1
|
Asdemir A, Özgür A. Molecular mechanism of anticancer effect of heat shock protein 90 inhibitor BIIB021 in human bladder cancer cell line. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5167-5177. [PMID: 38240781 PMCID: PMC11166791 DOI: 10.1007/s00210-024-02950-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/10/2024] [Indexed: 06/12/2024]
Abstract
Bladder cancer is a type of urologic malignancy that exhibits significant morbidity, mortality, and treatment costs. Inhibition of heat shock protein 90 (HSP90) activity has been a promising pharmacological strategy for blocking of bladder cancer pathogenesis. BIIB021 is a next-generation HSP90 inhibitor which interrupts ATP hydrolysis process of HSP90 and inhibits the stabilization and correct folding of client proteins. In current study, we aimed to investigate the molecular mechanism of the anticancer activity of BIIB021 in human bladder cancer T24 cells. Our results revealed that nanomolar concentration of BIIB021 decreased viability of T24 cell. BIIB021 downregulated HSP90 expression in T24 cells and inhibited the refolding activity of luciferase in the presence of T24 cell lysate. PCR array data indicated a significant alteration in transcript levels of cancer-related genes involved in metastases, apoptotic cell death, cell cycle, cellular senescence, DNA damage and repair mechanisms, epithelial-to-mesenchymal transition, hypoxia, telomeres and telomerase, and cancer metabolism pathways in T24 cells. All findings hypothesize that BIIB021 could exhibit as effective HSP90 inhibitor in the future for treatment of bladder cancer patients.
Collapse
Affiliation(s)
- Aydemir Asdemir
- Faculty of Medicine, Department of Urology, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Aykut Özgür
- Artova Vocational School, Department of Veterinary Medicine, Laboratory and Veterinary Health Program, Tokat Gaziosmanpasa University, Tokat, Turkey
| |
Collapse
|
2
|
Wei H, Zhang Y, Jia Y, Chen X, Niu T, Chatterjee A, He P, Hou G. Heat shock protein 90: biological functions, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e470. [PMID: 38283176 PMCID: PMC10811298 DOI: 10.1002/mco2.470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/30/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024] Open
Abstract
Heat shock protein 90 (Hsp90) is a predominant member among Heat shock proteins (HSPs), playing a central role in cellular protection and maintenance by aiding in the folding, stabilization, and modification of diverse protein substrates. It collaborates with various co-chaperones to manage ATPase-driven conformational changes in its dimer during client protein processing. Hsp90 is critical in cellular function, supporting the proper operation of numerous proteins, many of which are linked to diseases such as cancer, Alzheimer's, neurodegenerative conditions, and infectious diseases. Recognizing the significance of these client proteins across diverse diseases, there is a growing interest in targeting Hsp90 and its co-chaperones for potential therapeutic strategies. This review described biological background of HSPs and the structural characteristics of HSP90. Additionally, it discusses the regulatory role of heat shock factor-1 (HSF-1) in modulating HSP90 and sheds light on the dynamic chaperone cycle of HSP90. Furthermore, the review discusses the specific contributions of HSP90 in various disease contexts, especially in cancer. It also summarizes HSP90 inhibitors for cancer treatment, offering a thoughtful analysis of their strengths and limitations. These advancements in research expand our understanding of HSP90 and open up new avenues for considering HSP90 as a promising target for therapeutic intervention in a range of diseases.
Collapse
Affiliation(s)
- Huiyun Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Yingying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Yilin Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Xunan Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Tengda Niu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Aniruddha Chatterjee
- Department of PathologyDunedin School of MedicineUniversity of OtagoDunedinNew Zealand
| | - Pengxing He
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
| | - Guiqin Hou
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of EducationSchool of Pharmaceutical SciencesZhengzhou UniversityZhengzhouChina
- Department of PathologyDunedin School of MedicineUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
3
|
Alimardan Z, Abbasi M, Hasanzadeh F, Aghaei M, Khodarahmi G, Kashfi K. Heat shock proteins and cancer: The FoxM1 connection. Biochem Pharmacol 2023; 211:115505. [PMID: 36931349 PMCID: PMC10134075 DOI: 10.1016/j.bcp.2023.115505] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Heat shock proteins (Hsp) and FoxM1 have significant roles in carcinogenesis. According to their relative molecular weight, Hsps are divided into Hsp110, Hsp90, Hsp70, Hsp60, Hsp40, and small Hsps. Hsp70 can play essential functions in cancer initiation and is overexpressed in several human cancers. Hsp70, in combination with cochaperones HIP and HOP, refolds partially denatured proteins and acts as a cochaperone for Hsp90. Also, Hsp70, in combination with BAG3, regulates the FoxM1 signaling pathway. FoxM1 protein is a transcription factor of the Forkhead family that is overexpressed in most human cancers and is involved in many cancers' development features, including proliferation, migration, invasion, angiogenesis, metastasis, and resistance to apoptosis. This review discusses the Hsp70, Hsp90, and FoxM1 structure and function, the known Hsp70 cochaperones, and Hsp70, Hsp90, and FoxM1 inhibitors.
Collapse
Affiliation(s)
- Zahra Alimardan
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Pharmacology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Abbasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farshid Hasanzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahmud Aghaei
- Department of Biochemistry, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ghadamali Khodarahmi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, NY, USA.
| |
Collapse
|
4
|
Heterocyclic Compounds as Hsp90 Inhibitors: A Perspective on Anticancer Applications. Pharmaceutics 2022; 14:pharmaceutics14102220. [PMID: 36297655 PMCID: PMC9610671 DOI: 10.3390/pharmaceutics14102220] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
Heat shock proteins (Hsps) have garnered special attention in cancer therapy as molecular chaperones with regulatory/mediatory effects on folding, maintenance/stability, maturation, and conformation of proteins as well as their effects on prevention of protein aggregation. Hsp90 ensures the stability of various client proteins needed for the growth of cells or the survival of tumor cells; therefore, they are overexpressed in tumor cells and play key roles in carcinogenesis. Accordingly, Hsp90 inhibitors are recognized as attractive therapeutic agents for investigations pertaining to tumor suppression. Natural Hsp90 inhibitors comprising geldanamycin (GM), reclaimed analogs of GM including 17-AAG and DMAG, and radicicol, a natural macrocyclic antifungal, are among the first potent Hsp90 inhibitors. Herein, recently synthesized heterocyclic compounds recognized as potent Hsp90 inhibitors are reviewed along with the anticancer effects of heterocyclic compounds, comprising purine, pyrazole, triazine, quinolines, coumarin, and isoxazoles molecules.
Collapse
|
5
|
Tustumi F, Agareno GA, Galletti RP, da Silva RBR, Quintas JG, Sesconetto LDA, Szor DJ, Wolosker N. The Role of the Heat-Shock Proteins in Esophagogastric Cancer. Cells 2022; 11:2664. [PMID: 36078072 PMCID: PMC9454628 DOI: 10.3390/cells11172664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 01/05/2023] Open
Abstract
Heat-shock proteins (HSPs) are a family of proteins that have received considerable attention over the last several years. They have been classified into six prominent families: high-molecular-mass HSP, 90, 70, 60, 40, and small heat shock proteins. HSPs participate in protein folding, stability, and maturation of several proteins during stress, such as in heat, oxidative stress, fever, and inflammation. Due to the immunogenic host's role in the combat against cancer cells and the role of the inflammation in the cancer control or progression, abnormal expression of these proteins has been associated with many types of cancer, including esophagogastric cancer. This study aims to review all the evidence concerning the role of HSPs in the pathogenesis and prognosis of esophagogastric cancer and their potential role in future treatment options. This narrative review gathers scientific evidence concerning HSPs in relation to esophagus and gastric cancer. All esophagogastric cancer subtypes are included. The role of HSPs in carcinogenesis, prognostication, and therapy for esophagogastric cancer are discussed. The main topics covered are premalignant conditions for gastric cancer atrophic gastritis, Barrett esophagus, and some viral infections such as human papillomavirus (HPV) and Epstein-Barr virus (EBV). HSPs represent new perspectives on the development, prognostication, and treatment of esophagogastric cancer.
Collapse
Affiliation(s)
- Francisco Tustumi
- Department of Gastroenterology, Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 255, São Paulo 05403-000, SP, Brazil
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Gabriel Andrade Agareno
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Ricardo Purchio Galletti
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Rafael Benjamim Rosa da Silva
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Julia Grams Quintas
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Lucas de Abreu Sesconetto
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Daniel José Szor
- Department of Gastroenterology, Universidade de São Paulo, Av. Dr. Enéas Carvalho de Aguiar, 255, São Paulo 05403-000, SP, Brazil
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| | - Nelson Wolosker
- Department of Surgery, Hospital Israelita Albert Einstein, Av. Albert Einstein, 627, São Paulo 05652-900, SP, Brazil
| |
Collapse
|
6
|
Abstract
INTRODUCTION Heat shock proteins (HSPs) constitute a large family of proteins involved in protein folding and maturation. HSP expression is induced by heat shock or other stressors including cellular damage and hypoxia. The major groups, which are classified based on their molecular weight, include HSP27, HSP40, HSP60, HSP70, HSP90, and large HSP (HSP110 and glucose-regulated protein 170). HSPs play a significant role in cellular proliferation, differentiation, survival, apoptosis, and carcinogenesis. The human HSP90 family consists of five members and has a strong association with cancer. OBJECTIVES The primary objective is to review the important functions of heat shock protein 90 in cancer, especially as an anti-cancer drug target. RESULTS The HSP90 proteins not only play important roles in cancer development, progression, and metastasis, but also have potential clinical use as biomarkers for cancer diagnosis or assessing disease progression, and as therapeutic targets for cancer therapy. In this chapter, we discuss the roles of HSP90 in cancer biology and pharmacology, focusing on HSP90 as an anti-cancer drug target. An understanding of the functions and molecular mechanisms of HSP90 is critical for enhancing the accuracy of cancer diagnosis as well as for developing more effective and less toxic chemotherapeutic agents. CONCLUSION We have provided an overview of the complex relationship between cancer and HSP90. HSP90 proteins play an important role in tumorigenesis and may be used as potential clinical biomarkers for the diagnosis and predicting prognostic outcome of patients with cancer. HSP90 proteins may be used as therapeutic targets for cancer therapy, prompting discovery and development of novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Anthony Aswad
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, United States
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV, United States.
| |
Collapse
|
7
|
Mehta RK, Pal S, Kondapi K, Sitto M, Dewar C, Devasia T, Schipper MJ, Thomas DG, Basrur V, Pai MP, Morishima Y, Osawa Y, Pratt WB, Lawrence TS, Nyati MK. Low-Dose Hsp90 Inhibitor Selectively Radiosensitizes HNSCC and Pancreatic Xenografts. Clin Cancer Res 2020; 26:5246-5257. [PMID: 32718999 PMCID: PMC7541797 DOI: 10.1158/1078-0432.ccr-19-3102] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 04/21/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Treatment approaches using Hsp90 inhibitors at their maximum tolerated doses (MTDs) have not produced selective tumor toxicity. Inhibition of Hsp90 activity causes degradation of client proteins including those involved in recognizing and repairing DNA lesions. We hypothesized that if DNA repair proteins were degraded by concentrations of an Hsp90 inhibitor below those required to cause nonspecific cytotoxicity, significant tumor-selective radiosensitization might be achieved. EXPERIMENTAL DESIGN Tandem mass tagged-mass spectrometry was performed to determine the effect of a subcytotoxic concentration of the Hsp90 inhibitor, AT13387 (onalespib), on global protein abundance. The effect of AT13387 on in vitro radiosensitization was assessed using a clonogenic assay. Pharmacokinetics profiling was performed in mice bearing xenografts. Finally, the effect of low-dose AT13387 on the radiosensitization of three tumor models was assessed. RESULTS A subcytotoxic concentration of AT13387 reduced levels of DNA repair proteins, without affecting the majority of Hsp90 clients. The pharmacokinetics study using one-third of the MTD showed 40-fold higher levels of AT13387 in tumors compared with plasma. This low dose enhanced Hsp70 expression in peripheral blood mononuclear cells (PBMCs), which is a biomarker of Hsp90 inhibition. Low dose monotherapy was ineffective, but when combined with radiotherapy, produced significant tumor growth inhibition. CONCLUSIONS This study shows that a significant therapeutic ratio can be achieved by a low dose of Hsp90 inhibitor in combination with radiotherapy. Hsp90 inhibition, even at a low dose, can be monitored by measuring Hsp70 expression in PBMCs in human studies.
Collapse
Affiliation(s)
- Ranjit K Mehta
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Sanjima Pal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Koushik Kondapi
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Merna Sitto
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Cuyler Dewar
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Theresa Devasia
- School of Public Health, University of Michigan, Ann Arbor, Michigan
| | | | - Dafydd G Thomas
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Venkatesha Basrur
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Manjunath P Pai
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| | | | - Yoichi Osawa
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - William B Pratt
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Mukesh K Nyati
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
8
|
Zhang J, Si J, Gan L, Zhou R, Guo M, Zhang H. Harnessing the targeting potential of differential radiobiological effects of photon versus particle radiation for cancer treatment. J Cell Physiol 2020; 236:1695-1711. [PMID: 32691425 DOI: 10.1002/jcp.29960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/09/2020] [Indexed: 01/04/2023]
Abstract
Radiotherapy is one of the major modalities for malignancy treatment. High linear energy transfer (LET) charged-particle beams, like proton and carbon ions, exhibit favourable depth-dose distributions and radiobiological enhancement over conventional low-LET photon irradiation, thereby marking a new era in high precision medicine. Tumour cells have developed multicomponent signal transduction networks known as DNA damage responses (DDRs), which initiate cell-cycle checkpoints and induce double-strand break (DSB) repairs in the nucleus by nonhomologous end joining or homologous recombination pathways, to manage ionising radiation (IR)-induced DNA lesions. DNA damage induction and DSB repair pathways are reportedly dependent on the quality of radiation delivered. In this review, we summarise various types of DNA lesion and DSB repair mechanisms, upon irradiation with low and high-LET radiation, respectively. We also analyse factors influencing DNA repair efficiency. Inhibition of DNA damage repair pathways and dysfunctional cell-cycle checkpoint sensitises tumour cells to IR. Radio-sensitising agents, including DNA-PK inhibitors, Rad51 inhibitors, PARP inhibitors, ATM/ATR inhibitors, chk1 inhibitors, wee1 kinase inhibitors, Hsp90 inhibitors, and PI3K/AKT/mTOR inhibitors have been found to enhance cell killing by IR through interference with DDRs, cell-cycle arrest, or other cellular processes. The cotreatment of these inhibitors with IR may represent a promising therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Si
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lu Gan
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Rong Zhou
- Research Center for Ecological Impacts and Environmental Health Effects of Toxic and Hazardous Chemicals, Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment of the People's Republic of China, Nanjing, China
| | - Menghuan Guo
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hong Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Berei J, Eckburg A, Miliavski E, Anderson AD, Miller RJ, Dein J, Giuffre AM, Tang D, Deb S, Racherla KS, Patel M, Vela MS, Puri N. Potential Telomere-Related Pharmacological Targets. Curr Top Med Chem 2020; 20:458-484. [DOI: 10.2174/1568026620666200109114339] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022]
Abstract
Telomeres function as protective caps at the terminal portion of chromosomes, containing
non-coding nucleotide sequence repeats. As part of their protective function, telomeres preserve genomic
integrity and minimize chromosomal exposure, thus limiting DNA damage responses. With
continued mitotic divisions in normal cells, telomeres progressively shorten until they reach a threshold
at a point where they activate senescence or cell death pathways. However, the presence of the enzyme
telomerase can provide functional immortality to the cells that have reached or progressed past
senescence. In senescent cells that amass several oncogenic mutations, cancer formation can occur due
to genomic instability and the induction of telomerase activity. Telomerase has been found to be expressed
in over 85% of human tumors and is labeled as a near-universal marker for cancer. Due to this
feature being present in a majority of tumors but absent in most somatic cells, telomerase and telomeres
have become promising targets for the development of new and effective anticancer therapeutics.
In this review, we evaluate novel anticancer targets in development which aim to alter telomerase
or telomere function. Additionally, we analyze the progress that has been made, including preclinical
studies and clinical trials, with therapeutics directed at telomere-related targets. Furthermore, we review
the potential telomere-related therapeutics that are used in combination therapy with more traditional
cancer treatments. Throughout the review, topics related to medicinal chemistry are discussed,
including drug bioavailability and delivery, chemical structure-activity relationships of select therapies,
and the development of a unique telomere assay to analyze compounds affecting telomere elongation.
Collapse
Affiliation(s)
- Joseph Berei
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Adam Eckburg
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Edward Miliavski
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Austin D. Anderson
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Rachel J. Miller
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Joshua Dein
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Allison M. Giuffre
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Diana Tang
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Shreya Deb
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Kavya Sri Racherla
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Meet Patel
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Monica Saravana Vela
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| | - Neelu Puri
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL 61107, United States
| |
Collapse
|
10
|
Jiang YY, Lin DC, Mayakonda A, Hazawa M, Ding LW, Chien WW, Xu L, Chen Y, Xiao JF, Senapedis W, Baloglu E, Kanojia D, Shang L, Xu X, Yang H, Tyner JW, Wang MR, Koeffler HP. Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut 2017; 66:1358-1368. [PMID: 27196599 PMCID: PMC5912916 DOI: 10.1136/gutjnl-2016-311818] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Oesophageal squamous cell carcinoma (OSCC) is an aggressive malignancy and the major histological subtype of oesophageal cancer. Although recent large-scale genomic analysis has improved the description of the genetic abnormalities of OSCC, few targetable genomic lesions have been identified, and no molecular therapy is available. This study aims to identify druggable candidates in this tumour. DESIGN High-throughput small-molecule inhibitor screening was performed to identify potent anti-OSCC compounds. Whole-transcriptome sequencing (RNA-Seq) and chromatin immunoprecipitation sequencing (ChIP-Seq) were conducted to decipher the mechanisms of action of CDK7 inhibition in OSCC. A variety of in vitro and in vivo cellular assays were performed to determine the effects of candidate genes on OSCC malignant phenotypes. RESULTS The unbiased high-throughput small-molecule inhibitor screening led us to discover a highly potent anti-OSCC compound, THZ1, a specific CDK7 inhibitor. RNA-Seq revealed that low-dose THZ1 treatment caused selective inhibition of a number of oncogenic transcripts. Notably, further characterisation of the genomic features of these THZ1-sensitive transcripts demonstrated that they were frequently associated with super-enhancer (SE). Moreover, SE analysis alone uncovered many OSCC lineage-specific master regulators. Finally, integrative analysis of both THZ1-sensitive and SE-associated transcripts identified a number of novel OSCC oncogenes, including PAK4, RUNX1, DNAJB1, SREBF2 and YAP1, with PAK4 being a potential druggable kinase. CONCLUSIONS Our integrative approaches led to a catalogue of SE-associated master regulators and oncogenic transcripts, which may significantly promote both the understanding of OSCC biology and the development of more innovative therapies.
Collapse
Affiliation(s)
- Yan-Yi Jiang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - De-Chen Lin
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Masaharu Hazawa
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ling-Wen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Wen-Wen Chien
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Ye Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jin-Fen Xiao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - William Senapedis
- Department of Drug Discovery, Karyopharm Therapeutics Inc., Newton, Massachusetts, USA
| | - Erkan Baloglu
- Department of Drug Discovery, Karyopharm Therapeutics Inc., Newton, Massachusetts, USA
| | - Deepika Kanojia
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Shang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Xu
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Ming-Rong Wang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, California, USA
- National University Cancer Institute, National University Health System and National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Li Z, Zhou Y, Tu B, Bu Y, Liu A, Kong J. Long noncoding RNA MALAT1 affects the efficacy of radiotherapy for esophageal squamous cell carcinoma by regulating Cks1 expression. J Oral Pathol Med 2017; 46:583-590. [PMID: 27935117 DOI: 10.1111/jop.12538] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) has been well studied in the progression of many malignancies. However, its association with the radioresistance of tumors has not been well understood yet. This study tried to explore the role of MALAT1 in regulating the radiosensitivity of esophageal cancer (EC), especially esophageal squamous cell carcinoma (ESCC), involving its regulation on Cks1 expression. METHODS KYSE150 cells were subcutaneously inoculated into nude mice to establish ESCC xenografts. Real-time PCR and Western blot analysis were performed to detect the expression of MALAT1 and Cks1 in irradiated xenografts and cells. Functional analysis was performed in both EC9706 and KYSE150 cells via the transfection of corresponding plasmids or small interfering RNAs (siRNAs). Irradiation-induced damage was examined by the detection of cell viability and apoptosis using MTT and TUNEL assays, respectively. RESULTS Both MALAT1 and Cks1 were downregulated in irradiated xenografts and cells. Cks1FER1L4 showed significant downregulation. Overexpression of MALAT1 inhibited irradiation-induced decrease in cell viability, increase in apoptosis, and downregulation of Cks1. Cks1 expression was also downregulated by MALAT1 siRNA, while Cks1 siRNA strongly recovered MALAT1-induced radioresistance in vitro. Moreover, better tumor growth, accompanied by Cks1 upregulation, was observed in KYSE150 xenografts with MALAT1 overexpression, especially under radiation treatment. CONCLUSION MALAT1 acted as one positive regulator of the radioresistance of ESCC, at least partly due to its promotion on Cks1 expression. Furthermore, MALAT1-targeted therapies showed great potential in enhancing the radiotherapeutic effect on ESCC.
Collapse
Affiliation(s)
- Zhijun Li
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Yang Zhou
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Bo Tu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Yu Bu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Aqiu Liu
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| | - Jianmin Kong
- Department of Radiation Oncology, Inner Mongolia Autonomous Region Peoples Hospital, Hohhot, China
| |
Collapse
|
12
|
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: 0.9] [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
|
13
|
Wu J, Liu T, Rios Z, Mei Q, Lin X, Cao S. Heat Shock Proteins and Cancer. Trends Pharmacol Sci 2016; 38:226-256. [PMID: 28012700 DOI: 10.1016/j.tips.2016.11.009] [Citation(s) in RCA: 444] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/23/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Heat shock proteins (HSPs) constitute a large family of proteins involved in protein folding and maturation whose expression is induced by heat shock or other stressors. The major groups are classified based on their molecular weights and include HSP27, HSP40, HSP60, HSP70, HSP90, and large HSPs. HSPs play a significant role in cellular proliferation, differentiation, and carcinogenesis. In this article we comprehensively review the roles of major HSPs in cancer biology and pharmacology. HSPs are thought to play significant roles in the molecular mechanisms leading to cancer development and metastasis. HSPs may also have potential clinical uses as biomarkers for cancer diagnosis, for assessing disease progression, or as therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Jianming Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tuoen Liu
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, WV 24901, USA.
| | - Zechary Rios
- University of Illinois College of Medicine at Chicago, Chicago, IL 60612, USA
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
14
|
Bao CH, Wang XT, Ma W, Wang NN, Un Nesa E, Wang JB, Wang C, Jia YB, Wang K, Tian H, Cheng YF. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun 2015; 458:441-7. [PMID: 25677618 DOI: 10.1016/j.bbrc.2015.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/01/2015] [Indexed: 12/31/2022]
Abstract
Recent evidence suggested that nonirradiated cancer-associated fibroblasts (CAFs) promoted aggressive phenotypes of cancer cells through epithelial-mesenchymal transition (EMT). Hepatoma-derived growth factor (HDGF) is a radiosensitive gene of esophageal squamous cell carcinoma (ESCC). This study aimed to investigate the effect of irradiated fibroblasts on EMT and HDGF expression of ESCC. Our study demonstrated that coculture with nonirradiated fibroblasts significantly increased the invasive ability of ESCC cells and the increased invasiveness was further accelerated when they were cocultured with irradiated fibroblasts. Scattering of ESCC cells was also accelerated by the supernatant from irradiated fibroblasts. Exposure of ESCC cells to supernatant from irradiated fibroblasts resulted in decreased E-cadherin, increased vimentin in vitro and β-catenin was demonstrated to localize to the nucleus in tumor cells with irradiated fibroblasts in vivo models. The expression of HDGF and β-catenin were increased in both fibroblasts and ESCC cells of irradiated group in vitro and in vivo models. Interestingly, the tumor cells adjoining the stromal fibroblasts displayed strong nuclear HDGF immunoreactivity, which suggested the occurrence of a paracrine effect of fibroblasts on HDGF expression. These data suggested that irradiated fibroblasts promoted invasion, growth, EMT and HDGF expression of ESCC.
Collapse
Affiliation(s)
- Ci-Hang Bao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xin-Tong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Wei Ma
- Department of Radiation Oncology, Cancer Hospital, Genaral Hospital of Ningxia Medical University, Yinchuan 750000, China
| | - Na-Na Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Effat Un Nesa
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jian-Bo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yi-Bin Jia
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Kai Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu-Feng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|