51
|
Dong L, Dong Q, Chen Y, Li Y, Zhang B, Zhou F, Lyu X, Chen GG, Lai P, Kung HF, He ML. Novel HDAC5-interacting motifs of Tbx3 are essential for the suppression of E-cadherin expression and for the promotion of metastasis in hepatocellular carcinoma. Signal Transduct Target Ther 2018; 3:22. [PMID: 30151243 PMCID: PMC6107554 DOI: 10.1038/s41392-018-0025-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/22/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Tbx3, a transcriptional repressor, is essential in the organogenesis of vertebrates, stem cell self-renewal and differentiation, and the carcinogenesis of multiple tumor types. However, the mechanism by which Tbx3 participates in the metastasis of hepatocellular carcinoma (HCC) remains largely unknown. In this study, we show that Tbx3 was dramatically upregulated in clinical HCC samples and that elevated expression of Tbx3 promoted cancer progression. To determine the underlying mechanism, systematic glycine scan mutagenesis and deletion assays were performed. We identified two critical motifs, 585LFSYPYT591 and 604HRH606, that contribute to the repression of transcriptional activity. These motifs are also essential for Tbx3 to promote cell migration and metastasis both in vitro and in vivo via the suppression of E-cadherin expression. More importantly, Tbx3 directly interacts with HDAC5 via these motifs, and an HDAC inhibitor blocks Tbx3-mediated cell migration and the downregulation of E-cadherin in HCC. As Tbx3 is involved in the carcinogenesis of multiple types of human cancers, our findings suggest an important target for anti-cancer drug development. A regulatory protein that represses gene activity interacts with an enzyme involved in chromosome remodeling to promote the migration and metastasis of liver cancer cells. Ming-Liang He from the City University of Hong Kong and colleagues found that levels of the T-box transcription factor Tbx3 were dramatically increased in tissue biopsies of liver tumors. They injected Tbx3-expressing human liver cancer cells into mice and saw a positive correlation between Tbx3 activity and cancer progression. By mutating and deleting parts of Tbx3, the researchers identified two particular stretches of the protein that bind histone deacetylase 5, an enzyme involved in ensuring DNA coils, are wound tight to suppress gene activity. This interaction is needed for Tbx3’s tumor-promoting function and may be targetable with drugs in order to prevent metastasis in patients with aggressive liver cancer.
Collapse
Affiliation(s)
- Liang Dong
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Qi Dong
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Ying Chen
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yichen Li
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Bao Zhang
- 2School of Public Health and Tropical Medicine, Southern Medical University, 1023 Shatai Road, 510515 Guangzhou, China
| | - Fanghang Zhou
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xiaoming Lyu
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - George G Chen
- 3Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul Lai
- 3Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hsiang-Fu Kung
- 4Key Laboratory of Tumor Immunopathology, Ministry of Education of China, and Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, 400038 Chongqing, China
| | - Ming-Liang He
- 1Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Biotechnology and Health Center, CityU Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
52
|
Apatorsen plus docetaxel versus docetaxel alone in platinum-resistant metastatic urothelial carcinoma (Borealis-2). Br J Cancer 2018; 118:1434-1441. [PMID: 29765151 PMCID: PMC5988804 DOI: 10.1038/s41416-018-0087-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 03/23/2018] [Accepted: 03/28/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND A randomised study to assess the addition of apatorsen, an antisense oligonucleotide that inhibits Hsp27 expression, to docetaxel in patients with metastatic urothelial carcinoma (mUC) relapsed after prior platinum-based chemotherapy. METHODS Multicentre, phase II study with 1:1 randomisation to apatorsen (three loading doses at 600 mg intravenous followed by weekly doses) plus docetaxel (75 mg/m2 intravenous every 21 days) (A/D) or docetaxel alone. Overall survival (OS) was the primary end point with a P value <0.1 (one-sided) being positive. Progression-free survival (PFS), objective response rate (ORR), safety, and effect of Hsp27 levels on outcomes were secondary end points. RESULTS Patients randomised to A/D (n = 99) had improved OS compared to docetaxel alone (n = 101): HR: 0.80, 80% CI: 0.65-0.98, P = 0.0784, median 6.4 vs 5.9 months. PFS and ORR were similar in both arms. A/D had more incidence of sepsis and urinary tract infections. Patients with baseline Hsp27 levels <5.7 ng/mL had improved OS compared to those with levels ≥5.7 ng/mL. Patients with a decline or ≤20.5% increase in Hsp27 from baseline benefited more from A/D than those with >20.5% increase. CONCLUSIONS A/D met its predefined OS end point in patients with platinum-refractory mUC in this phase II trial. This trial is hypothesis generating requiring further study before informing practice.
Collapse
|
53
|
MacLeod AR, Crooke ST. RNA Therapeutics in Oncology: Advances, Challenges, and Future Directions. J Clin Pharmacol 2018; 57 Suppl 10:S43-S59. [PMID: 28921648 DOI: 10.1002/jcph.957] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/08/2017] [Indexed: 12/18/2022]
Abstract
RNA-based therapeutic technologies represent a rapidly expanding class of therapeutic opportunities with the power to modulate cellular biology in ways never before possible. With RNA-targeted therapeutics, inhibitors of previously undruggable proteins, gene expression modulators, and even therapeutic proteins can be rationally designed based on sequence information alone, something that is not possible with other therapeutic modalities. The most advanced RNA therapeutic modalities are antisense oligonucleotides (ASOs) and small interfering RNAs. Particularly with ASOs, recent clinical data have demonstrated proof of mechanism and clinical benefit with these approaches across several nononcology disease areas by multiple routes of administration. In cancer, next-generation ASOs have recently demonstrated single-agent activity in patients with highly refractory cancers. Here we discuss advances in RNA therapeutics for the treatment of cancer and the challenges that remain to solidify these as mainstay therapeutic modalities to bridge the pharmacogenomic divide that remains in cancer drug discovery.
Collapse
Affiliation(s)
- A Robert MacLeod
- Vice President, Oncology Discovery, Ionis Pharmaceuticals, Carlsbad, CA, USA
| | - Stanley T Crooke
- CEO and Chairman of the Board, Ionis Pharmaceuticals, Carlsbad, CA, USA
| |
Collapse
|
54
|
Abstract
RNA-targeted therapies represent a platform for drug discovery involving chemically modified oligonucleotides, a wide range of cellular RNAs, and a novel target-binding motif, Watson-Crick base pairing. Numerous hurdles considered by many to be impassable have been overcome. Today, four RNA-targeted therapies are approved for commercial use for indications as diverse as Spinal Muscular Atrophy (SMA) and reduction of low-density lipoprotein cholesterol (LDL-C) and by routes of administration including subcutaneous, intravitreal, and intrathecal delivery. The technology is efficient and supports approaching "undruggable" targets. Three additional agents are progressing through registration, and more are in clinical development, representing several chemical and structural classes. Moreover, progress in understanding the molecular mechanisms by which these drugs work has led to steadily better clinical performance and a wide range of mechanisms that may be exploited for therapeutic purposes. Here we summarize the progress, future challenges, and opportunities for this drug discovery platform.
Collapse
Affiliation(s)
- Stanley T Crooke
- Ionis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA.
| | - Joseph L Witztum
- University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - C Frank Bennett
- Ionis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Brenda F Baker
- Ionis Pharmaceuticals, Inc., 2855 Gazelle Court, Carlsbad, CA 92010, USA
| |
Collapse
|
55
|
Heinrich JC, Donakonda S, Haupt VJ, Lennig P, Zhang Y, Schroeder M. New HSP27 inhibitors efficiently suppress drug resistance development in cancer cells. Oncotarget 2018; 7:68156-68169. [PMID: 27626687 PMCID: PMC5356546 DOI: 10.18632/oncotarget.11905] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 08/26/2016] [Indexed: 12/13/2022] Open
Abstract
Drug resistance is an important open problem in cancer treatment. In recent years, the heat shock protein HSP27 (HSPB1) was identified as a key player driving resistance development. HSP27 is overexpressed in many cancer types and influences cellular processes such as apoptosis, DNA repair, recombination, and formation of metastases. As a result cancer cells are able to suppress apoptosis and develop resistance to cytostatic drugs. To identify HSP27 inhibitors we follow a novel computational drug repositioning approach. We exploit a similarity between a predicted HSP27 binding site to a viral thymidine kinase to generate lead inhibitors for HSP27. Six of these leads were verified experimentally. They bind HSP27 and down-regulate its chaperone activity. Most importantly, all six compounds inhibit development of drug resistance in cellular assays. One of the leads – chlorpromazine – is an antipsychotic, which has a positive effect on survival time in human breast cancer. In summary, we make two important contributions: First, we put forward six novel leads, which inhibit HSP27 and tackle drug resistance. Second, we demonstrate the power of computational drug repositioning.
Collapse
Affiliation(s)
- Jörg C Heinrich
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sainitin Donakonda
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - V Joachim Haupt
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| | - Petra Lennig
- B CUBE - Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Yixin Zhang
- B CUBE - Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Michael Schroeder
- Biotechnology Center, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
56
|
Hwang WL, Pleskow HM, Miyamoto DT. Molecular analysis of circulating tumors cells: Biomarkers beyond enumeration. Adv Drug Deliv Rev 2018; 125:122-131. [PMID: 29326053 DOI: 10.1016/j.addr.2018.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/15/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022]
Abstract
Advances in our molecular understanding of cancer biology have paved the way to an expanding compendium of molecularly-targeted therapies, accompanied by the urgent need for biomarkers that enable the precise selection of the most appropriate therapies for individual cancer patients. Circulating biomarkers such as circulating tumor cells (CTCs) are poised to fill this need, since they are "liquid biopsies" that can be performed non-invasively and serially, and may capture the spectrum of spatial and temporal tumor heterogeneity better than conventional tissue biopsies. Increasing evidence suggests that moving beyond the enumeration of CTCs towards more sophisticated molecular analyses can provide actionable data that may predict and potentially improve clinical outcomes. In this review, we discuss the potential of molecular CTC analyses to serve as prognostic and predictive biomarkers to guide cancer therapy and early cancer detection. As technologies to capture and analyze CTCs continue to increase in sophistication, we anticipate that the potential clinical applications of CTCs will grow exponentially in the coming years.
Collapse
Affiliation(s)
- William L Hwang
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital Cancer Center, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Haley M Pleskow
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, United States
| | - David T Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital Cancer Center, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
57
|
Clinical, prognostic, and therapeutic significance of heat shock protein 27 in bladder cancer. Oncotarget 2018; 9:7961-7974. [PMID: 29487706 PMCID: PMC5814273 DOI: 10.18632/oncotarget.24091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/03/2018] [Indexed: 01/26/2023] Open
Abstract
Heat shock protein 27 (HSP27) is highly expressed in many cancers, and its prognostic and predictive value has been reported. HSP27 knockdown using siRNA or OGX-427 (an anti-sense oligonucleotide sequence targeting HSP27) is reported to have anti-cancer effects and to enhance chemosensitivity of cancer cells to chemotherapeutic agents. However, conflicting results have been reported regarding the clinical significance of HSP27 in bladder cancer (BC). Furthermore, long-term suppression of HSP27 has not been investigated in BC. In this study, we investigated the association between HSP27 expression and BC characteristics in 132 BC patient samples, as well as its prognostic value to determine the potential of HSP27 as a clinical biomarker. Additionally, we applied five different shRNAs targeting HSP27 in three invasive BC cell lines to analyze the long-term knockdown effects of HSP27. Our study revealed a significant association between HSP27 expression and adverse pathological characteristics such as high-stage and -grade BC. However, HSP27 expression was not associated with clinical outcomes such as tumor recurrence, progression, and patient survival. Interestingly, although our shRNAs had obvious knockdown effects on HSP27 in BC cells, we did not find consistent effects on apoptosis of BC cells or chemotherapeutic sensitivity of BC cells to cisplatin. Therefore, although HSP27 may be a predictor of adverse pathological characteristics in BC, its role as a prognostic biomarker and therapeutic target seems to be limited.
Collapse
|
58
|
A randomized phase 2 study of a HSP27 targeting antisense, apatorsen with prednisone versus prednisone alone, in patients with metastatic castration resistant prostate cancer. Invest New Drugs 2017; 36:278-287. [PMID: 29250742 DOI: 10.1007/s10637-017-0553-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023]
Abstract
Purpose Heat shock protein 27 (Hsp27) is implicated in prostate cancer progression. Apatorsen is a second generation phosphorothioate antisense inhibitor of Hsp27 expression. We evaluated apatorsen in patients with metastatic castration resistant prostate cancer (mCRPC). Experimental design Eligible patients were randomized 1:1 to receive intravenous apatorsen (3 loading doses of 600 mg within 5-9 days followed by weekly doses of 1000 mg) with oral prednisone 5 mg twice daily or prednisone alone. The primary endpoint was disease progression at 12 weeks. Crossover from prednisone alone was allowed after radiographic progression. Results 74 patients received apatorsen + prednisone (n = 36) or prednisone alone (n = 38). Twenty-five patients crossed-over to receive apatorsen + prednisone. Apatorsen treated patients received a median of 19 infusions. 50% of apatorsen + prednisone patients (95% CI: 32.9%, 67.1%) compared with 42% of prednisone patients (95% CI: 26.3%, 59.2%) did not have disease progression at week 12 (P = 0.33). A PSA decline of ≥50% was observed in 47% of apatorsen + prednisone and 24% of prednisone patients (P = 0.04), with a median duration of response of 24.1 weeks (95% CI: 12.0, 52) and 14.0 weeks (95% CI: 4.0, 44.4), respectively. A PSA decline of ≥50% was observed in 5 patients (20%) that received cross-over apatorsen. Infusion reactions were the most commonly reported adverse event occurring in 77% of apatorsen-treated patients. Conclusions Apatorsen + prednisone did not change the proportion of CRPC patients without disease progression at 12 weeks compared to prednisone but was associated with significant PSA declines. Further evaluation of Hsp27 targeting in prostate cancer is warranted.
Collapse
|
59
|
Ko AH, Murphy PB, Peyton JD, Shipley DL, Al-Hazzouri A, Rodriguez FA, Womack MS, Xiong HQ, Waterhouse DM, Tempero MA, Guo S, Lane CM, Earwood C, DeBusk LM, Bendell JC. A Randomized, Double-Blinded, Phase II Trial of Gemcitabine and Nab-Paclitaxel Plus Apatorsen or Placebo in Patients with Metastatic Pancreatic Cancer: The RAINIER Trial. Oncologist 2017; 22:1427-e129. [PMID: 28935773 PMCID: PMC5728028 DOI: 10.1634/theoncologist.2017-0066] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 07/07/2017] [Indexed: 01/05/2023] Open
Abstract
LESSONS LEARNED The addition of the heat shock protein 27 (Hsp27)-targeting antisense oligonucleotide, apatorsen, to a standard first-line chemotherapy regimen did not result in improved survival in unselected patients with metastatic pancreatic cancer.Findings from this trial hint at the possible prognostic and predictive value of serum Hsp27 that may warrant further investigation. BACKGROUND This randomized, double-blinded, phase II trial evaluated the efficacy of gemcitabine/nab-paclitaxel plus either apatorsen, an antisense oligonucleotide targeting heat shock protein 27 (Hsp27) mRNA, or placebo in patients with metastatic pancreatic cancer. METHODS Patients were randomized 1:1 to Arm A (gemcitabine/nab-paclitaxel plus apatorsen) or Arm B (gemcitabine/nab-paclitaxel plus placebo). Treatment was administered in 28-day cycles, with restaging every 2 cycles, until progression or intolerable toxicity. Serum Hsp27 levels were analyzed at baseline and on treatment. The primary endpoint was overall survival (OS). RESULTS One hundred thirty-two patients were enrolled, 66 per arm. Cytopenias and fatigue were the most frequent grade 3/4 treatment-related adverse events for both arms. Median progression-free survival (PFS) and OS were 2.7 and 5.3 months, respectively, for arm A, and 3.8 and 6.9 months, respectively, for arm B. Objective response rate was 18% for both arms. Patients with high serum level of Hsp27 represented a poor-prognosis subgroup who may have derived modest benefit from addition of apatorsen. CONCLUSION Addition of apatorsen to chemotherapy does not improve outcomes in unselected patients with metastatic pancreatic cancer in the first-line setting, although a trend toward prolonged PFS and OS in patients with high baseline serum Hsp27 suggests this therapy may warrant further evaluation in this subgroup.
Collapse
Affiliation(s)
- Andrew H Ko
- Division of Hematology and Oncology, University of California, San Francisco, California, USA
| | | | - James D Peyton
- Tennessee Oncology, PLLC/SCRI, Nashville, Tennessee, USA
| | | | | | | | - Mark S Womack
- Tennessee Oncology, PLLC/SCRI, Chattanooga, Tennessee, USA
| | - Henry Q Xiong
- The Center for Cancer and Blood Disorders/SCRI, Fort Worth, Texas, USA
| | | | - Margaret A Tempero
- Division of Hematology and Oncology, University of California, San Francisco, California, USA
| | - Shuangli Guo
- Sarah Cannon Research Institute (SCRI), Nashville, Tennessee, USA
| | - Cassie M Lane
- Sarah Cannon Research Institute (SCRI), Nashville, Tennessee, USA
| | - Chris Earwood
- Sarah Cannon Research Institute (SCRI), Nashville, Tennessee, USA
| | - Laura M DeBusk
- Sarah Cannon Research Institute (SCRI), Nashville, Tennessee, USA
| | | |
Collapse
|
60
|
Karam J, Fadous-Khalifé MC, Tannous R, Fakhreddine S, Massoud M, Hadchity J, Aftimos G, Hadchity E. Role of Krüppel-like factor 4 and heat shock protein 27 in cancer of the larynx. Mol Clin Oncol 2017; 7:808-814. [PMID: 29181170 DOI: 10.3892/mco.2017.1412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/07/2017] [Indexed: 12/27/2022] Open
Abstract
Late detection and lack of standard treatment strategies in larynx cancer patients result in high levels of mortality and poor prognosis. Prognostic stratification of larynx cancer patients based on molecular prognostic tumor biomarkers may lead to more efficient clinical management. Krüppel-like factor 4 (KLF4) and Heat Shock Protein 27 (HSP27) have an important role in tumorigenesis and are considered promising candidate biomarkers for various types of cancer. However, their role in larynx carcinoma remains to be elucidated. The present study aimed to determine KLF4 and HSP27 expression profiles in laryngeal tumors. The protein and mRNA expression levels of KLF4 and HSP27 were evaluated by immunohistochemical and reverse transcription-polymerase chain reaction analyses in 44 larynx carcinoma samples and 21 normal tissue samples, and then correlated with clinical characteristics. A differential expression of KLF4 and HSP27 was observed between normal and tumor tissues. The protein and mRNA expression levels of KLF4 were significantly decreased in larynx squamous cell carcinoma (LSCC) compared with normal tissue, whereas HSP27 was significantly overexpressed in tumor tissues compared with normal tissues, at the protein and mRNA levels. KLF4 expression decreased gradually with tumor progression whereas HSP27 expression increased. A significant difference was observed between stages I and IV. KLF4 and HSP27 exhibit opposite functions and roles in the carcinogenic process of LSCC. Their role in laryngeal cancer initiation and progression emphasizes their use as potential future targets for prognosis and treatment. KLF4 and HSP27 expression levels may act as potential biomarkers in patients with cancer of the larynx.
Collapse
Affiliation(s)
- Jihad Karam
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon
| | - Marie Claude Fadous-Khalifé
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon.,Notre Dame de Secours University Hospital, Jbeil 1401, Lebanon
| | - Rita Tannous
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon
| | - Sally Fakhreddine
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon
| | - Marcel Massoud
- Notre Dame de Secours University Hospital, Jbeil 1401, Lebanon
| | - Joseph Hadchity
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon.,Department of Surgery, St. Therese Hospital, Hadat 1003, Lebanon
| | | | - Elie Hadchity
- Anti-Tumor Therapeutic Targeting Laboratory, Faculty of Sciences, Lebanese University, Hadat 1003, Lebanon
| |
Collapse
|
61
|
Vau N, Volavsek M, Blanca A, Montironi R, Raspollini MR, Massari F, Cheng M, Scarpelli M, Lopez-Beltran A. Prospects for precision therapy of bladder urothelial carcinoma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1389273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Nuno Vau
- Urologic Oncology, Champalimaud Clinical Center, Lisbon, Portugal
| | - Metka Volavsek
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Ana Blanca
- Maimonides Biomedical Research Institute of Cordoba, Cordoba, Spain
| | - Rodolfo Montironi
- Institute of Pathological Anatomy and Histopathology, School of Medicine, Polytechnic University of the Marche Region (Ancona), United Hospitals, Ancona, Italy
| | - Maria R. Raspollini
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Florence, Italy
| | | | - Monica Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marina Scarpelli
- Institute of Pathological Anatomy and Histopathology, School of Medicine, Polytechnic University of the Marche Region (Ancona), United Hospitals, Ancona, Italy
| | - Antonio Lopez-Beltran
- Department of Pathology, Unit of Anatomical Pathology, Department of Surgery, Faculty of Medicine, Cordoba, Spain
| |
Collapse
|
62
|
Aunan JR, Cho WC, Søreide K. The Biology of Aging and Cancer: A Brief Overview of Shared and Divergent Molecular Hallmarks. Aging Dis 2017; 8:628-642. [PMID: 28966806 PMCID: PMC5614326 DOI: 10.14336/ad.2017.0103] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022] Open
Abstract
Aging is the inevitable time-dependent decline in physiological organ function and is a major risk factor for cancer development. Due to advances in health care, hygiene control and food availability, life expectancy is increasing and the population in most developed countries is shifting to an increasing proportion of people at a cancer susceptible age. Mechanisms of aging are also found to occur in carcinogenesis, albeit with shared or divergent end-results. It is now clear that aging and cancer development either share or diverge in several disease mechanisms. Such mechanisms include the role of genomic instability, telomere attrition, epigenetic changes, loss of proteostasis, decreased nutrient sensing and altered metabolism, but also cellular senescence and stem cell function. Cancer cells and aged cells are also fundamentally opposite, as cancer cells can be thought of as hyperactive cells with advantageous mutations, rapid cell division and increased energy consumption, while aged cells are hypoactive with accumulated disadvantageous mutations, cell division inability and a decreased ability for energy production and consumption. Nonetheless, aging and cancer are tightly interconnected and many of the same strategies and drugs may be used to target both, while in other cases antagonistic pleiotrophy come into effect and inhibition of one can be the activation of the other. Cancer can be considered an aging disease, though the shared mechanisms underpinning the two processes remain unclear. Better understanding of the shared and divergent pathways of aging and cancer is needed.
Collapse
Affiliation(s)
- Jan R Aunan
- 1Gastrointestinal Translational Research Unit, Molecular Lab, Stavanger University Hospital, Stavanger, Norway.,2Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
| | - William C Cho
- 3Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Kjetil Søreide
- 1Gastrointestinal Translational Research Unit, Molecular Lab, Stavanger University Hospital, Stavanger, Norway.,2Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway.,4Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
63
|
Cattrini C, Zanardi E, Vallome G, Cavo A, Cerbone L, Di Meglio A, Fabbroni C, Latocca MM, Rizzo F, Messina C, Rubagotti A, Barboro P, Boccardo F. Targeting androgen-independent pathways: new chances for patients with prostate cancer? Crit Rev Oncol Hematol 2017; 118:42-53. [PMID: 28917268 DOI: 10.1016/j.critrevonc.2017.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 02/08/2023] Open
Abstract
Androgen deprivation therapy (ADT) is the mainstay treatment for advanced prostate cancer (PC). Most patients eventually progress to a condition known as castration-resistant prostate cancer (CRPC), characterized by lack of response to ADT. Although new androgen receptor signaling (ARS) inhibitors and chemotherapeutic agents have been introduced to overcome resistance to ADT, many patients progress because of primary or acquired resistance to these agents. This comprehensive review aims at exploring the mechanisms of resistance and progression of PC, with specific focus on alterations which lead to the activation of androgen receptor (AR)-independent pathways of survival. Our work integrates available clinical and preclinical data on agents which target these pathways, assessing their potential clinical implication in specific settings of patients. Given the rising interest of the scientific community in cancer immunotherapy strategies, further attention is dedicated to the role of immune evasion in PC.
Collapse
Affiliation(s)
- C Cattrini
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy.
| | - E Zanardi
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - G Vallome
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - A Cavo
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - L Cerbone
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - A Di Meglio
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - C Fabbroni
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - M M Latocca
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - F Rizzo
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - C Messina
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| | - A Rubagotti
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Health Sciences (DISSAL), University of Genoa, Via A. Pastore 1, 16132, Genoa, Italy
| | - P Barboro
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy
| | - F Boccardo
- Academic Unit of Medical Oncology, San Martino University Hospital - IST National Cancer Research Institute, L.go R. Benzi 10, 16132, Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, V.le Benedetto XV 6, 16132, Genoa, Italy
| |
Collapse
|
64
|
Targeting Hsp27/eIF4E interaction with phenazine compound: a promising alternative for castration-resistant prostate cancer treatment. Oncotarget 2017; 8:77317-77329. [PMID: 29100389 PMCID: PMC5652782 DOI: 10.18632/oncotarget.20469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023] Open
Abstract
The actual strategy to improve current therapies in advanced prostate cancer involves targeting genes activated by androgen withdrawal, either to delay or prevent the emergence of the castration-refractory phenotype. However, these genes are often implicated in several physiological processes, and long-term inhibition of survival proteins might be accompanied with cytotoxic effects. To avoid this problem, an alternative therapeutic strategy relies on the identification and use of compounds that disrupt specific protein-protein interactions involved in androgen withdrawal. Specifically, the interaction of the chaperone protein Hsp27 with the initiation factor eIF4E leads to the protection of protein synthesis initiation process and enhances cell survival during cell stress induced by castration or chemotherapy. Thus, in this work we aimed at i) identifying the interaction site of the Hsp27/eIF4E complex and ii) interfere with the relevant protein/protein association mechanism involved in castration-resistant progression of prostate cancer. By a combination of experimental and modeling techniques, we proved that eIF4E interacts with the C-terminal part of Hsp27, preferentially when Hsp27 is phosphorylated. We also observed that the loss of this interaction increased cell chemo-and hormone-sensitivity. In order to find a potential inhibitor of Hsp27/eIF4E interaction, BRET assays in combination with molecular simulations identified the phenazine derivative 14 as the compound able to efficiently interfere with this protein/protein interaction, thereby inhibiting cell viability and increasing cell death in chemo- and castration-resistant prostate cancer models in vitro and in vivo.
Collapse
|
65
|
Huang Z, Yang C, Sun S, Nan Y, Lang Z, Wang X, Zhao J, Liu Y. Heat Shock Protein 27, a Novel Regulator of Transforming Growth Factor β Induced Resistance to Cisplatin in A549 Cell. Pharmacology 2017; 100:283-291. [PMID: 28848138 DOI: 10.1159/000479320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/10/2017] [Indexed: 12/20/2022]
Abstract
Lung cancer is one of the major causes of cancer morbidity and mortality around the world, and the resistance to cisplatin is a critical issue to chemotherapy in lung cancer patients. Transforming growth factor β (TGF-β) signal pathway abnormality is widely observed in drug resistance during lung cancer chemotherapy. Here, we investigated the effects of heat-shock protein 27 (HSP27) in the TGF-β-induced cisplatin resistance in lung cancer cell. In this study, our results indicated that the mRNA and protein expression of HSP27 were significantly increased in human lung cancer tissues. TGF-β induced the mRNA and protein expression of HSP27 in human lung cancer cell (A549). Treatment of TGF-β-induced cisplatin resistance in A549 cell through blocking the cisplatin-induced apoptosis and cell death, which characterized as the increasing of cell viability and decreasing of PARP and caspase3 cleavage in the cisplatin-treated cell. Knockdown of SMAD3 attenuated the TGF-β-induced HSP27 expression and restored the TGF-β-induced cisplatin resistance in A549 cell. Additionally, the knockdown of HSP27 blocked TGF-β-induced cisplatin resistance via decreasing cell viability and increasing cell apoptosis in A549 cell. These data therefore suggested that HSP27 is critical to lung cancer progression and TGF-β-induced cisplatin resistance in human lung cancer cell, and may provide an effective clinical strategy in lung cancer patients with resistance to chemotherapy.
Collapse
Affiliation(s)
- Zhicheng Huang
- Department of Radiology, Jilin Province Cancer Hospital, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Manerba M, Di Ianni L, Govoni M, Roberti M, Recanatini M, Di Stefano G. LDH inhibition impacts on heat shock response and induces senescence of hepatocellular carcinoma cells. Eur J Pharm Sci 2017; 105:91-98. [DOI: 10.1016/j.ejps.2017.05.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 12/15/2022]
|
67
|
Idippily ND, Zheng Q, Gan C, Quamine A, Ashcraft MM, Zhong B, Su B. Copalic acid analogs down-regulate androgen receptor and inhibit small chaperone protein. Bioorg Med Chem Lett 2017; 27:2292-2295. [PMID: 28442254 DOI: 10.1016/j.bmcl.2017.04.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 12/28/2022]
Abstract
Copalic acid, one of the diterpenoid acids in copaiba oil, inhibited the chaperone function of α-crystallin and heat shock protein 27kD (HSP27). It also showed potent activity in decreasing an HSP27 client protein, androgen receptor (AR), which makes it useful in prostate cancer treatment or prevention. To develop potent drug candidates to decrease the AR level in prostate cancer cells, more copalic acid analogs were synthesized. Using the level of AR as the readout, 15 of the copalic acid analogs were screened and two compounds were much more potent than copalic acid. The compounds also dose-dependently inhibited AR positive prostate cancer cell growth. Furthermore, they inhibited the chaperone activity of α-crystallin as well.
Collapse
Affiliation(s)
- Nethrie D Idippily
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Qiaoyun Zheng
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Chunfang Gan
- College of Chemistry and Material Science, Key Laboratory of Beibu Gulf Environment Change and Resources Utilization, Guangxi Teachers Education University, Nanning 530001, China
| | - Aicha Quamine
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Morgan M Ashcraft
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Bo Zhong
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Bin Su
- Department of Chemistry, Center for Gene Regulation in Health and Disease, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Ave., Cleveland, OH 44115, USA.
| |
Collapse
|
68
|
Hendriks LEL, Dingemans AMC. Heat shock protein antagonists in early stage clinical trials for NSCLC. Expert Opin Investig Drugs 2017; 26:541-550. [PMID: 28274158 DOI: 10.1080/13543784.2017.1302428] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Cancer cells have a higher need of chaperones than normal cells to prevent the toxic effects of intracellular protein misfolding and aggregation. Heat shock proteins (Hsps) belong to these chaperones; they are classified into families according to molecular size. Hsps are upregulated in many cancers and inhibition can inhibit tumor growth by destabilizing proteins necessary for tumor survival. In non-small cell lung cancer (NSCLC), there are three different Hsp antagonist classes that are in (early) clinical trials: Hsp90, Hsp70 and Hsp27 inhibitors. Areas covered: The rationale to use Hsp inhibitors in NSCLC will be summarized and phase I-III trials will be reviewed. Expert opinion: Several Hsp90 inhibitors have been tested in phase I-III trials, until now none was positive in unselected NSCLC; therefore development of AUY922, ganetespib and retaspimycin was halted. Results seem more promising in molecularly selected patients, especially in ALK-rearranged NSCLC. Hsp27 is overexpressed in squamous NSCLC and is a mechanism of chemotherapy resistance. The Hsp27 inhibitor apatorsen is now tested in squamous NSCLC. No phase II/III data are known for Hsp70 inhibitors. Combination of Hsp inhibitors with heat shock transcription factor 1 inhibitors or focal adhesion kinase inhibitors might be of interest for future trials.
Collapse
Affiliation(s)
- Lizza E L Hendriks
- a Department of Pulmonary Diseases, GROW - School for oncology and developmental biology , Maastricht University Medical Center+ , Maastricht , The Netherlands
| | - Anne-Marie C Dingemans
- a Department of Pulmonary Diseases, GROW - School for oncology and developmental biology , Maastricht University Medical Center+ , Maastricht , The Netherlands
| |
Collapse
|
69
|
Hwang WL, Hwang KL, Miyamoto DT. The promise of circulating tumor cells for precision cancer therapy. Biomark Med 2016; 10:1269-1285. [PMID: 27924634 PMCID: PMC5827810 DOI: 10.2217/bmm-2016-0192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022] Open
Abstract
The rapidly growing array of therapeutic options in cancer requires informative biomarkers to guide the rational selection and precision application of appropriate therapies. Circulating biomarkers such as circulating tumor cells have immense potential as noninvasive, serial 'liquid biopsies' that may be more representative of the complete spectrum of a patient's individual malignancy than spatially and temporally restricted tumor biopsies. In this review, we discuss the current state-of-the-art in the isolation and molecular characterization of circulating tumor cells as well as their utility in a wide range of clinical applications such as prognostics, treatment monitoring and identification of novel therapeutic targets and resistance mechanisms to enable real-time adjustments in the clinical management of cancer.
Collapse
Affiliation(s)
- William L Hwang
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Katie L Hwang
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
- Medical Scientist Training Program, Harvard Medical School, Boston, MA 02115, USA
| | - David T Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| |
Collapse
|
70
|
Abstract
Recent studies have led to a greater appreciation of the diverse roles RNAs play in maintaining normal cellular function and how they contribute to disease pathology, broadening the number of potential therapeutic targets. Antisense oligonucleotides are the most direct means to target RNA in a selective manner and have become an established platform technology for drug discovery. There are multiple molecular mechanisms by which antisense oligonucleotides can be used to modulate RNAs in cells, including promoting the degradation of the targeted RNA or modulating RNA function without degradation. Antisense drugs utilizing various antisense mechanisms are demonstrating therapeutic potential for the treatment of a broad variety of diseases. This review focuses on some of the advances that have taken place in translating antisense technology from the bench to the clinic.
Collapse
Affiliation(s)
| | | | - Nguyen Pham
- Ionis Pharmaceuticals, Carlsbad, California 92010;
| | - Eric Swayze
- Ionis Pharmaceuticals, Carlsbad, California 92010;
| | | |
Collapse
|