1
|
Tien Anh D, Hai Nam N, Kircher B, Baecker D. The Impact of Fluorination on the Design of Histone Deacetylase Inhibitors. Molecules 2023; 28:molecules28041973. [PMID: 36838960 PMCID: PMC9965134 DOI: 10.3390/molecules28041973] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
In recent years, histone deacetylases (HDACs) have emerged as promising targets in the treatment of cancer. The approach is to inhibit HDACs with drugs known as HDAC inhibitors (HDACis). Such HDACis are broadly classified according to their chemical structure, e.g., hydroxamic acids, benzamides, thiols, short-chain fatty acids, and cyclic peptides. Fluorination plays an important role in the medicinal-chemical design of new active representatives. As a result of the introduction of fluorine into the chemical structure, parameters such as potency or selectivity towards isoforms of HDACs can be increased. However, the impact of fluorination cannot always be clearly deduced. Nevertheless, a change in lipophilicity and, hence, solubility, as well as permeability, can influence the potency. The selectivity towards certain HDACs isoforms can be explained by special interactions of fluorinated compounds with the structure of the slightly different enzymes. Another aspect is that for a more detailed investigation of newly synthesized fluorine-containing active compounds, fluorination is often used for the purpose of labeling. Aside from the isotope 19F, which can be detected by nuclear magnetic resonance spectroscopy, the positron emission tomography of 18F plays a major role. However, to our best knowledge, a survey of the general effects of fluorination on HDACis development is lacking in the literature to date. Therefore, the aim of this review is to highlight the introduction of fluorine in the course of chemical synthesis and the impact on biological activity, using selected examples of recently developed fluorinated HDACis.
Collapse
Affiliation(s)
- Duong Tien Anh
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 10000, Vietnam
| | - Nguyen Hai Nam
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 10000, Vietnam
| | - Brigitte Kircher
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
- Correspondence: (B.K.); (D.B.)
| | - Daniel Baecker
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
- Correspondence: (B.K.); (D.B.)
| |
Collapse
|
2
|
Zhang F, Zhou Q, Yang G, An L, Li F, Wang J. A genetically encoded 19F NMR probe for lysine acetylation. Chem Commun (Camb) 2018; 54:3879-3882. [PMID: 29595201 DOI: 10.1039/c7cc09825a] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Advances in acetylated protein-protein/DNA interactions depend on the development of a novel NMR (nuclear magnetic resonance) probe to study the conformational changes of acetylated proteins. However, the method for detecting the acetylated protein conformation is underdeveloped. Herein, an acetyllysine mimic has been exploited for detecting the conformational changes of acetylated p53-protein/DNA interactions by genetic code expansion and 19F NMR. This 19F NMR probe shows high structural similarity to acetyllysine and could not be deacetylated by sirtuin deacetylase in vitro/vivo. Moreover, acetylation of p53 K164 is reported to be deacetylated by SIRT2 for the first time.
Collapse
Affiliation(s)
- Feng Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Science, Shandong Normal University, No. 88 East Wenhua Road, Jinan, 250014, China.
| | | | | | | | | | | |
Collapse
|
3
|
In Vivo 6-([ 18F]Fluoroacetamido)-1-hexanoicanilide PET Imaging of Altered Histone Deacetylase Activity in Chemotherapy-Induced Neurotoxicity. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:3612027. [PMID: 29755299 PMCID: PMC5884410 DOI: 10.1155/2018/3612027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/19/2018] [Accepted: 01/31/2018] [Indexed: 12/26/2022]
Abstract
Background Histone deacetylases (HDACs) regulate gene expression by changing histone deacetylation status. Neurotoxicity is one of the major side effects of cisplatin, which reacts with deoxyribonucleic acid (DNA) and has excellent antitumor effects. Suberoylanilide hydroxamic acid (SAHA) is an HDAC inhibitor with neuroprotective effects against cisplatin-induced neurotoxicity. Purpose We investigated how cisplatin with and without SAHA pretreatment affects HDAC expression/activity in the brain by using 6-([18F]fluoroacetamido)-1-hexanoicanilide ([18F]FAHA) as a positron emission tomography (PET) imaging agent for HDAC IIa. Materials and Methods [18F]FAHA and [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG) PET studies were done in 24 mice on 2 consecutive days and again 1 week later. The mice were divided into three groups according to drug administration between the first and second imaging sessions (Group A: cisplatin 2 mg/kg, twice; Group B: cisplatin 4 mg/kg, twice; Group C: cisplatin 4 mg/kg, twice, and SAHA 300 mg/kg pretreatment, 4 times). Results The Ki value of [18F]FAHA was increased and the percentage of injected dose/tissue g (% ID/g) of [18F]FDG was decreased in the brains of animals in Groups A and B. The Ki value of [18F]FAHA and % ID/g of [18F]FDG were not significantly different in Group C. Conclusions [18F]FAHA PET clearly showed increased HDAC activity suggestive of cisplatin neurotoxicity in vivo, which was blocked by SAHA pretreatment.
Collapse
|
4
|
Pera B, Krumsiek J, Assouline SE, Marullo R, Patel J, Phillip JM, Román L, Mann KK, Cerchietti L. Metabolomic Profiling Reveals Cellular Reprogramming of B-Cell Lymphoma by a Lysine Deacetylase Inhibitor through the Choline Pathway. EBioMedicine 2018; 28:80-89. [PMID: 29396295 PMCID: PMC5835559 DOI: 10.1016/j.ebiom.2018.01.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/12/2018] [Accepted: 01/15/2018] [Indexed: 01/24/2023] Open
Abstract
Despite the proven clinical antineoplastic activity of histone deacetylase inhibitors (HDACI), their effect has been reported to be lower than expected in B-cell lymphomas. Traditionally considered as “epigenetic drugs”, HDACI modify the acetylation status of an extensive proteome, acting as general lysine deacetylase inhibitors (KDACI), and thus potentially impacting various branches of cellular metabolism. Here, we demonstrate through metabolomic profiling of patient plasma and cell lines that the KDACI panobinostat alters lipid metabolism and downstream survival signaling in diffuse large B-cell lymphomas (DLBCL). Specifically, panobinostat induces metabolic adaptations resulting in newly acquired dependency on the choline pathway and activation of PI3K signaling. This metabolic reprogramming decreased the antineoplastic effect of panobinostat. Conversely, inhibition of these metabolic adaptations resulted in superior anti-lymphoma effect as demonstrated by the combination of panobinostat with a choline pathway inhibitor. In conclusion, our study demonstrates the power of metabolomics in identifying unknown effects of KDACI, and emphasizes the need for a better understanding of these drugs in order to achieve successful clinical implementation. Lysine deacetylase inhibitor (KDACI) treatment alters choline metabolism in B-cell lymphoma patients. KDACI-treated lymphoma cells acquire PI3K pathway dependency via increased choline kinase A (CHKA) activity. Targeting the acquired choline dependency improves the anti-lymphoma effect of KDACI.
Pera et al. explored the effects of the lysine deacetylase inhibitor panobinostat in the metabolism of patients with lymphoma. They demonstrated that panobinostat alters choline metabolism leading to PI3K pathway activation. Their findings revealed the mechanism behind the anti-lymphoma activity of dual lysine deacetylase/PI3K inhibitors, and uncovered a novel therapeutic strategy based on targeting choline pathway following panobinostat treatment.
Collapse
Affiliation(s)
- Benet Pera
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jan Krumsiek
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA; Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sarit E Assouline
- Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Rossella Marullo
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jayeshkumar Patel
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jude M Phillip
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lidia Román
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Koren K Mann
- Segal Cancer Center, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Leandro Cerchietti
- Hematology and Oncology Division, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
5
|
Arlauckas SP, Popov AV, Delikatny EJ. Choline kinase alpha-Putting the ChoK-hold on tumor metabolism. Prog Lipid Res 2016; 63:28-40. [PMID: 27073147 PMCID: PMC5360181 DOI: 10.1016/j.plipres.2016.03.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/14/2016] [Accepted: 03/26/2016] [Indexed: 12/24/2022]
Abstract
It is well established that lipid metabolism is drastically altered during tumor development and response to therapy. Choline kinase alpha (ChoKα) is a key mediator of these changes, as it represents the first committed step in the Kennedy pathway of phosphatidylcholine biosynthesis and ChoKα expression is upregulated in many human cancers. ChoKα activity is associated with drug resistant, metastatic, and malignant phenotypes, and represents a robust biomarker and therapeutic target in cancer. Effective ChoKα inhibitors have been developed and have recently entered clinical trials. ChoKα's clinical relevance was, until recently, attributed solely to its production of second messenger intermediates of phospholipid synthesis. The recent discovery of a non-catalytic scaffolding function of ChoKα may link growth receptor signaling to lipid biogenesis and requires a reinterpretation of the design and validation of ChoKα inhibitors. Advances in positron emission tomography, magnetic resonance spectroscopy, and optical imaging methods now allow for a comprehensive understanding of ChoKα expression and activity in vivo. We will review the current understanding of ChoKα metabolism, its role in tumor biology and the development and validation of targeted therapies and companion diagnostics for this important regulatory enzyme. This comes at a critical time as ChoKα-targeting programs receive more clinical interest.
Collapse
Affiliation(s)
- Sean P Arlauckas
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anatoliy V Popov
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E James Delikatny
- Department of Radiology, 317 Anatomy-Chemistry Building, 3620 Hamilton Walk, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
6
|
Bonomi R, Mukhopadhyay U, Shavrin A, Yeh HH, Majhi A, Dewage SW, Najjar A, Lu X, Cisneros GA, Tong WP, Alauddin MM, Liu RS, Mangner TJ, Turkman N, Gelovani JG. Novel Histone Deacetylase Class IIa Selective Substrate Radiotracers for PET Imaging of Epigenetic Regulation in the Brain. PLoS One 2015; 10:e0133512. [PMID: 26244761 PMCID: PMC4526562 DOI: 10.1371/journal.pone.0133512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 06/29/2015] [Indexed: 01/14/2023] Open
Abstract
Histone deacetylases (HDAC's) became increasingly important targets for therapy of various diseases, resulting in a pressing need to develop HDAC class- and isoform-selective inhibitors. Class IIa deacetylases possess only minimal deacetylase activity against acetylated histones, but have several other client proteins as substrates through which they participate in epigenetic regulation. Herein, we report the radiosyntheses of the second generation of HDAC class IIa-specific radiotracers: 6-(di-fluoroacetamido)-1-hexanoicanilide (DFAHA) and 6-(tri-fluoroacetamido)-1-hexanoicanilide ([18F]-TFAHA). The selectivity of these radiotracer substrates to HDAC class IIa enzymes was assessed in vitro, in a panel of recombinant HDACs, and in vivo using PET/CT imaging in rats. [18F]TFAHA showed significantly higher selectivity for HDAC class IIa enzymes, as compared to [18F]DFAHA and previously reported [18F]FAHA. PET imaging with [18F]TFAHA can be used to visualize and quantify spatial distribution and magnitude of HDAC class IIa expression-activity in different organs and tissues in vivo. Furthermore, PET imaging with [18F]TFAHA may advance the understanding of HDACs class IIa mediated epigenetic regulation of normal and pathophysiological processes, and facilitate the development of novel HDAC class IIa-specific inhibitors for therapy of different diseases.
Collapse
Affiliation(s)
- Robin Bonomi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Uday Mukhopadhyay
- Center for Advanced Biomedical Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Aleksandr Shavrin
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Hsien-Hsien Yeh
- National Cyclotron and Radiochemistry Center, National Yang Ming University, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming University, Taipei, Taiwan
| | - Anjoy Majhi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Sajeewa W. Dewage
- Department of Chemistry, Wayne State University, Detroit, MI 48202, United States of America
| | - Amer Najjar
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Xin Lu
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - G. Andrés Cisneros
- Department of Chemistry, Wayne State University, Detroit, MI 48202, United States of America
| | - William P. Tong
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Mian M. Alauddin
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Ren-Shuan Liu
- National Cyclotron and Radiochemistry Center, National Yang Ming University, Taipei, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming University, Taipei, Taiwan
| | - Thomas J. Mangner
- Positron Emission Tomography Center, Wayne State University, Detroit, MI 48202, United States of America
| | - Nashaat Turkman
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Juri G. Gelovani
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| |
Collapse
|
7
|
Visualizing epigenetics: current advances and advantages in HDAC PET imaging techniques. Neuroscience 2013; 264:186-97. [PMID: 24051365 DOI: 10.1016/j.neuroscience.2013.09.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/27/2013] [Accepted: 09/09/2013] [Indexed: 12/19/2022]
Abstract
Abnormal gene regulation as a consequence of flawed epigenetic mechanisms may be central to the initiation and persistence of many human diseases. However, the association of epigenetic dysfunction with disease and the development of therapeutic agents for treatment are slow. Developing new methodologies used to visualize chromatin-modifying enzymes and their function in the human brain would be valuable for the diagnosis of brain disorders and drug discovery. We provide an overview of current invasive and noninvasive techniques for measuring expression and functions of chromatin-modifying enzymes in the brain, emphasizing tools applicable to histone deacetylase (HDAC) enzymes as a leading example. The majority of current techniques are invasive and difficult to translate to what is happening within a human brain in vivo. However, recent progress in molecular imaging provides new, noninvasive ways to visualize epigenetics in the human brain. Neuroimaging tool development presents a unique set of challenges in order to identify and validate CNS radiotracers for HDACs and other histone-modifying enzymes. We summarize advances in the effort to image HDACs and HDAC inhibitory effects in the brain using positron emission tomography (PET) and highlight generalizable techniques that can be adapted to investigate other specific components of epigenetic machinery. Translational tools like neuroimaging by PET and magnetic resonance imaging provide the best way to link our current understanding of epigenetic changes with in vivo function in normal and diseased brains. These tools will be a critical addition to ex vivo methods to evaluate - and intervene - in CNS dysfunction.
Collapse
|
8
|
Ward CS, Eriksson P, Izquierdo-Garcia JL, Brandes AH, Ronen SM. HDAC inhibition induces increased choline uptake and elevated phosphocholine levels in MCF7 breast cancer cells. PLoS One 2013; 8:e62610. [PMID: 23626839 PMCID: PMC3633900 DOI: 10.1371/journal.pone.0062610] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 03/26/2013] [Indexed: 11/18/2022] Open
Abstract
Histone deacetylase (HDAC) inhibitors have emerged as effective antineoplastic agents in the clinic. Studies from our lab and others have reported that magnetic resonance spectroscopy (MRS)-detectable phosphocholine (PC) is elevated following SAHA treatment, providing a potential noninvasive biomarker of response. Typically, elevated PC is associated with cancer while a decrease in PC accompanies response to antineoplastic treatment. The goal of this study was therefore to elucidate the underlying biochemical mechanism by which HDAC inhibition leads to elevated PC. We investigated the effect of SAHA on MCF-7 breast cancer cells using 13C MRS to monitor [1,2-13C] choline uptake and phosphorylation to PC. We found that PC synthesis was significantly higher in treated cells, representing 154±19% of control. This was within standard deviation of the increase in total PC levels detected by 31P MRS (129±7% of control). Furthermore, cellular choline kinase activity was elevated (177±31%), while cytidylyltransferase activity was unchanged. Expression of the intermediate-affinity choline transporter SLC44A1 and choline kinase α increased (144% and 161%, respectively) relative to control, as determined by mRNA microarray analysis with protein-level confirmation by Western blotting. Taken together, our findings indicate that the increase in PC levels following SAHA treatment results from its elevated synthesis. Additionally, the concentration of glycerophosphocholine (GPC) increased significantly with treatment to 210±45%. This is likely due to the upregulated expression of several phospholipase A2 (PLA2) isoforms, resulting in increased PLA2 activity (162±18%) in SAHA-treated cells. Importantly, the levels of total choline (tCho)-containing metabolites, comprised of choline, PC and GPC, are readily detectable clinically using 1H MRS. Our findings thus provide an important step in validating clinically translatable non-invasive imaging methods for follow-up diagnostics of HDAC inhibitor treatment.
Collapse
Affiliation(s)
- Christopher S. Ward
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, United States of America
| | - Pia Eriksson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, United States of America
| | - Jose L. Izquierdo-Garcia
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, United States of America
| | - Alissa H. Brandes
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, United States of America
| | - Sabrina M. Ronen
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
9
|
Yu JX, Hallac RR, Chiguru S, Mason RP. New frontiers and developing applications in 19F NMR. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2013; 70:25-49. [PMID: 23540575 PMCID: PMC3613763 DOI: 10.1016/j.pnmrs.2012.10.001] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 10/23/2012] [Indexed: 05/06/2023]
Affiliation(s)
- Jian-Xin Yu
- Laboratory of Prognostic Radiology, Division of Advanced Radiological Sciences, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| | - Rami R. Hallac
- Laboratory of Prognostic Radiology, Division of Advanced Radiological Sciences, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| | - Srinivas Chiguru
- Laboratory of Prognostic Radiology, Division of Advanced Radiological Sciences, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| | - Ralph P. Mason
- Laboratory of Prognostic Radiology, Division of Advanced Radiological Sciences, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
10
|
Metabolic biomarkers for response to PI3K inhibition in basal-like breast cancer. Breast Cancer Res 2013; 15:R16. [PMID: 23448424 PMCID: PMC3672699 DOI: 10.1186/bcr3391] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 02/28/2013] [Indexed: 12/18/2022] Open
Abstract
Introduction The phosphatidylinositol 3-kinase (PI3K) pathway is frequently activated in cancer cells through numerous mutations and epigenetic changes. The recent development of inhibitors targeting different components of the PI3K pathway may represent a valuable treatment alternative. However, predicting efficacy of these drugs is challenging, and methods for therapy monitoring are needed. Basal-like breast cancer (BLBC) is an aggressive breast cancer subtype, frequently associated with PI3K pathway activation. The objectives of this study were to quantify the PI3K pathway activity in tissue sections from xenografts representing basal-like and luminal-like breast cancer before and immediately after treatment with PI3K inhibitors, and to identify metabolic biomarkers for treatment response. Methods Tumor-bearing animals (n = 8 per treatment group) received MK-2206 (120 mg/kg/day) or BEZ235 (50 mg/kg/day) for 3 days. Activity in the PI3K/Akt/mammalian target of rapamycin pathway in xenografts and human biopsies was evaluated using a novel method for semiquantitative assessment of Aktser473 phosphorylation. Metabolic changes were assessed by ex vivo high-resolution magic angle spinning magnetic resonance spectroscopy. Results Using a novel dual near-infrared immunofluorescent imaging method, basal-like xenografts had a 4.5-fold higher baseline level of pAktser473 than luminal-like xenografts. Following treatment, basal-like xenografts demonstrated reduced levels of pAktser473 and decreased proliferation. This correlated with metabolic changes, as both MK-2206 and BEZ235 reduced lactate concentration and increased phosphocholine concentration in the basal-like tumors. BEZ235 also caused increased glucose and glycerophosphocholine concentrations. No response to treatment or change in metabolic profile was seen in luminal-like xenografts. Analyzing tumor sections from five patients with BLBC demonstrated that two of these patients had an elevated pAktser473 level. Conclusion The activity of the PI3K pathway can be determined in tissue sections by quantitative imaging using an antibody towards pAktser473. Long-term treatment with MK-2206 or BEZ235 resulted in significant growth inhibition in basal-like, but not luminal-like, xenografts. This indicates that PI3K inhibitors may have selective efficacy in basal-like breast cancer with increased PI3K signaling, and identifies lactate, phosphocholine and glycerophosphocholine as potential metabolic biomarkers for early therapy monitoring. In human biopsies, variable pAktser473 levels were observed, suggesting heterogeneous PI3K signaling activity in BLBC.
Collapse
|
11
|
Yeh HH, Tian M, Hinz R, Young D, Shavrin A, Mukhapadhyay U, Flores LG, Balatoni J, Soghomonyan S, Jeong HJ, Pal A, Uthamanthil R, Jackson JN, Nishii R, Mizuma H, Onoe H, Kagawa S, Higashi T, Fukumitsu N, Alauddin M, Tong W, Herholz K, Gelovani JG. Imaging epigenetic regulation by histone deacetylases in the brain using PET/MRI with ¹⁸F-FAHA. Neuroimage 2012; 64:630-9. [PMID: 22995777 DOI: 10.1016/j.neuroimage.2012.09.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 08/31/2012] [Accepted: 09/05/2012] [Indexed: 01/12/2023] Open
Abstract
Epigenetic modifications mediated by histone deacetylases (HDACs) play important roles in the mechanisms of different neurologic diseases and HDAC inhibitors (HDACIs) have shown promise in therapy. However, pharmacodynamic profiles of many HDACIs in the brain remain largely unknown due to the lack of validated methods for noninvasive imaging of HDAC expression-activity. In this study, dynamic PET/CT imaging was performed in 4 rhesus macaques using [(18)F]FAHA, a novel HDAC substrate, and [(18)F]fluoroacetate, the major radio-metabolite of [(18)F]FAHA, and fused with corresponding MR images of the brain. Quantification of [(18)F]FAHA accumulation in the brain was performed using a customized dual-tracer pharmacokinetic model. Immunohistochemical analyses of brain tissue revealed the heterogeneity of expression of individual HDACs in different brain structures and cell types and confirmed that PET/CT/MRI with [(18)F]FAHA reflects the level of expression-activity of HDAC class IIa enzymes. Furthermore, PET/CT/MRI with [(18)F]FAHA enabled non-invasive, quantitative assessment of pharmacodynamics of HDAC inhibitor SAHA in the brain.
Collapse
Affiliation(s)
- Hsin-Hsien Yeh
- Department of Experimental Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
McIntyre DJO, Madhu B, Lee SH, Griffiths JR. Magnetic resonance spectroscopy of cancer metabolism and response to therapy. Radiat Res 2012; 177:398-435. [PMID: 22401303 DOI: 10.1667/rr2903.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Magnetic resonance spectroscopy allows noninvasive in vivo measurements of biochemical information from living systems, ranging from cultured cells through experimental animals to humans. Studies of biopsies or extracts offer deeper insights by detecting more metabolites and resolving metabolites that cannot be distinguished in vivo. The pharmacokinetics of certain drugs, especially fluorinated drugs, can be directly measured in vivo. This review briefly describes these methods and their applications to cancer metabolism, including glycolysis, hypoxia, bioenergetics, tumor pH, and tumor responses to radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Dominick J O McIntyre
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
| | | | | | | |
Collapse
|
13
|
Beloueche-Babari M, Arunan V, Troy H, te Poele RH, Fong ACWT, Jackson LE, Payne GS, Griffiths JR, Judson IR, Workman P, Leach MO, Chung YL. Histone deacetylase inhibition increases levels of choline kinase α and phosphocholine facilitating noninvasive imaging in human cancers. Cancer Res 2012; 72:990-1000. [PMID: 22194463 PMCID: PMC3378496 DOI: 10.1158/0008-5472.can-11-2688] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are currently approved for cutaneous T-cell lymphoma and are in mid-late stage trials for other cancers. The HDAC inhibitors LAQ824 and SAHA increase phosphocholine (PC) levels in human colon cancer cells and tumor xenografts as observed by magnetic resonance spectroscopy (MRS). In this study, we show that belinostat, an HDAC inhibitor with an alternative chemical scaffold, also caused a rise in cellular PC content that was detectable by (1)H and (31)P MRS in prostate and colon carcinoma cells. In addition, (1)H MRS showed an increase in branched chain amino acid and alanine concentrations. (13)C-choline labeling indicated that the rise in PC resulted from increased de novo synthesis and correlated with an induction of choline kinase α expression. Furthermore, metabolic labeling experiments with (13)C-glucose showed that differential glucose routing favored alanine formation at the expense of lactate production. Additional analysis revealed increases in the choline/water and phosphomonoester (including PC)/total phosphate ratios in vivo. Together, our findings provide mechanistic insights into the impact of HDAC inhibition on cancer cell metabolism and highlight PC as a candidate noninvasive imaging biomarker for monitoring the action of HDAC inhibitors.
Collapse
Affiliation(s)
- Mounia Beloueche-Babari
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Vaitha Arunan
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Helen Troy
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Robert H te Poele
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Anne-Christine Wong Te Fong
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - L Elizabeth Jackson
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Geoffrey S Payne
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - John R Griffiths
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge CB2 ORE, UK
| | - Ian R Judson
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Martin O Leach
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| | - Yuen-Li Chung
- Cancer Research UK and EPSRC Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT, UK
| |
Collapse
|
14
|
Abstract
Abnormal choline metabolism is emerging as a metabolic hallmark that is associated with oncogenesis and tumour progression. Following transformation, the modulation of enzymes that control anabolic and catabolic pathways causes increased levels of choline-containing precursors and breakdown products of membrane phospholipids. These increased levels are associated with proliferation, and recent studies emphasize the complex reciprocal interactions between oncogenic signalling and choline metabolism. Because choline-containing compounds are detected by non-invasive magnetic resonance spectroscopy (MRS), increased levels of these compounds provide a non-invasive biomarker of transformation, staging and response to therapy. Furthermore, enzymes of choline metabolism, such as choline kinase, present novel targets for image-guided cancer therapy.
Collapse
Affiliation(s)
- Kristine Glunde
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, The Russell H. Morgan Department of Radiology and Radiological Science, 720 Rutland Avenue, 212 Traylor Building, Baltimore, Maryland 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland 21231, USA
| | - Zaver M. Bhujwalla
- The Johns Hopkins University In Vivo Cellular and Molecular Imaging Center, The Russell H. Morgan Department of Radiology and Radiological Science, 720 Rutland Avenue, 212 Traylor Building, Baltimore, Maryland 21205, USA
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland 21231, USA
| | - Sabrina M. Ronen
- Department of Radiology, University of California San Francisco School of Medicine, UCSF Mission Bay Campus, Byers Hall, San Francisco, California CA94158-2330, USA
| |
Collapse
|
15
|
Podo F, Canevari S, Canese R, Pisanu ME, Ricci A, Iorio E. MR evaluation of response to targeted treatment in cancer cells. NMR IN BIOMEDICINE 2011; 24:648-672. [PMID: 21387442 DOI: 10.1002/nbm.1658] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 11/22/2010] [Accepted: 11/29/2010] [Indexed: 05/30/2023]
Abstract
The development of molecular technologies, together with progressive sophistication of molecular imaging methods, has allowed the further elucidation of the multiple mutations and dysregulatory effects of pathways leading to oncogenesis. Acting against these pathways by specifically targeted agents represents a major challenge for current research efforts in oncology. As conventional anatomically based pharmacological endpoints may be inadequate to monitor the tumor response to these targeted treatments, the identification and use of more appropriate, noninvasive pharmacodynamic biomarkers appear to be crucial to optimize the design, dosage and schedule of these novel therapeutic approaches. An aberrant choline phospholipid metabolism and enhanced flux of glucose derivatives through glycolysis, which sustain the redirection of mitochondrial ATP to glucose phosphorylation, are two major hallmarks of cancer cells. This review focuses on the changes detected in these pathways by MRS in response to targeted treatments. The progress and limitations of our present understanding of the mechanisms underlying MRS-detected phosphocholine accumulation in cancer cells are discussed in the light of gene and protein expression and the activation of different enzymes involved in phosphatidylcholine biosynthesis and catabolism. Examples of alterations induced in the MRS choline profile of cells exposed to different agents or to tumor environmental factors are presented. Current studies aimed at the identification in cancer cells of MRS-detected pharmacodynamic markers of therapies targeted against specific conditional or constitutive cell receptor stimulation are then reviewed. Finally, the perspectives of present efforts addressed to identify enzymes of the phosphatidylcholine cycle as possible novel targets for anticancer therapy are summarized.
Collapse
Affiliation(s)
- Franca Podo
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | | | |
Collapse
|
16
|
Moestue SA, Engebraaten O, Gribbestad IS. Metabolic effects of signal transduction inhibition in cancer assessed by magnetic resonance spectroscopy. Mol Oncol 2011; 5:224-41. [PMID: 21536506 DOI: 10.1016/j.molonc.2011.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 12/31/2022] Open
Abstract
Despite huge efforts in development of drugs targeting oncogenic signalling, the number of such drugs entering clinical practice to date remains limited. Rational use of biomarkers for drug candidate selection and early monitoring of response to therapy may accelerate this process. Magnetic resonance spectroscopy (MRS) can be used to assess metabolic effects of drug treatment both in vivo and in vitro, and technological advances are continuously increasing the utility of this non-invasive method. In this review, we summarise the use of MRS for monitoring the effect of targeted anticancer drugs, and discuss the potential role of MRS in the context of personalised cancer treatment.
Collapse
Affiliation(s)
- Siver Andreas Moestue
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | |
Collapse
|
17
|
Metabolic assessment of the action of targeted cancer therapeutics using magnetic resonance spectroscopy. Br J Cancer 2009; 102:1-7. [PMID: 19935796 PMCID: PMC2813738 DOI: 10.1038/sj.bjc.6605457] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Developing rational targeted cancer drugs requires the implementation of pharmacodynamic (PD), preferably non-invasive, biomarkers to aid response assessment and patient follow-up. Magnetic resonance spectroscopy (MRS) allows the non-invasive study of tumour metabolism. We describe the MRS-detectable PD biomarkers resulting from the action of targeted therapeutics, and discuss their biological significance and future translation into clinical use.
Collapse
|
18
|
Smith BC, Hallows WC, Denu JM. A continuous microplate assay for sirtuins and nicotinamide-producing enzymes. Anal Biochem 2009; 394:101-9. [PMID: 19615966 DOI: 10.1016/j.ab.2009.07.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 07/07/2009] [Accepted: 07/13/2009] [Indexed: 11/28/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases (sirtuins) and other enzymes that produce nicotinamide are integral to many cellular processes. Yet current activity measurements involve expensive and time-consuming assays. Here we present a spectroscopic assay that circumvents many issues of previous methods. This assay permits continuous product monitoring over time, allows determination of steady-state kinetic parameters, and is readily adaptable to high-throughput screening. The methodology uses an enzyme-coupled system in which nicotinamide is converted to nicotinic acid and ammonia by nicotinamidase. The ammonia is transferred to alpha-ketoglutarate via glutamate dehydrogenase, yielding glutamate and the oxidation of NAD(P)H to NAD(P)+, which is measured spectrophotometrically at 340 nm. Using this continuous assay with sirtuin-1 (Sirt1) and the ADP-ribosyl cyclase CD38, the resulting steady-state kinetic parameters are in excellent agreement with values obtained by other published methods. Importantly, this assay permitted determination of k(cat) and K(m) values with the native acetylated substrate acetyl-CoA synthetase-1; measurement of Sirt1, Sirt2, and Sirt3 activities from mammalian cell extracts; and determination of IC(50) values of various Sirt1 inhibitors. This assay is applicable to any nicotinamide-forming enzyme and will be an important tool to address many outstanding questions surrounding their regulation.
Collapse
Affiliation(s)
- Brian C Smith
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | |
Collapse
|
19
|
Beloueche-Babari M, Peak JC, Jackson LE, Tiet MY, Leach MO, Eccles SA. Changes in choline metabolism as potential biomarkers of phospholipase C{gamma}1 inhibition in human prostate cancer cells. Mol Cancer Ther 2009; 8:1305-11. [PMID: 19417158 DOI: 10.1158/1535-7163.mct-09-0039] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Phosphoinositide-specific phospholipase Cγ1 (PLCγ1) is activated downstream of many receptor tyrosine kinases to promote cell motility. Inhibition of this protein is being explored as a therapeutic strategy for blocking cancer cell invasion and metastasis. The clinical development of such cytostatic therapies requires the implementation of pharmacodynamic biomarkers of target modulation. In this study, we use magnetic resonance spectroscopy to explore metabolic biomarkers of PLCγ1 down-regulation in PC3LN3 prostate cancer cells. We show that inhibition of PLCγ1 via an inducible short hairpin RNA system causes a reduction in phosphocholine levels by up to 50% relative to the control as detected by (1)H and (31)P magnetic resonance spectroscopy analyses. This correlated with a rounded-up morphology and reduced cell migration. Interestingly, the fall in phosphocholine levels was not recorded in cells with constitutive PLCγ1 knockdown where the rounded-up phenotype was no longer apparent. This study reveals alterations in metabolism that accompany the cellular effects of PLCγ1 knockdown and highlights phosphocholine as a potential pharmacodynamic biomarker for monitoring the action of inhibitors targeting PLCγ1 signaling.
Collapse
Affiliation(s)
- Mounia Beloueche-Babari
- Cancer Research UK Clinical Magnetic Resonance Research Group, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sutton, Surrey, United Kingdom.
| | | | | | | | | | | |
Collapse
|
20
|
Taplin ME. Androgen receptor: role and novel therapeutic prospects in prostate cancer. Expert Rev Anticancer Ther 2008; 8:1495-508. [PMID: 18759700 DOI: 10.1586/14737140.8.9.1495] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Androgen receptor (AR) signaling is necessary for the development of prostate cancer. Androgen-deprivation therapy (ADT) for prostate cancer was described over 50 years ago and ADT remains the mainstay of systemic therapy. AR signaling remains intact as the disease evolves to castration-resistant prostate cancer (CRPC). Through cellular adaptations, CRPC continues to rely on androgens and AR growth signaling, and thus AR remains an important therapeutic target. CRPC cells upregulate enzymes used in androgen synthesis, thus providing an intracellular source of androgen despite systemic castration. Compounds in development, such as antiandrogens, lyase inhibitors, heat-shock protein-90 inhibitors, histone deacetylase inhibitors and others, will provide new tools to more effectively reduce ligand, inhibit AR and/or inhibit costimulatory pathways and result in improved clinical outcomes.
Collapse
Affiliation(s)
- Mary-Ellen Taplin
- Harvard Medical School, Dana-Farber Cancer Institute, Lank Center for Genitourinary Oncology, 44 Binney Street, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Ross J, Najjar AM, Sankaranarayanapillai M, Tong WP, Kaluarachchi K, Ronen SM. Fatty acid synthase inhibition results in a magnetic resonance-detectable drop in phosphocholine. Mol Cancer Ther 2008; 7:2556-65. [PMID: 18723500 DOI: 10.1158/1535-7163.mct-08-0015] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Expression of fatty acid synthase (FASN), the key enzyme in de novo synthesis of long-chain fatty acids, is normally low but increases in cancer. Consequently, FASN is a novel target for cancer therapy. However, because FASN inhibitors can lead to tumor stasis rather than shrinkage, noninvasive methods for assessing FASN inhibition are needed. To this end, we combined (1)H, (31)P, and (13)C magnetic resonance spectroscopy (MRS) (a) to monitor the metabolic consequences of FASN inhibition and (b) to identify MRS-detectable metabolic biomarkers of response. Treatment of PC-3 cells with the FASN inhibitor Orlistat for up to 48 h resulted in inhibition of FASN activity by 70%, correlating with 74% inhibition of fatty acid synthesis. Furthermore, we have determined that FASN inhibition results not only in lower phosphatidylcholine levels but also in a 59% drop in the phospholipid precursor phosphocholine (PCho). This drop resulted from inhibition in PCho synthesis as a result of a reduction in the cellular activity of its synthetic enzyme choline kinase. The drop in PCho levels following FASN inhibition was confirmed in SKOV-3 ovarian cancer cells treated with Orlistat and in MCF-7 breast cancer cells treated with Orlistat as well as cerulenin. Combining data from all treated cells, the drop in PCho significantly correlated with the drop in de novo synthesized fatty acid levels, identifying PCho as a potential noninvasive MRS-detectable biomarker of FASN inhibition in vivo.
Collapse
Affiliation(s)
- James Ross
- Department of Radiology, University of California-San Francisco, 1700 4th Street, San Francisco, CA 94158, USA
| | | | | | | | | | | |
Collapse
|
22
|
Smith BC, Hallows WC, Denu JM. Mechanisms and molecular probes of sirtuins. CHEMISTRY & BIOLOGY 2008; 15:1002-13. [PMID: 18940661 PMCID: PMC2626554 DOI: 10.1016/j.chembiol.2008.09.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Revised: 09/08/2008] [Accepted: 09/17/2008] [Indexed: 12/12/2022]
Abstract
Sirtuins are critical regulators of many cellular processes, including insulin secretion, the cell cycle, and apoptosis. Sirtuins are associated with a variety of age-associated diseases such as type II diabetes, obesity, and Alzheimer's disease. A thorough understanding of sirtuin chemical mechanisms will aid toward developing novel therapeutics that regulate metabolic disorders and combat associated diseases. In this review, we discuss the unique deacetylase mechanism of sirtuins and how this information might be employed to develop inhibitors and other molecular probes for therapeutic and basic research applications. We also cover physiological regulation of sirtuin activity and how these modes of regulation may be exploited to manipulate sirtuin activity in live cells. Development of molecular probes and drugs that specifically target sirtuins will further understanding of sirtuin biology and potentially afford new treatments of several human diseases.
Collapse
Affiliation(s)
- Brian C. Smith
- Department of Biomolecular Chemistry; University of Wisconsin, Medical School; Madison, WI 53706; USA
| | - William C. Hallows
- Department of Biomolecular Chemistry; University of Wisconsin, Medical School; Madison, WI 53706; USA
| | - John M. Denu
- Department of Biomolecular Chemistry; University of Wisconsin, Medical School; Madison, WI 53706; USA
| |
Collapse
|
23
|
Chung YL, Troy H, Kristeleit R, Aherne W, Jackson LE, Atadja P, Griffiths JR, Judson IR, Workman P, Leach MO, Beloueche-Babari M. Noninvasive magnetic resonance spectroscopic pharmacodynamic markers of a novel histone deacetylase inhibitor, LAQ824, in human colon carcinoma cells and xenografts. Neoplasia 2008; 10:303-13. [PMID: 18392140 PMCID: PMC2288545 DOI: 10.1593/neo.07834] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2007] [Revised: 01/27/2008] [Accepted: 01/28/2008] [Indexed: 11/18/2022]
Abstract
The aim of this work was to use phosphorus magnetic resonance spectroscopy ((31)P MRS) to investigate the pharmacodynamic effects of LAQ824, a histone deacetylase (HDAC) inhibitor. Human HT29 colon carcinoma cells were examined by (31)P MRS after treatment with LAQ824 and another HDAC inhibitor, suberoylanilide hydroxamic acid. HT29 xenografts and tumor extracts were also examined using (31)P MRS, pre- and post-LAQ824 treatment. Histone H3 acetylation was determined using Western blot analysis, and tumor microvessel density by immunohistochemical staining of CD31. Phosphocholine showed a significant increase in HT29 cells after treatment with LAQ824 and suberoylanilide hydroxamic acid. In vivo, the ratio of phosphomonoester/total phosphorus (TotP) signal was significantly increased in LAQ824-treated HT29 xenografts, and this ratio was inversely correlated with changes in tumor volume. Statistically significant decreases in intracellular pH, beta-nucleoside triphosphate (beta-NTP)/TotP, and beta-NTP/inorganic phosphate (Pi) and an increase in Pi/TotP were also seen in LAQ824-treated tumors. Tumor extracts showed many significant metabolic changes after LAQ824 treatment, in parallel with increased histone acetylation and decreased microvessel density. Treatment with LAQ824 resulted in altered phospholipid metabolism and compromised tumor bioenergetics. The phosphocholine and phosphomonoester increases may have the potential to act as pharmacodynamic markers for noninvasively monitoring tumor response after treatment with LAQ824 or other HDAC inhibitors.
Collapse
Affiliation(s)
- Yuen-Li Chung
- Cancer Research UK, Biomedical Magnetic Resonance Research Group, Department of Basic Medical Sciences, St. George's University of London, London, UK.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Sankaranarayanapillai M, Tong WP, Yuan Q, Bankson JA, Dafni H, Bornmann WG, Soghomonyan S, Pal A, Ramirez MS, Webb D, Kaluarachchi K, Gelovani JG, Ronen SM. Monitoring Histone Deacetylase Inhibition In Vivo: Noninvasive Magnetic Resonance Spectroscopy Method. Mol Imaging 2008. [DOI: 10.2310/7290.2008.0011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Madhuri Sankaranarayanapillai
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - William P. Tong
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Qing Yuan
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - James A. Bankson
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Hagit Dafni
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - William G. Bornmann
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Suren Soghomonyan
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Ashutosh Pal
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Marc S. Ramirez
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Douglas Webb
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Kumaralal Kaluarachchi
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Juri G. Gelovani
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| | - Sabrina M. Ronen
- From Experimental Diagnostic Imaging, Imaging Physics, and Experimental Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX; and Department of Radiology, University of California-San Francisco, San Francisco, CA
| |
Collapse
|
25
|
Chung YL, Griffiths JR. Using metabolomics to monitor anticancer drugs. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2008:55-78. [PMID: 18811053 DOI: 10.1007/2789_2008_089] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The metabolome of a cancer cell is likely to show changes after responding to an anticancer drug. These changes could be used to decide whether to continue treatment or, in the context of a drug trial, to indicate whether the drug is working and perhaps its mechanism of action. (Nuclear) magnetic resonance spectroscopy (NMR/MRS) methods can offer important insights into novel anticancer agents in order to accelerate the drug development process including time-course studies on the effect of a drug on its site of action (termed pharmacodynamics), in this case the cancer. In addition, some classes of anticancer agents currently under development (e.g. antiangiogenics) are designed to be used in combination with other drugs and will not cause tumour shrinkage when used as single agents in Phase 1 clinical trials. Thus NMR/MRS may have a special role in monitoring the pharmacodynamic actions of such drugs in early-phase clinical trials. This review focuses on the use of ex vivo NMR and in vivo MRS methods for monitoring the effect of some novel anticancer drugs on the cancer metabolome. Ex vivo NMR methods are complementary to in vivo measurements, as they can provide additional information and help in the interpretation of the in vivo data.
Collapse
Affiliation(s)
- Y-L Chung
- St. George's University of London, UK
| | | |
Collapse
|
26
|
Ciossek T, Julius H, Wieland H, Maier T, Beckers T. A homogeneous cellular histone deacetylase assay suitable for compound profiling and robotic screening. Anal Biochem 2007; 372:72-81. [PMID: 17868634 DOI: 10.1016/j.ab.2007.07.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 07/16/2007] [Accepted: 07/26/2007] [Indexed: 10/23/2022]
Abstract
Most cellular assays that quantify the efficacy of histone deacetylase (HDAC) inhibitors measure hyperacetylation of core histone proteins H3 and H4. Here we describe a new approach, directly measuring cellular HDAC enzymatic activity using the substrate Boc-K(Ac)-7-amino-4-methylcoumarin (AMC). After penetration into HeLa cervical carcinoma or K562 chronic myeloid leukemia cells, the deacetylated product Boc-K-AMC is formed which, after cell lysis, is cleaved by trypsin, finally releasing the fluorophor AMC. The cellular potency of suberoylanilide hydroxamic acid, LBH589, trichostatin A, and MS275 as well-known HDAC inhibitors was determined using this assay. IC(50) values derived from concentration-effect curves correlated well with EC(50) values derived from a cellomics array scan histone H3 hyperacetylation assay. The cellular HDAC activity assay was adapted to a homogeneous format, fully compatible with robotic screening. Concentration-effect curves generated on a Tecan Genesis Freedom workstation were highly reproducible with a signal-to-noise ratio of 5.7 and a Z' factor of 0.88, indicating a very robust assay. Finally, a HDAC-inhibitor focused library was profiled in a medium-throughput screening campaign. Inhibition of cellular HDAC activity correlated well with cytotoxicity and histone H3 hyperacetylation in HeLa cells and with inhibition of human recombinant HDAC1 in a biochemical assay. Thus, by using Boc-K(Ac)-AMC as a cell-permeable HDAC substrate, the activity of various protein lysine-specific deacetylases including HDAC1-containing complexes is measurable in intact cells in a simple and homogeneous manner.
Collapse
Affiliation(s)
- Thomas Ciossek
- Therapeutic Area Oncology, ALTANA Pharma-a member of the Nycomed Group, Byk-Gulden Str. 2, 78467 Konstanz, Germany
| | | | | | | | | |
Collapse
|
27
|
Piccioni F, Borioni A, Delfini M, Del Giudice MR, Mustazza C, Rodomonte A, Risuleo G. Metabolic alterations in cultured mouse fibroblasts induced by an inhibitor of the tyrosine kinase receptor Fibroblast Growth Factor Receptor 1. Anal Biochem 2007; 367:111-21. [PMID: 17512489 DOI: 10.1016/j.ab.2007.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2007] [Revised: 04/06/2007] [Accepted: 04/09/2007] [Indexed: 11/15/2022]
Abstract
Proton nuclear magnetic resonance (NMR) spectroscopy was used to identify and quantify the metabolites present in cultured mouse fibroblast cells 3T6 in their native state and after treatment with PD166866, an inhibitor of the fibroblast growth factor receptor. Cell extracts were prepared according to the Bligh-Dyer protocol which prevents artifacts deriving from the chemical demolition of macromolecules. Also the growth medium was subjected to the same extraction procedure. The NMR approach made possible the identification and quantification of about 40 different metabolites at nanomoles/mg of protein level: the biological relevance of the variation of some metabolite levels is discussed. Our experimental procedure offers a prospective method for the evaluation of variations of the metabolic profile deriving from different biochemical treatments of these cells.
Collapse
Affiliation(s)
- Fabiana Piccioni
- Department of Chemistry, University of Rome La Sapienza, P.le A. Moro 5, 00185 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Riester D, Hildmann C, Schwienhorst A. Histone deacetylase inhibitors--turning epigenic mechanisms of gene regulation into tools of therapeutic intervention in malignant and other diseases. Appl Microbiol Biotechnol 2007; 75:499-514. [PMID: 17377788 DOI: 10.1007/s00253-007-0912-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 02/26/2007] [Accepted: 02/26/2007] [Indexed: 12/22/2022]
Abstract
Histone deacetylase inhibitors reside among the most promising targeted anticancer agents that are potent inducers of growth arrest, differentiation, and/or apoptotic cell death of transformed cells. In October 2006, the US Food and Drug Administration approved the first drug of this new class, vorinostat (1, Zolinza, Merck). Several histone deacetylase (HDAC) inhibitors more are in clinical trials. HDAC inhibitors have shown significant activity against a variety of hematological and solid tumors at doses that are well tolerated by patients, both in monotherapy as well as in combination therapy with other drugs. This paper reviews the most recent developments in HDAC inhibitor design, particularly in the context of anticancer therapy, and other possible pharmaceutical applications.
Collapse
Affiliation(s)
- Daniel Riester
- Department of Molecular Genetics and Preparative Molecular Biology, Institute for Microbiology und Genetics, Grisebachstr. 8, 37077, Göttingen, Germany
| | | | | |
Collapse
|