02/04/2013] [Indexed: 12/19/2022]
Abstract
The benzoquinone ansamycin geldanamycin and its derivatives are inhibitors of heat shock protein Hsp90, an emerging target for novel therapeutic agents both in cancer and in neurodegeneration. However, the toxicity of these compounds to normal cells has been ascribed to reaction with thiol nucleophiles at the quinone 19-position. We reasoned that blocking this position would ameliorate toxicity, and that it might also enforce a favourable conformational switch of the trans-amide group into the cis-form required for protein binding. Here, we report an efficient synthesis of such 19-substituted compounds and realization of our hypotheses. Protein crystallography established that the new compounds bind to Hsp90 with, as expected, a cis-amide conformation. Studies on Hsp90 inhibition in cells demonstrated the molecular signature of Hsp90 inhibitors: decreases in client proteins with compensatory increases in other heat shock proteins in both human breast cancer and dopaminergic neural cells, demonstrating their potential for use in the therapy of cancer or neurodegenerative diseases.
Collapse
|
50
|
Barrott JJ, Haystead TAJ. Hsp90, an unlikely ally in the war on cancer. FEBS J 2013; 280:1381-96. [PMID: 23356585 PMCID: PMC3815692 DOI: 10.1111/febs.12147] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/10/2013] [Accepted: 01/22/2013] [Indexed: 12/25/2022]
Abstract
On the surface heat shock protein 90 (Hsp90) is an unlikely drug target for the treatment of any disease, let alone cancer. Hsp90 is highly conserved and ubiquitously expressed in all cells. There are two major isoforms α and β encoded by distinct genes and together they may constitute 1%-3% of the cellular protein. Deletion of the protein is embryonic lethal and there are no recognized polymorphisms suggesting an association or causal relationship with any human disease. With respect to cancer, the proteins absence from two recent high profile articles, 'Hallmarks of cancer: the next generation' [Hanahan & Weinberg (2011) Cell 144, 646-674] and 'Comprehensive molecular portraits of human breast tumours' [Koboldt et al. (2012) Nature] underlines the perception that it is an unlikely bona fide target to treat this disease. Yet, to date, there are 17 distinct Hsp90 inhibitors in clinical trials for multiple indications in cancer. The protein has been championed for over 20 years by the National Cancer Institute (Bethesda, MD, USA) as a cancer target since the discovery of the antitumor activity of the natural product geldanamycin. This review aims to look at the conundrum of why Hsp90 can even be considered a druggable target for the treatment of cancer. We propose that in contrast to the majority of chemotherapeutics our growing armamentarium of investigational Hsp90 drugs represents an elegant choice that offers real hope in the long-term treatment of certain cancers.
Collapse
Affiliation(s)
- Jared J Barrott
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | |
Collapse
|