1
|
Chang C, Tang X, Woodley DT, Chen M, Li W. Previously unrecognized and potentially consequential challenges facing Hsp90 inhibitors in cancer clinical trials. Cell Stress Chaperones 2024; 29:642-653. [PMID: 39181529 PMCID: PMC11402043 DOI: 10.1016/j.cstres.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024] Open
Abstract
Targeting the heat shock protein-90 (Hsp90) chaperone machinery in various cancers with 200 monotherapy or combined-therapy clinical trials since 1999 has not yielded any success of food and drug administration approval. Blames for the failures were unanimously directed at the Hsp90 inhibitors or tumors or both. However, analyses of recent cellular and genetic studies together with the Hsp90 data from the Human Protein Atlas database suggest that the vast variations in Hsp90 expression among different organs in patients might have been the actual cause. It is evident now that Hsp90β is the root of dose-limiting toxicity (DLT), whereas Hsp90α is a buffer of penetrated Hsp90 inhibitors. The more Hsp90α, the safer Hsp90β, and the lower DLT are for the host. Unfortunately, the dramatic variations of Hsp90, from total absence in the eye, muscle, pancreas, and heart to abundance in reproduction organs, lung, liver, and gastrointestinal track, would cause the selection of any fair toxicity biomarker and an effective maximum tolerable dose (MTD) of Hsp90 inhibitor extremely challenging. In theory, a safe MTD for the organs with high Hsp90 could harm the organs with low Hsp90. In reverse, a safe MTD for organs with low or undetectable Hsp90 would have little impact on the tumors, whose cells exhibit average 3-7% Hsp90 over the average 2-3% Hsp90 in normal cells. Moreover, not all tumor cell lines tested follow the "inhibitor binding-client protein degradation" paradigm. It is likely why the oral Hsp90 inhibitor TAS-116 (Pimitespib), which bypasses blood circulation and other organs, showed some beneficiary efficacy by conveniently hitting tumors along the gastrointestinal track. The critical question is what the next step will be for the Hsp90 chaperone as a cancer therapeutic target.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Xin Tang
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - David T Woodley
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Mei Chen
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA
| | - Wei Li
- Department of Dermatology and USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, CA 90033, USA.
| |
Collapse
|
2
|
Sharma S, Wang SA, Yang WB, Lin HY, Lai MJ, Chen HC, Kao TY, Hsu FL, Nepali K, Hsu TI, Liou JP. First-in-Class Dual EZH2-HSP90 Inhibitor Eliciting Striking Antiglioblastoma Activity In Vitro and In Vivo. J Med Chem 2024; 67:2963-2985. [PMID: 38285511 PMCID: PMC10895674 DOI: 10.1021/acs.jmedchem.3c02053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Structural analysis of tazemetostat, an FDA-approved EZH2 inhibitor, led us to pinpoint a suitable site for appendage with a pharmacophoric fragment of second-generation HSP90 inhibitors. Resultantly, a magnificent dual EZH2/HSP90 inhibitor was pinpointed that exerted striking cell growth inhibitory efficacy against TMZ-resistant Glioblastoma (GBM) cell lines. Exhaustive explorations of chemical probe 7 led to several revelations such as (i) compound 7 increased apoptosis/necrosis-related gene expression, whereas decreased M phase/kinetochore/spindle-related gene expression as well as CENPs protein expression in Pt3R cells; (ii) dual inhibitor 7 induced cell cycle arrest at the M phase; (iii) compound 7 suppressed reactive oxygen species (ROS) catabolism pathway, causing the death of TMZ-resistant GBM cells; and (iv) compound 7 elicited substantial in vivo anti-GBM efficacy in experimental mice xenografted with TMZ-resistant Pt3R cells. Collectively, the study results confirm the potential of dual EZH2-HSP90 inhibitor 7 as a tractable anti-GBM agent.
Collapse
Affiliation(s)
- Sachin Sharma
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
| | - Shao-An Wang
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Bin Yang
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
| | - Hong-Yi Lin
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mei-Jung Lai
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
| | - Hsien-Chung Chen
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- Department
of Neurosurgery, Shuang Ho Hospital, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Tzu-Yuan Kao
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Feng-Lin Hsu
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kunal Nepali
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-I Hsu
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
- International
Master Program in Medical Neuroscience, College of Medical Science
and Technology, Taipei Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
| | - Jing-Ping Liou
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
3
|
Varlı M, Ngo MT, Kim SM, Taş İ, Zhou R, Gamage CD, Pulat S, Park SY, Sesal NC, Hur JS, Kang KB, Kim H. A fatty acid-rich fraction of an endolichenic fungus Phoma sp. suppresses immune checkpoint markers via AhR/ARNT and ESR1. Heliyon 2023; 9:e19185. [PMID: 37662726 PMCID: PMC10474435 DOI: 10.1016/j.heliyon.2023.e19185] [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] [Received: 03/27/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023] Open
Abstract
Lung cancer has the highest mortality rates worldwide. The disease is caused by environmental pollutants, smoking, and many other factors. Recent treatments include immunotherapeutics, which have shown some success; however, the search for new therapeutics is ongoing. Endolichenic fungi produce a whale of a lot of secondary metabolites, the therapeutic effects of which are being evaluated. Here, we used a crude extract and subfractions of the endolichenic fungus, Phoma sp. (EL006848), isolated from the Pseudevernia furfuracea. It was identified the fatty acid components, palmitic acid, stearic acid, and oleic acid, exist in subfractions E1 and E2. In addition, EL006848 and its fatty acids fractions suppressed benzo[a]pyrene (an AhR ligand)- induced expression of PD-L1 to inhibit the activity of multiple immune checkpoints. E2 subfraction, which had a higher fatty acid content than E1, downregulated expression of AhR/ARNT and several human transcription factors related to ESR1. Moreover, E2 showed a strong inhibitory effect on STAT3 expression and mild effect on NF-kB activity. These results suggest that fatty acids extracted from an endolichenic fungus can exert strong immunotherapeutic effects.
Collapse
Affiliation(s)
- Mücahit Varlı
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Men Thi Ngo
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Seong-Min Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D.B. Gamage
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Nüzhet Cenk Sesal
- Faculty of Arts and Sciences, Department of Biology, Marmara University, Istanbul, Turkey
| | - Jae-Seoun Hur
- Korean Lichen Research Institute, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Kyo Bin Kang
- College of Pharmacy, Sookmyung Women's University, 100 Cheongpa-ro 47 gil, Seoul 04310, Republic of Korea
| | - Hangun Kim
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
4
|
Manunu B, Serafin AM, Akudugu JM. BAG1, MGMT, FOXO1, and DNAJA1 as potential drug targets for radiosensitizing cancer cell lines. Int J Radiat Biol 2023; 99:292-307. [PMID: 35511481 DOI: 10.1080/09553002.2022.2074164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE Activation of some signaling pathways can promote cell survival and have a negative impact on tumor response to radiotherapy. Here, the role of differences in expression levels of genes related to the poly(ADP-ribose) polymerase-1 (PARP-1), heat shock protein 90 (Hsp90), B-cell lymphoma 2 (Bcl-2), and phosphoinositide 3-kinase (PI3K) pathways in the survival or death of cells following X-ray exposure was investigated. METHODS Eight human cell cultures (MCF-7 and MDA-MB-231: breast cancers; MCF-12A: apparently normal breast; A549: lung cancer; L132: normal lung; G28, G44 and G112: glial cancers) were irradiated with X-rays. The colony-forming and real-time PCR based on a custom human pathway RT2 Profiler PCR Array assays were used to evaluate cell survival and gene expression, respectively. RESULTS The surviving fractions at 2 Gy for the cell lines, in order of increasing radioresistance, were found to be as follows: MCF-7 (0.200 ± 0.011), G44 (0.277 ± 0.065), L132 (0.367 ± 0.023), MDA-MB-231 (0.391 ± 0.057), G112 (0.397 ± 0.113), A549 (0.490 ± 0.048), MCF-12A (0.526 ± 0.004), and G28 (0.633 ± 0.094). The rank order of radioresistance at 6 Gy was: MCF-7 < L132 < G44 < MDA-MB-231 < A549 < G28 < G112 < MCF-12A. PCR array data analysis revealed that several genes were differentially expressed between irradiated and unirradiated cell cultures. The following genes, with fold changes: BCL2A1 (21.91), TP53 (8743.75), RAD51 (11.66), FOX1 (65.86), TCP1 (141.32), DNAJB1 (3283.64), RAD51 (51.52), and HSPE1 (12887.29) were highly overexpressed, and BAX (-127.21), FOX1 (-81.79), PDPK1 (-1241.78), BRCA1 (-8.70), MLH1 (-12143.95), BCL2 (-18.69), CCND1 (-46475.98), and GJA1 (-2832.70) were highly underexpressed in the MDA-MB-231, MCF-7, MCF-12A, A549, L132, G28, G44, and G112 cell lines, respectively. The radioresistance in the malignant A549 and G28 cells was linked to upregulation in the apoptotic, DNA repair, PI3K, and Hsp90 pathway genes BAG1, MGMT, FOXO1, and DNAJA1, respectively, and inhibition of these genes resulted in significant radiosensitization. CONCLUSIONS Targeting BAG1, MGMT, FOXO1, and DNAJA1 with specific inhibitors might effectively sensitize radioresistant tumors to radiotherapy.
Collapse
Affiliation(s)
- Bayanika Manunu
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Antonio M Serafin
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - John M Akudugu
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
5
|
Yingsunthonwattana W, Junprung W, Supungul P, Tassanakajon A. Heat shock protein 90 of Pacific white shrimp (Litopenaeus vannamei) is possibly involved in promoting white spot syndrome virus infection. FISH & SHELLFISH IMMUNOLOGY 2022; 128:405-418. [PMID: 35964878 DOI: 10.1016/j.fsi.2022.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Viruses cause up to 60% of disease-associated losses in shrimp aquaculture, and the white spot syndrome virus (WSSV) is a major viral pathogen in shrimp. Heat shock proteins (HSPs) are host chaperones that help promote many viral infections. We investigated the involvement of Litopenaeus vannamei (Lv) HSP90 in WSSV infections. Expression of LvHSP90 at the transcript and protein levels were upregulated after WSSV infection. Silencing LvHSP90 resulted in the increased cumulative mortality rate and the reduction of circulating hemocytes. The inhibition of LvHSP90 also induced the expression of apoptosis-related genes which indicated the induction of apoptotic pathway and might lead to shrimp death. However, lower the number of WSSV-infected cells and viral copy numbers were detected in the LvHSP90-silenced shrimp compared with those of the controls, corresponding with significantly decreased expressions of viral genes, including the immediate-early genes WSV083 and WSV249 and viral DNA polymerase. Conversely, injecting shrimp with WSSV that had been co-incubated with a recombinant LvHSP90 (rLvHSP90) promoted WSSV infection as evidenced by an increased cumulative mortality rate and viral copy numbers at 40-48 h post infection (hpi). Subcellular localization of LvHSP90 in WSSV-infected hemocytes at 3, 6 and 12 hpi demonstrated increased expression and translocation of LvHSP90 into the nucleus where WSSV DNA can replicate. Thus, LvHSP90 might be involved in the WSSV pathogenesis by promoting WSSV replication.
Collapse
Affiliation(s)
- Warumporn Yingsunthonwattana
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Wisarut Junprung
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Premruethai Supungul
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, 12120, Thailand
| | - Anchalee Tassanakajon
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
6
|
Maleki MP, Soltanzade H, Tanomand A, Shahsavari G. The layered double hydroxide (LDH) nanosheets decrease anticancer potential of Satureja khuzestanica in HepG2 hepatocellular carcinoma cells. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
7
|
Abstract
Survivin is one of the rare proteins that is differentially expressed in normal and cancer cells and is directly or indirectly involved in numerous pathways required for tumor maintenance. It is expressed in almost all cancers and its expression has been detected at early stages of cancer. These traits make survivin an exceptionally attractive target for cancer therapeutics. Even with these promising features to be an oncotherapeutic target, there has been limited success in the clinical trials targeting survivin. Only recently it has emerged that survivin was not being specifically targeted which could have resulted in the negative clinical outcome. Also, focus of research has now shifted from survivin expression in the overall heterogeneous tumor cell populations to survivin expression in cancer stem cells as these cells have proved to be the major drivers of tumors. Therefore, in this review we have analyzed the expression of survivin in normal and cancer cells with a particular focus on its expression in cancer stem cell compartment. We have discussed the major signaling pathways involved in regulation of survivin. We have explored the current development status of various types of interventions for inhibition of survivin. Furthermore, we have discussed the challenges involving the development of potent and specific survivin inhibitors for cancer therapeutics. Finally we have given insights for some of the promising future anticancer treatments.
Collapse
|
8
|
Inhibition of Cervical Cancer Cell Line Hela by Human Wharton’s Jelly Stem Cells Through Induction of Apoptosis. ACTA ACUST UNITED AC 2020. [DOI: 10.5812/gct.99206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
9
|
Rapozzi V, D’Este F, Xodo LE. Molecular pathways in cancer response to photodynamic therapy. J PORPHYR PHTHALOCYA 2019. [DOI: 10.1142/s1088424619300064] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This minireview describes the complexity of the molecular mechanisms involved in the tumor response to photodynamic treatment (PDT). Different aspects of reactive oxygen (ROS) and nitrogen species (RNS) induced by PDT will be examined. In particular, we will discuss the effect of ROS and RNS on cell compartments and the main mechanisms of cell death induced by the treatment. Moreover, we will also examine host defense mechanisms as well as resistance to PDT.
Collapse
Affiliation(s)
- Valentina Rapozzi
- Department of Medicine, University of Udine, P.le Kolbe 4, Udine, 33100, Italy
| | - Francesca D’Este
- Department of Medicine, University of Udine, P.le Kolbe 4, Udine, 33100, Italy
| | - Luigi E. Xodo
- Department of Medicine, University of Udine, P.le Kolbe 4, Udine, 33100, Italy
| |
Collapse
|
10
|
Yan L, Zhang W, Zhang B, Xuan C, Wang D. BIIB021: A novel inhibitor to heat shock protein 90–addicted oncology. Tumour Biol 2017; 39:1010428317698355. [PMID: 28443462 DOI: 10.1177/1010428317698355] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heat shock protein 90 is induced in response to the cell stress. Its overexpression has been reported in many cancers with poor prognosis. It acts as a chaperone to the client proteins, especially the activated oncoproteins in malignancies to protect them from degradation. Heat shock protein 90 inhibition represented anti-cancer effects in many studies. Previous natural product–based compounds are limited by their association with target toxicities. BIIB021 is an orally available, fully synthetic novel small-molecule heat shock protein 90 inhibitor that has shown strong antitumor activities in a large number of preclinical models and is now under clinical investigation. This review will summarize its therapeutic effects and highlight the prospect of targeting heat shock protein 90 in the cancer therapy.
Collapse
Affiliation(s)
- Liang Yan
- Department of Oncology, Binzhou People’s Hospital, Binzhou, People’s Republic of China
| | - Weiming Zhang
- Department of Oncology, The Affiliated Hospital of Binzhou Medical College, Binzhou, People’s Republic of China
| | - Beibei Zhang
- Department of Molecular Microbiology, Oslo University Hospital, Oslo, Norway
| | - Chao Xuan
- Department of Clinical Laboratory, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, People’s Republic of China
| | - Daogang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| |
Collapse
|
11
|
Khan Z, Khan AA, Yadav H, Prasad GBKS, Bisen PS. Survivin, a molecular target for therapeutic interventions in squamous cell carcinoma. Cell Mol Biol Lett 2017; 22:8. [PMID: 28536639 PMCID: PMC5415770 DOI: 10.1186/s11658-017-0038-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 03/27/2017] [Indexed: 12/14/2022] Open
Abstract
Squamous cell carcinoma (SCC) is the most common cancer worldwide. The treatment of locally advanced disease generally requires various combinations of radiotherapy, surgery, and systemic therapy. Despite aggressive multimodal treatment, most of the patients relapse. Identification of molecules that sustain cancer cell growth and survival has made molecular targeting a feasible therapeutic strategy. Survivin is a member of the Inhibitor of Apoptosis Protein (IAP) family, which is overexpressed in most of the malignancies including SCC and totally absent in most of the normal tissues. This feature makes survivin an ideal target for cancer therapy. It orchestrates several important mechanisms to support cancer cell survival including inhibition of apoptosis and regulation of cell division. Overexpression of survivin in tumors is also associated with poor prognosis, aggressive tumor behavior, resistance to therapy, and high tumor recurrence. Various strategies have been developed to target survivin expression in cancer cells, and their effects on apoptosis induction and tumor growth attenuation have been demonstrated. In this review, we discuss recent advances in therapeutic potential of survivin in cancer treatment.
Collapse
Affiliation(s)
- Zakir Khan
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474001 MP India.,Department of Biomedical Sciences, Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Abdul Arif Khan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hariom Yadav
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | | | - Prakash Singh Bisen
- School of Studies in Biotechnology, Jiwaji University, Gwalior, 474001 MP India
| |
Collapse
|
12
|
Mohammadian M, Zeynali S, Azarbaijani AF, Khadem Ansari MH, Kheradmand F. Cytotoxic effects of the newly-developed chemotherapeutic agents 17-AAG in combination with oxaliplatin and capecitabine in colorectal cancer cell lines. Res Pharm Sci 2017; 12:517-525. [PMID: 29204180 PMCID: PMC5691578 DOI: 10.4103/1735-5362.217432] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The use of heat shock protein 90 inhibitors like 17-allylamino-17-demethoxy-geldanamycin (17-AAG) has been recently introduced as an attractive anticancer therapy. It has been shown that 17-AAG may potentiate the inhibitory effects of some classical anticolorectal cancer (CRC) agents. In this study, two panels of colorectal carcinoma cell lines were used to evaluate the effects of 17-AAG in combination with capecitabine and oxaliplatin as double and triple combination therapies on the proliferation of CRC cell lines. HT-29 and all HCT-116 cell lines were seeded in culture media in the presence of different doses of the mentioned drugs in single, double, and triple combinations. Water-soluble tetrazolium-1 (WST-1) assay was used to investigate cell proliferation 24 h after treatments. Then, dose-response curves were plotted using WST-1outputs, and IC50 values were determined. For double and triple combinations respectively 0.5 × IC50 and 0.25 × IC50 were used. Data was analyzed with the software CompuSyn. Drug interactions were analyzed using Chou-Talalay method to calculate the combination index (CI).The data revealed that 17-AAG shows a potent synergistic interaction (CI < 1) with oxaliplatin and capecitabine in double combinations (0.5 × IC50) in both cell lines. In the case of triple combinations, the findings showed an antagonistic interaction (CI > 1) in HT-29 and a synergistic effect (CI < 1) in HCT-116 (0.25 × IC50) cell lines. It was concluded that double combinations of 17-AAG with oxaliplatin or capecitabine might be effective against HCT-116 and HT-29 cell lines. However, in triple combinations, positive results were seen only against HCT-116. Further investigation is suggested to confirm the effectiveness of these combinations in clinical trials.
Collapse
Affiliation(s)
- Mahshid Mohammadian
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical sciences, Urmia, I.R. Iran
| | - Shima Zeynali
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical sciences, Urmia, I.R. Iran
| | - Anahita Fathi Azarbaijani
- Department of Pharmaceutics, School of Pharmacy, Urmia University of Medical sciences, Urmia, I.R. Iran
| | | | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical sciences, Urmia, I.R. Iran.,Solid Tumor Research Center and Cellular and Molecular Research Center, Urmia University of Medical sciences, Urmia, I.R. Iran
| |
Collapse
|
13
|
Peng CL, Chen YI, Liu HJ, Lee PC, Luo TY, Shieh MJ. A novel temperature-responsive micelle for enhancing combination therapy. Int J Nanomedicine 2016; 11:3357-69. [PMID: 27524894 PMCID: PMC4966578 DOI: 10.2147/ijn.s100469] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
A novel thermosensitive polymer p(N-isopropylacrylamide-co-poly[ethylene glycol] methyl ether acrylate)-block-poly(epsilon-caprolactone), p(NIPAAM-co-PEGMEA)-b-PCL, was synthesized and developed as nanomicelles. The hydrophobic heat shock protein 90 inhibitor 17-allylamino-17-demethoxygeldanamycin and the photosensitizer cyanine dye infrared-780 were loaded into the core of the micelles to achieve both chemotherapy and photothermal therapy simultaneously at the tumor site. The release of the drug could be controlled by varying the temperature due to the thermosensitive nature of the micelles. The micelles were less than 200 nm in size, and the drug encapsulation efficiency was >50%. The critical micelle concentrations were small enough to allow micelle stability upon dilution. Data from cell viability and animal experiments indicate that this combination treatment using photothermal therapy with chemotherapy had synergistic effects while decreasing side effects.
Collapse
Affiliation(s)
- Cheng-Liang Peng
- Isotope application Division, Institute of Nuclear energy research, Taoyuan
| | - Yuan-I Chen
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei; Department of Oncology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Hung-Jen Liu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei
| | - Pei-Chi Lee
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei
| | - Tsai-Yueh Luo
- Isotope application Division, Institute of Nuclear energy research, Taoyuan
| | - Ming-Jium Shieh
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei; Department of Oncology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| |
Collapse
|
14
|
Kao CY, Yang PM, Wu MH, Huang CC, Lee YC, Lee KH. Heat shock protein 90 is involved in the regulation of HMGA2-driven growth and epithelial-to-mesenchymal transition of colorectal cancer cells. PeerJ 2016; 4:e1683. [PMID: 26893968 PMCID: PMC4756735 DOI: 10.7717/peerj.1683] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/21/2016] [Indexed: 01/05/2023] Open
Abstract
High Mobility Group AT-hook 2 (HMGA2) is a nonhistone chromatin-binding protein which acts as a transcriptional regulating factor involved in gene transcription. In particular, overexpression of HMGA2 has been demonstrated to associate with neoplastic transformation and tumor progression in Colorectal Cancer (CRC). Thus, HMGA2 is a potential therapeutic target in cancer therapy. Heat Shock Protein 90 (Hsp90) is a chaperone protein required for the stability and function for a number of proteins that promote the growth, mobility, and survival of cancer cells. Moreover, it has shown strong positive connections were observed between Hsp90 inhibitors and CRC, which indicated their potential for use in CRC treatment by using combination of data mining and experimental designs. However, little is known about the effect of Hsp90 inhibition on HMGA2 protein expression in CRC. In this study, we tested the hypothesis that Hsp90 may regulate HMGA2 expression and investigated the relationship between Hsp90 and HMGA2 signaling. The use of the second-generation Hsp90 inhibitor, NVP-AUY922, considerably knocked down HMGA2 expression, and the effects of Hsp90 and HMGA2 knockdown were similar. In addition, Hsp90 knockdown abrogates colocalization of Hsp90 and HMGA2 in CRC cells. Moreover, the suppression of HMGA2 protein expression in response to NVP-AUY922 treatment resulted in ubiquitination and subsequent proteasome-dependant degradation of HMGA2. Furthermore, RNAi-mediated silencing of HMGA2 reduced the survival of CRC cells and increased the sensitivity of these cells to chemotherapy. Finally, we found that the NVP-AUY922-dependent mitigation of HMGA2 signaling occurred also through indirect reactivation of the tumor suppressor microRNA (miRNA), let-7a, or the inhibition of ERK-regulated HMGA2 involved in regulating the growth of CRC cells. Collectively, our studies identify the crucial role for the Hsp90-HMGA2 interaction in maintaining CRC cell survival and migration. These findings have significant implications for inhibition HMGA2-dependent tumorigenesis by clinically available Hsp90 inhibitors.
Collapse
Affiliation(s)
- Chun-Yu Kao
- Department of Pediatric Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Pei-Ming Yang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Heng Wu
- Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chi-Chen Huang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chao Lee
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
15
|
Rodríguez ME, Cogno IS, Milla Sanabria LS, Morán YS, Rivarola VA. Heat shock proteins in the context of photodynamic therapy: autophagy, apoptosis and immunogenic cell death. Photochem Photobiol Sci 2016; 15:1090-1102. [DOI: 10.1039/c6pp00097e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heat shock proteins can mediate resistance to photodynamic therapy by inhibiting apoptosis and modulating autophagy which, in turn, prevents apoptosis and immunogenic cell death.
Collapse
Affiliation(s)
- Matías E. Rodríguez
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Ingrid S. Cogno
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Laura S. Milla Sanabria
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Yanina S. Morán
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| | - Viviana A. Rivarola
- Departamento de Biología Molecular
- Universidad Nacional de Río Cuarto
- Río Cuarto (5800)
- Argentina
| |
Collapse
|
16
|
Anand N, Kanwar RK, Dubey ML, Vahishta RK, Sehgal R, Verma AK, Kanwar JR. Effect of lactoferrin protein on red blood cells and macrophages: mechanism of parasite-host interaction. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3821-35. [PMID: 26251568 PMCID: PMC4524381 DOI: 10.2147/dddt.s77860] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Lactoferrin is a natural multifunctional protein known to have antitumor, antimicrobial, and anti-inflammatory activity. Apart from its antimicrobial effects, lactoferrin is known to boost the immune response by enhancing antioxidants. Lactoferrin exists in various forms depending on its iron saturation. The present study was done to observe the effect of lactoferrin, isolated from bovine and buffalo colostrum, on red blood cells (RBCs) and macrophages (human monocytic cell line-derived macrophages THP1 cells). Methods Lactoferrin obtained from both species and in different iron saturation forms were used in the present study, and treatment of host cells were given with different forms of lactoferrin at different concentrations. These treated host cells were used for various studies, including morphometric analysis, viability by MTT assay, survivin gene expression, production of reactive oxygen species, phagocytic properties, invasion assay, and Toll-like receptor-4, Toll-like receptor-9, and MDR1 expression, to investigate the interaction between lactoferrin and host cells and the possible mechanism of action with regard to parasitic infections. Results The mechanism of interaction between host cells and lactoferrin have shown various aspects of gene expression and cellular activity depending on the degree of iron saturation of lactoferrin. A significant increase (P<0.05) in production of reactive oxygen species, phagocytic activity, and Toll-like receptor expression was observed in host cells incubated with iron-saturated lactoferrin when compared with an untreated control group. However, there was no significant (P>0.05) change in percentage viability in the different groups of host cells treated, and no downregulation of survivin gene expression was found at 48 hours post-incubation. Upregulation of the Toll-like receptor and downregulation of the P-gp gene confirmed the immunomodulatory potential of lactoferrin protein. Conclusion The present study details the interaction between lactoferrin and parasite host cells, ie, RBCs and macrophages, using various cellular processes and expression studies. The study reveals the possible mechanism of action against various intracellular pathogens such as Toxoplasma, Plasmodium, Leishmania, Trypanosoma, and Mycobacterium. The presence of iron in lactoferrin plays an important role in enhancing the various activities taking place inside these cells. This work provides a lot of information about targeting lactoferrin against many parasitic infections which can rule out the exact pathways for inhibition of diseases caused by intracellular microbes mainly targeting RBCs and macrophages for their survival. Therefore, this initial study can serve as a baseline for further evaluation of the mechanism of action of lactoferrin against parasitic diseases, which is not fully understood to date.
Collapse
Affiliation(s)
- Namrata Anand
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rupinder K Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research, School of Medicine, Molecular and Medical Research Strategic Research Centre, Faculty of Health, Deakin University, Geelong, VIC, Australia
| | - Mohan Lal Dubey
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - R K Vahishta
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh
| | - Rakesh Sehgal
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anita K Verma
- Nanobiotech Laboratory, Department of Zoology, Kirorimal College, University of Delhi, Delhi, India
| | - Jagat R Kanwar
- Nanomedicine Laboratory of Immunology and Molecular Biomedical Research, School of Medicine, Molecular and Medical Research Strategic Research Centre, Faculty of Health, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
17
|
Adeno-associated virus mediated gene transfer of Shepherdin inhibits gallbladder carcinoma growth in vitro and in vivo. Gene 2015; 572:87-94. [PMID: 26143116 DOI: 10.1016/j.gene.2015.06.080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 01/08/2023]
Abstract
Gene therapy, a significantly crucial strategy for treatment of malignancies, has been gradually accepted in recent years. However, this therapeutic approach has being facing great challenges concerning problems which include complicated development of cancer with multiple gene control, effective target shortage, low efficiency of gene transferring and safety of the vector delivery system. Shepherdin, a novel peptidomimetic molecule designed from Lys-79 to Leu-87 of survivin, has been identified as a tumor suppressor with the function that can not only competitively interfere with the interaction between survivin and Hsp90 (heat shock protein-90) leading to the degradation of survivin to anti-tumor, but also competitively target the ATP-dependent binding pocket of Hsp90 resulting in the dysfunction of Hsp90 chaperone to cell apoptosis via a mitochondrial dependent or independent pathway. In the present study, we designed and constructed a recombinant Adeno-associated virus (rAAV) loading fusion gene NT4-TAT-6His-Shepherdin. The expression of Shepherdin in gallbladder carcinoma (GBC) cells was detected and its strong inhibitory effects against GBC growth were evaluated after AAV mediated gene transfer of Shepherdin into GBC cells and xenograft tumors. MTT assay and flow cytometric analysis demonstrated that rAAV containing Shepherdin gene could significantly inhibit the growth of GBC and this effect was closely associated with apoptosis. These results indicated that rAAV-NT4-TAT-6His-Shepherdin may be considered a novel therapeutic strategy in the gene therapy for gallbladder carcinoma.
Collapse
|
18
|
Data supporting the identification of compound for inhibition of survivin of colorectal cancer by using ingenuity pathway analysis of gene expression profiling of colorectal cancer tissues. Data Brief 2015. [PMID: 26217796 PMCID: PMC4510466 DOI: 10.1016/j.dib.2015.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The data in this article is related to the research article entitled, “Targeting of Multiple Oncogenic Signaling Pathways by Hsp90 Inhibitor Alone or in Combination with Berberine for Treatment of Colorectal Cancer” [1]. Overexpression of survivin induces resistance to various anticancer therapies such as chemotherapy and radiation therapy in colorectal cancer (CRC) cells. To determine significant correlations of biological functions/pathways with survivin, 4567 significant genes were analyzed from the GEO DataSet (GSE21815) of CRC and these were overlaid onto a global molecular network developed from information contained in the Ingenuity Pathway Analysis (IPA) database. The data here present the most significant disease and disordered biological functions, significant molecular/cellular functions and significant categories in physiological development/system functions which were associated with CRC. The top 10 canonical signaling pathways associated with CRC were categorize in order based on the level of statistical significance.
Collapse
|
19
|
Su YH, Tang WC, Cheng YW, Sia P, Huang CC, Lee YC, Jiang HY, Wu MH, Lai IL, Lee JW, Lee KH. Targeting of multiple oncogenic signaling pathways by Hsp90 inhibitor alone or in combination with berberine for treatment of colorectal cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2261-72. [PMID: 25982393 DOI: 10.1016/j.bbamcr.2015.05.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/24/2015] [Accepted: 05/08/2015] [Indexed: 12/24/2022]
Abstract
There is a wide range of drugs and combinations under investigation and/or approved over the last decade to treat colorectal cancer (CRC), but the 5-year survival rate remains poor at stages II-IV. Therefore, new, more-efficient drugs still need to be developed that will hopefully be included in first-line therapy or overcome resistance when it appears, as part of second- or third-line treatments in the near future. In this study, we revealed that heat shock protein 90 (Hsp90) inhibitors have high therapeutic potential in CRC according to combinative analysis of NCBI's Gene Expression Omnibus (GEO) repository and chemical genomic database of Connectivity Map (CMap). We found that second generation Hsp90 inhibitor, NVP-AUY922, significantly downregulated the activities of a broad spectrum of kinases involved in regulating cell growth arrest and death of NVP-AUY922-sensitive CRC cells. To overcome NVP-AUY922-induced upregulation of survivin expression which causes drug insensitivity, we found that combining berberine (BBR), a herbal medicine with potency in inhibiting survivin expression, with NVP-AUY922 resulted in synergistic antiproliferative effects for NVP-AUY922-sensitive and -insensitive CRC cells. Furthermore, we demonstrated that treatment of NVP-AUY922-insensitive CRC cells with the combination of NVP-AUY922 and BBR caused cell growth arrest through inhibiting CDK4 expression and induction of microRNA-296-5p (miR-296-5p)-mediated suppression of Pin1-β-catenin-cyclin D1 signaling pathway. Finally, we found that the expression level of Hsp90 in tumor tissues of CRC was positively correlated with CDK4 and Pin1 expression levels. Taken together, these results indicate that combination of NVP-AUY922 and BBR therapy can inhibit multiple oncogenic signaling pathways of CRC.
Collapse
Affiliation(s)
- Yen-Hao Su
- Department of Surgery, Taipei Medical University-Shuang Ho Hospital, New Taipei City, Taiwan
| | - Wan-Chun Tang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ya-Wen Cheng
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Peik Sia
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chi-Chen Huang
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chao Lee
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Yi Jiang
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ming-Heng Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - I-Lu Lai
- Division of Medicinal Chemistry, College of Pharmacy and Comprehensive Cancer Center, Ohio State University, Columbus, OH, USA
| | - Jun-Wei Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Kuen-Haur Lee
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
20
|
Singh N, Krishnakumar S, Kanwar RK, Cheung CHA, Kanwar JR. Clinical aspects for survivin: a crucial molecule for targeting drug-resistant cancers. Drug Discov Today 2015; 20:578-87. [DOI: 10.1016/j.drudis.2014.11.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/12/2014] [Accepted: 11/20/2014] [Indexed: 12/24/2022]
|
21
|
Zhou X, Fan LX, Peters DJM, Trudel M, Bradner JE, Li X. Therapeutic targeting of BET bromodomain protein, Brd4, delays cyst growth in ADPKD. Hum Mol Genet 2015; 24:3982-93. [PMID: 25877301 DOI: 10.1093/hmg/ddv136] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023] Open
Abstract
In this study, we identified a BET bromodomain (BRD) protein, Brd4, not only as a novel epigenetic regulator of autosomal dominant polycystic kidney disease (ADPKD) but also as a novel client protein of Hsp90. We found that Brd4 was upregulated in Pkd1 mutant mouse renal epithelial cells and tissues. This upregulation of Brd4 appears to result from the chaperone activity of Hsp90 and escape proteasomal degradation. We further identify that Brd4 is an upstream regulator of the expression of c-Myc which has been upregulated in all rodent models of PKD and ADPKD patients with unknown mechanism. Inhibition of Brd4 in Pkd1 mutant renal epithelial cells with JQ1, a selective small-molecular inhibitor of BET BRD protein(s), (1) decreased the levels of c-Myc mRNA and protein; (2) increased the levels of p21 mRNA and protein, which was transcriptionally repressed by c-Myc; (3) decreased the phosphorylation of Rb; and (4) decreased cystic epithelial cell proliferation as shown by inhibition of S-phase entry. Most importantly, treatment with JQ1 strikingly delayed cyst growth and kidney enlargement, and preserved renal function in two early stage genetic mouse strains with Pkd1 mutations. This study not only provides one of the mechanisms of how c-Myc is upregulated in PKD but also suggests that targeting Brd4 with JQ1 may function as a novel epigenetic approach in ADPKD. The unraveled link between Brd4 and Hsp90 in ADPKD may also be a general mechanism for the upregulation of Brd4 in cancer cells and opens up avenues for combination therapies against ADPKD and cancer.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Internal Medicine, Kidney Institute
| | - Lucy X Fan
- Department of Internal Medicine, Kidney Institute
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Trudel
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montreal, Université de Montreal, Faculté de Médecine, Montreal, Quebec, Canada and
| | - James E Bradner
- Department of Medicine, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaogang Li
- Department of Internal Medicine, Kidney Institute, Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA,
| |
Collapse
|
22
|
Lazenby M, Hills R, Burnett AK, Zabkiewicz J. The HSP90 inhibitor ganetespib: A potential effective agent for Acute Myeloid Leukemia in combination with cytarabine. Leuk Res 2015; 39:617-24. [PMID: 25882550 PMCID: PMC4452084 DOI: 10.1016/j.leukres.2015.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 02/23/2015] [Accepted: 03/19/2015] [Indexed: 01/21/2023]
Abstract
HSP90 is a multi-client chaperone involved in regulating a large array of cellular processes and is commonly overexpressed in many different cancer types including hematological malignancies. Inhibition of HSP90 holds promise for targeting multiple molecular abnormalities and is therefore an attractive target for heterogeneous malignancies such as Acute Myeloid Leukemia (AML). Ganetespib is a highly potent second generation HSP90 inhibitor which we show is significantly more effective against primary AML blasts at nanomolar concentrations when compared with cytarabine (p<0.001). Dose dependant cytotoxicity was observed with an apoptotic response coordinate with the loss of pro-survival signaling through the client protein AKT. Combination treatment of primary blasts with ganetespib and cytarabine showed good synergistic interaction (combination index (CI): 0.47) across a range of drug effects with associated reduction in HSP70 feedback and AKT signaling levels. In summary, we show ganetespib to have high activity in primary AMLs as a monotherapy and a synergistic relationship with cytarabine when combined. The combination of cytotoxic cell death, suppression of cytoprotective/drug resistance mechanisms such as AKT and reduced clinical toxicity compared to other HSP90 inhibitors provide strong rationale for the clinical assessment of ganetespib in AML.
Collapse
Affiliation(s)
- M Lazenby
- Cardiff Experimental Cancer Medicine Centre (ECMC), Department of Haematology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - R Hills
- Cardiff Experimental Cancer Medicine Centre (ECMC), Department of Haematology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - A K Burnett
- Cardiff Experimental Cancer Medicine Centre (ECMC), Department of Haematology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - J Zabkiewicz
- Cardiff Experimental Cancer Medicine Centre (ECMC), Department of Haematology, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
23
|
Bongiovanni L, Romanucci M, Malatesta D, D’Andrea A, Ciccarelli A, Della Salda L. Survivin and Related Proteins in Canine Mammary Tumors. Vet Pathol 2014; 52:269-75. [DOI: 10.1177/0300985814529312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Survivin is reexpressed in most human breast cancers, where its expression has been associated with tumor aggressiveness, poor prognosis, and poor response to therapy. Survivin expression was evaluated in 41 malignant canine mammary tumors (CMTs) by immunohistochemistry, in relation to histological grade and stage, and correlated with that of some related molecules (β-catenin, caspase 3, heat shock proteins) to understand their possible role in canine mammary tumorigenesis. An increase in nuclear survivin expression, compared with healthy mammary glands, was observed in CMTs, where nuclear immunolabeling was related to the presence of necrosis. No statistically significant relation was found between the expression of the investigated molecules and the histological grade or stage. The present study may suggest an important involvement of survivin in CMT tumorigenesis. Its overexpression in most of the cases evaluated might suggest that targeting survivin in CMTs may be a valid anticancer therapy.
Collapse
Affiliation(s)
- L. Bongiovanni
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - M. Romanucci
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - D. Malatesta
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - A. D’Andrea
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - A. Ciccarelli
- Faculty of Political Sciences, University of Teramo, Teramo, Italy
| | - L. Della Salda
- Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| |
Collapse
|
24
|
Wang J, Li W. Discovery of novel second mitochondria-derived activator of caspase mimetics as selective inhibitor of apoptosis protein inhibitors. J Pharmacol Exp Ther 2014; 349:319-29. [PMID: 24623800 DOI: 10.1124/jpet.113.212019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Inhibitor of apoptosis (IAP) proteins are widely considered as promising cancer drug targets, especially for drug-resistant tumors. Mimicking the IAP-binding motif of second mitochondria-derived activator of caspases (SMAC) is a rational strategy to design potential IAP inhibitors. In this report, we used the bioactive conformation of AVPI tetrapeptide in the N terminus of SMAC as a template and performed a shape-based virtual screening against a drug-like compound library to identify novel IAP inhibitors. Top hits were subsequently docked to available IAP crystal structures as a secondary screening followed by validation using in vitro biologic assays. Four novel hit compounds were identified to potently inhibit cell growth in two human melanoma (A375 and M14) and two human prostate (PC-3 and DU145) cancer cell lines. The best compound, UC-112 [5-((benzyloxy)methyl)-7-(pyrrolidin-1-ylmethyl)quinolin-8-ol], has IC50 values ranging from 0.7 to 3.4 µM. UC-112 also potently inhibits the growth of P-glycoprotein (P-gp)-overexpressed multidrug-resistant cancer cells, strongly activates caspase-3/7 and caspase-9 activities, and selectively downregulates survivin level at a concentration as low as 1 µM. Coincubation of UC-112 with a known proteasome inhibitor Z-Leu-Leu-Leu-CHO (MG-132) rescued survivin inhibition, consistent with the anticipated mechanism of action for UC-112. As a single agent, UC-112 strongly inhibits tumor growth and reduces both X chromosome-linked IAP and survivin levels in an A375 human melanoma xenograft model in vivo. Overall, our study identified novel scaffolds, especially UC-112, as new platforms on which potent and selective IAP antagonists can be developed.
Collapse
Affiliation(s)
- Jin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, Tennessee
| | | |
Collapse
|
25
|
Potential targets for colorectal cancer prevention. Int J Mol Sci 2013; 14:17279-303. [PMID: 23975167 PMCID: PMC3794728 DOI: 10.3390/ijms140917279] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 12/18/2022] Open
Abstract
The step-wise development of colorectal neoplasia from adenoma to carcinoma suggests that specific interventions could delay or prevent the development of invasive cancer. Several key factors involved in colorectal cancer pathogenesis have already been identified including cyclooxygenase 2 (COX-2), nuclear factor kappa B (NF-κB), survivin and insulin-like growth factor-I (IGF-I). Clinical trials of COX-2 inhibitors have provided the “proof of principle” that inhibition of this enzyme can prevent the formation of colonic adenomas and potentially carcinomas, however concerns regarding the potential toxicity of these drugs have limited their use as a chemopreventative strategy. Curcumin, resveratrol and quercetin are chemopreventive agents that are able to suppress multiple signaling pathways involved in carcinogenesis and hence are attractive candidates for further research.
Collapse
|
26
|
Margarucci L, Monti MC, Cassiano C, Mozzicafreddo M, Angeletti M, Riccio R, Tosco A, Casapullo A. Chemical proteomics-driven discovery of oleocanthal as an Hsp90 inhibitor. Chem Commun (Camb) 2013; 49:5844-6. [PMID: 23703283 DOI: 10.1039/c3cc41858h] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hsp90, a key target in cancer therapy, has been identified as the main partner of oleocanthal, an olive oil bioactive compound. A combination of chemical and biological assays disclosed its mechanism of action at the molecular level.
Collapse
Affiliation(s)
- Luigi Margarucci
- Dipartimento di Farmacia, Università degli Studi di Salerno, Via Ponte don Melillo, 84084 Fisciano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Wu X, Marmarelis ME, Hodi FS. Activity of the heat shock protein 90 inhibitor ganetespib in melanoma. PLoS One 2013; 8:e56134. [PMID: 23418523 PMCID: PMC3572008 DOI: 10.1371/journal.pone.0056134] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/05/2013] [Indexed: 01/09/2023] Open
Abstract
Heat shock protein 90 (HSP90) is involved in the regulation of diverse biological processes such as cell signaling, proliferation and survival, and has been recently recognized as a potential target for cancer therapy. Ganetespib is a potent ATP competitive inhibitor of HSP90. Ganetespib downregulated the expression of multiple signal transducing molecules including EGFR, IGF-1R, c-Met, Akt, B-RAF and C-RAF, resulting in pronounced decrease in phosphorylation of Akt and Erk1/2 in a panel of five cutaneous melanoma cell lines including those harboring B-RAF and N-RAS mutations. Ganetespib exhibited potent antiproliferative activity on all five of these cell lines, with IC50 values between 37.5 and 84 nM. Importantly, Ganetespib is active on B-RAF mutated melanoma cells that have acquired resistance to B-RAF inhibition. Ganetespib induced apoptosis and cell cycle arrest at G1 and/or G2/M phase. Ganetespib induced cell cycle arrest was accompanied by altered expression of cyclin-dependent kinase inhibitor (CDKI) p21(Cip1) and p27(Kip1), cyclins B1, D1 and E, and/or cyclin-dependent kinases 1, 2 and 4. HSP90 is functionally important for melanoma cells and HSP90 inhibitors such as ganetespib could potentially be effective therapeutics for melanoma with various genetic mutations and acquired resistance to B-RAF inhibition.
Collapse
Affiliation(s)
- Xinqi Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Melanoma Disease Center, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts, United States of America
| | | | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Melanoma Disease Center, Dana-Farber/Brigham and Women’s Cancer Center, Boston, Massachusetts, United States of America
| |
Collapse
|
28
|
Abstract
Fragment-based drug discovery (FBDD) concerns the screening of low-molecular weight compounds against macromolecular targets of clinical relevance. These compounds act as starting points for the development of drugs. FBDD has evolved and grown in popularity over the past 15 years. In this paper, the rationale and technology behind the use of X-ray crystallography in fragment based screening (FBS) will be described, including fragment library design and use of synchrotron radiation and robotics for high-throughput X-ray data collection. Some recent uses of crystallography in FBS will be described in detail, including interrogation of the drug targets β-secretase, phenylethanolamine N-methyltransferase, phosphodiesterase 4A and Hsp90. These examples provide illustrations of projects where crystallography is straightforward or difficult, and where other screening methods can help overcome the limitations of crystallography necessitated by diffraction quality.
Collapse
Affiliation(s)
- Zorik Chilingaryan
- School of Chemistry, University of Wollongong, Northfields Ave, Wollongong 2522, NSW, Australia.
| | | | | |
Collapse
|
29
|
The apoptotic effect and associated signalling of HSP90 inhibitor 17-DMAG in hepatocellular carcinoma cells. Cell Biol Int 2012; 36:893-9. [DOI: 10.1042/cbi20110473] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
30
|
Wang Q, Shu R, He H, Wang L, Ma Y, Zhu H, Wang Z, Wang S, Shen G, Lei P. Co-silencing of Birc5 (survivin) and Hspa5 (Grp78) induces apoptosis in hepatoma cells more efficiently than single gene interference. Int J Oncol 2012; 41:652-60. [PMID: 22581315 DOI: 10.3892/ijo.2012.1471] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 02/28/2012] [Indexed: 11/05/2022] Open
Abstract
Birc5 (previously known as survivin) is a cancer-specific protein. Due to the upregulation of its expression in various human malignancies and its key role in apoptosis, proliferation and angiogenesis, Birc5 has attracted attention as a target for anticancer therapies. In this study, when Birc5 was silenced in HepG2 cells, 29.7±3.3% cells underwent apoptosis as expected. It was found that the expression levels of glucose-regulated protein 78 (Hspa5, previously known as Grp78) was increased by almost 3-fold in Birc5-silenced HepG2 cells. Hspa5, a master regulator of the anti-apoptotic unfolded protein response signalling network, can also promote tumor proliferation, survival and metastasis. Hence, we hypothesized that the co-silencing of Birc5 and Hspa5 may exert a stronger apoptosis-inducing effect than single gene interference. To verify this, the expression levels of Birc5 and Hspa5 in human hepatocellular carcinoma tissues were determined. Immunohistochemical staining showed that the expression of Birc5 and Hspa5 was elevated in 28 out of 31 samples. Additionally, plasmid-based siRNA against Birc5 and/or Hspa5 were constructed and transfected into the human hepatocellular liver carcinoma cell line, HepG2. Compared with the HepG2 cells, in which Birc5 or Hspa5 were silenced alone, only 44.2±3.4% of the co-silenced cells proliferated, and 40.3±3.7% co-silenced cells underwent apoptosis (p<0.05). Furthermore, tumor formation from inoculated subcutaneous co-silenced cells in nude mice was inhibited significantly. The current study suggests that Birc5 and Hspa5 could be important survival factors for hepatoma carcinoma cells and that the simultaneous knockdown of Birc5 and Hspa5 is more effective in inducing apoptosis in HepG2 cells than the knockdown of Birc5 or Hspa5 alone. The co-silencing of Birc5 and Hspa5 could be warranted for cancer therapy.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Heat shock protein 90 is a putative therapeutic target in patients with recurrent advanced-stage ovarian carcinoma with serous effusions. Hum Pathol 2012; 43:529-35. [PMID: 21864883 DOI: 10.1016/j.humpath.2011.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/23/2011] [Accepted: 05/24/2011] [Indexed: 01/08/2023]
|
32
|
Paraiso KHT, Haarberg HE, Wood E, Rebecca VW, Chen YA, Xiang Y, Ribas A, Lo RS, Weber JS, Sondak VK, John JK, Sarnaik AA, Koomen JM, Smalley KSM. The HSP90 inhibitor XL888 overcomes BRAF inhibitor resistance mediated through diverse mechanisms. Clin Cancer Res 2012; 18:2502-14. [PMID: 22351686 DOI: 10.1158/1078-0432.ccr-11-2612] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE The clinical use of BRAF inhibitors is being hampered by the acquisition of drug resistance. This study shows the potential therapeutic use of the HSP90 inhibitor (XL888) in six different models of vemurafenib resistance. EXPERIMENTAL DESIGN The ability of XL888 to inhibit growth and to induce apoptosis and tumor regression of vemurafenib-resistant melanoma cell lines was shown in vitro and in vivo. A novel mass spectrometry-based pharmacodynamic assay was developed to measure intratumoral HSP70 levels following HSP90 inhibition in melanoma cell lines, xenografts, and melanoma biopsies. Mechanistic studies were carried out to determine the mechanism of XL888-induced apoptosis. RESULTS XL888 potently inhibited cell growth, induced apoptosis, and prevented the growth of vemurafenib-resistant melanoma cell lines in 3-dimensional cell culture, long-term colony formation assays, and human melanoma mouse xenografts. The reversal of the resistance phenotype was associated with the degradation of PDGFRβ, COT, IGFR1, CRAF, ARAF, S6, cyclin D1, and AKT, which in turn led to the nuclear accumulation of FOXO3a, an increase in BIM (Bcl-2 interacting mediator of cell death) expression, and the downregulation of Mcl-1. In most resistance models, XL888 treatment increased BIM expression, decreased Mcl-1 expression, and induced apoptosis more effectively than dual mitogen-activated protein-extracellular signal-regulated kinase/phosphoinositide 3-kinase (MEK/PI3K) inhibition. CONCLUSIONS HSP90 inhibition may be a highly effective strategy at managing the diverse array of resistance mechanisms being reported to BRAF inhibitors and appears to be more effective at restoring BIM expression and downregulating Mcl-1 expression than combined MEK/PI3K inhibitor therapy.
Collapse
Affiliation(s)
- Kim H T Paraiso
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, Tampa, Florida, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mechanisms of Resistance to Hsp90 Inhibitor Drugs: A Complex Mosaic Emerges. Pharmaceuticals (Basel) 2011; 4:1400-1422. [PMID: 27721330 PMCID: PMC4060131 DOI: 10.3390/ph4111400] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 10/04/2011] [Accepted: 10/17/2011] [Indexed: 01/07/2023] Open
Abstract
The molecular chaperone Hsp90 holds great promise as a cancer drug target, despite some of the initial clinical trials of Hsp90 inhibitor drugs having not lived up to expectation. Effective use of these drugs will benefit greatly from a much more detailed understanding of the factors that contribute to resistance, whether intrinsic or acquired. We review how cell culture studies have revealed a number of different mechanisms whereby cells can be rendered less susceptible to the effects of Hsp90 inhibitor treatment. A major influence is Hsp90 inhibition causing strong induction of the heat shock response, a stress response that increases cellular levels of prosurvival chaperones such as Hsp27 and Hsp70. Another problem seems to be that these inhibitors do not always access the Hsp90 proteins of the mitochondrion, forms of Hsp90 that—in cancer cells—are operating to suppress apoptosis. It should be possible to overcome these drawbacks through the appropriate drug redesign or with the combinatorial use of an Hsp90 inhibitor with a drug that targets either heat shock factor or the chaperone Hsp70. Still though, cells will often differ in the key antiapoptotic versus proapoptotic activities that are dependent on Hsp90, in the key steps in their apoptotic pathways responsive to Hsp90 inhibition or Hsp70 level, as well as the extents to which their survival is dependent on oncogenic tyrosine kinases that are clients of Hsp90. A systems approach will therefore often be required in order to establish the most prominent effects of Hsp90 inhibition in each type of cancer cell.
Collapse
|
34
|
Management of cytoskeleton architecture by molecular chaperones and immunophilins. Cell Signal 2011; 23:1907-20. [PMID: 21864675 DOI: 10.1016/j.cellsig.2011.07.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 07/22/2011] [Accepted: 07/26/2011] [Indexed: 11/20/2022]
Abstract
Cytoskeletal structure is continually remodeled to accommodate normal cell growth and to respond to pathophysiological cues. As a consequence, several cytoskeleton-interacting proteins become involved in a variety of cellular processes such as cell growth and division, cell movement, vesicle transportation, cellular organelle location and function, localization and distribution of membrane receptors, and cell-cell communication. Molecular chaperones and immunophilins are counted among the most important proteins that interact closely with the cytoskeleton network, in particular with microtubules and microtubule-associated factors. In several situations, heat-shock proteins and immunophilins work together as a functionally active heterocomplex, although both types of proteins also show independent actions. In circumstances where homeostasis is affected by environmental stresses or due to genetic alterations, chaperone proteins help to stabilize the system. Molecular chaperones facilitate the assembly, disassembly and/or folding/refolding of cytoskeletal proteins, so they prevent aberrant protein aggregation. Nonetheless, the roles of heat-shock proteins and immunophilins are not only limited to solve abnormal situations, but they also have an active participation during the normal differentiation process of the cell and are key factors for many structural and functional rearrangements during this course of action. Cytoskeleton modifications leading to altered localization of nuclear factors may result in loss- or gain-of-function of such factors, which affects the cell cycle and cell development. Therefore, cytoskeletal components are attractive therapeutic targets, particularly microtubules, to prevent pathological situations such as rapidly dividing tumor cells or to favor the process of cell differentiation in other cases. In this review we will address some classical and novel aspects of key regulatory functions of heat-shock proteins and immunophilins as housekeeping factors of the cytoskeletal network.
Collapse
|
35
|
|
36
|
Kanwar JR, Kamalapuram SK, Kanwar RK. Targeting survivin in cancer: the cell-signalling perspective. Drug Discov Today 2011; 16:485-94. [PMID: 21511051 DOI: 10.1016/j.drudis.2011.04.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 02/23/2011] [Accepted: 04/01/2011] [Indexed: 01/12/2023]
Abstract
Survivin, a prominent anticancer target, is ubiquitously expressed in a plethora of cancers and the evolving complexity in functional regulation of survivin is yet to be deciphered. However, pertaining to the recent studies, therapeutic modulation of survivin is critically regulated by interaction with prominent cell-signalling pathways [HIF-1α, HSP90, PI3K/AKT, mTOR, ERK, tumour suppressor genes (p53, PTEN), oncogenes (Bcl-2, Ras)] and a wide range of growth factors (EGFR, VEGF, among others). In our article we discuss, in detail, an overview of the recent developments in the pharmacological modulation of survivin via cell-signalling paradigms and antisurvivin therapeutics, along with an outlook on therapeutic management of survivin in drug-resistant cancers.
Collapse
Affiliation(s)
- Jagat R Kanwar
- Laboratory of Immunology and Molecular Biomedical Research (LIMBR), Centre for Biotechnology and Interdisciplinary Biosciences (BioDeakin), Institute for Technology Research and Innovation (ITRI), Deakin University, Victoria, Australia.
| | | | | |
Collapse
|
37
|
Kanwar JR, Kamalapuram SK, Kanwar RK. Targeting survivin in cancer: patent review. Expert Opin Ther Pat 2011; 20:1723-37. [PMID: 21083520 DOI: 10.1517/13543776.2010.533657] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
IMPORTANCE OF THE FIELD Survivin is a prominent anti-apoptotic molecule expressed widely in the majority of cancers. Overexpression of survivin leads to uncontrolled cancer cell growth and drug resistance. Efficient downregulation of survivin expression and its functions can sensitise the tumour cells to various therapeutic interventions such as chemotherapeutic agents leading to cell apoptosis. AREAS COVERED IN THIS REVIEW The article thoroughly analyses up-to-date information on the knowledge generated from the survivin patents. Various key areas of research in terms of understanding survivin biology and its targeting are discussed in detail. WHAT THE READER WILL GAIN The article clearly gives an insight on the recent developments undertaken to understand the roles of survivin in cancer and in validating various treatment paradigms that suppress survivin expression in cancer cells. TAKE HOME MESSAGE Most recent developments are helpful for effectively downregulating survivin expression by using various therapeutic platforms such as chemotherapeutic drugs, immunotechnology, antisense, dominant negative survivin mutant, RNA interference and peptide-based methods. However, selective and specific targeting of survivin in cancer cells still poses a major challenge. Nanotechnology-based platforms are currently under development to enable site-specific targeting of survivin in tumour cells.
Collapse
Affiliation(s)
- Jagat R Kanwar
- Deakin University, Institute for Technology Research and Innovation (ITRI), Centre for Biotechnology and Interdisciplinary Biosciences (BioDeakin), Laboratory of Immunology and Molecular Biomedical Research (LIMBR), Waurn Ponds, Victoria 3217, Australia.
| | | | | |
Collapse
|
38
|
Kanwar JR, Mahidhara G, Kanwar RK. Antiangiogenic therapy using nanotechnological-based delivery system. Drug Discov Today 2011; 16:188-202. [DOI: 10.1016/j.drudis.2011.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 11/19/2010] [Accepted: 01/14/2011] [Indexed: 10/18/2022]
|
39
|
Allegra A, Sant'antonio E, Penna G, Alonci A, D'Angelo A, Russo S, Cannavò A, Gerace D, Musolino C. Novel therapeutic strategies in multiple myeloma: role of the heat shock protein inhibitors. Eur J Haematol 2010; 86:93-110. [PMID: 21114539 DOI: 10.1111/j.1600-0609.2010.01558.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite advances in understanding the molecular pathogenesis of multiple myeloma and promising new therapies, almost all patients eventually relapse with resistant disease. There is therefore a strong rationale for combining novel therapies that target intrinsic molecular pathways mediating multiple myeloma cell resistance. One such protein family is the heat shock proteins (HSP), especially the HSP90 family. Heat shock protein inhibitors have been identified as promising cancer treatments as, while they only inhibit a single biologic function, the chaperone-protein association, their effect is widespread as it results in the destruction of numerous client proteins. This article reviews the preclinical and clinical data, which support the testing of HSP90 inhibitors as cancer drugs and update the reader on the current status of the ongoing clinical trials of HSP90 inhibitors in multiple myeloma.
Collapse
|
40
|
Baratchi S, Kanwar RK, Kanwar JR. Survivin: A target from brain cancer to neurodegenerative disease. Crit Rev Biochem Mol Biol 2010; 45:535-54. [DOI: 10.3109/10409238.2010.516740] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|