1
|
Pan TT, Jiang S, Yuan H, Wang ZY, He MT, Zhang CY. Construction of a chemiluminescent biosensor based on enzymatic extension and click chemistry for sensitive measurement of MGMT activity in human breast tissues. Talanta 2024; 282:127009. [PMID: 39383723 DOI: 10.1016/j.talanta.2024.127009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
O6-methylguanine methyltransferase (MGMT) is responsible for dealkylation of naturally occurring O6-methylguanines, and it is closely related with DNA replication, transcription, and cancers. Herein, we develop a chemiluminescent biosensor based on enzymatic extension and click chemistry for sensitive measurement of MGMT activity. When MGMT is present, the MGMT-catalyzed demethylation reaction initiates the cleavage of biotinylated dumbbell probes by PvuII restrictive enzyme, releasing two DNA fragments with 3'-OH end. The resultant DNA fragments can trigger terminal transferase (TdT)- and click chemistry-assisted isothermal amplification to obtain abundant G-rich sequences. The G-rich sequences can be captured by magnetic beads to produce a high chemiluminescence signal. This biosensor can greatly amplify the chemiluminescence signal, facilitating label-free and template-free measurement of MGMT. Especially, the introduction of dumbbell probe and PvuII enzyme can efficiently eliminate the false positive and improve the assay specificity. This biosensor possesses high sensitivity with a detection limit of 1.4 × 10-9 ng/μL, and it may accurately quantify the intracellular MGMT. Importantly, this biosensor can be used to screen the MGMT inhibitors and distinguish the MGMT level in breast tumor tissues and normal tissues, with great potential in drug discovery and cancer diagnosis.
Collapse
Affiliation(s)
- Ting-Ting Pan
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, China
| | - Su Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, China
| | - Huimin Yuan
- School of Chemistry and Chemical Engineering, State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Zi-Yue Wang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, 250014, China.
| | - Mao-Tao He
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, 261053, China.
| | - Chun-Yang Zhang
- School of Chemistry and Chemical Engineering, State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
2
|
Han Y, Li DL, Han Q, Ma F, Zhang CY. Integration of Demethylation-Activated DNAzyme with a Single Quantum Dot Nanosensor for Sensitive Detection of O 6-Methylguanine DNA Methyltransferase in Breast Tissues. Anal Chem 2024; 96:4487-4494. [PMID: 38451469 DOI: 10.1021/acs.analchem.3c05090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
O6-Methylguanine-DNA-methyltransferase (MGMT) is a demethylation protein that dynamically regulates the O6-methylguanine modification (O6 MeG), and dysregulated MGMT is implicated in various malignant tumors. Herein, we integrate demethylation-activated DNAzyme with a single quantum dot nanosensor to sensitively detect MGMT in breast tissues. The presence of MGMT induces the demethylation of the O6 MeG-caged DNAzyme and the restoration of catalytic activity. The activated DNAzyme then specifically cleaves the ribonucleic acid site of hairpin DNA to expose toehold sequences. The liberated toehold sequence may act as a primer to trigger a cyclic exponential amplification reaction for the generation of enormous signal strands that bind with the Cy5/biotin-labeled probes to form sandwich hybrids. The assembly of sandwich hybrids onto 605QD obtains 605QD-dsDNA-Cy5 nanostructures, inducing efficient FRET between the 605QD donor and Cy5 acceptor. Notably, the introduction of a mismatched base in hairpin DNA can greatly minimize the background and improve the signal-to-noise ratio. This nanosensor achieves a dynamic range of 1.0 × 10-8 to 0.1 ng/μL and a detection limit of 155.78 aM, and it can screen MGMT inhibitors and monitor cellular MGMT activity with single-cell sensitivity. Moreover, it can distinguish the MGMT level in tissues of breast cancer patients and healthy persons, holding great potential in clinical diagnostics and epigenetic research studies.
Collapse
Affiliation(s)
- Yun Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Dong-Ling Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Qian Han
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Fei Ma
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Chun-Yang Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
3
|
Bai P, Fan T, Sun G, Wang X, Zhao L, Zhong R. The dual role of DNA repair protein MGMT in cancer prevention and treatment. DNA Repair (Amst) 2023; 123:103449. [PMID: 36680944 DOI: 10.1016/j.dnarep.2023.103449] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/21/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Alkylating agents are genotoxic chemicals that can induce and treat various types of cancer. This occurs through covalent bonding with cellular macromolecules, in particular DNA, leading to the loss of functional integrity under the persistence of modifications upon replication. O6-alkylguanine (O6-AlkylG) adducts are proposed to be the most potent DNA lesions induced by alkylating agents. If not repaired correctly, these adducts can result, at the molecular level, in DNA point mutations, chromosome aberrations, recombination, crosslinking, and single- and double-strand breaks (SSB/DSBs). At the cellular level, these lesions can result in malignant transformation, senescence, or cell death. O6-methylguanine-DNA methyltransferase (MGMT) is a DNA repair protein capable of removing the alkyl groups from O6-AlkylG adducts in a damage reversal process that can prevent the adverse biological effects of DNA damage caused by guanine O6-alkylation. MGMT can thereby defend normal cells against tumor initiation, however it can also protect tumor cells against the beneficial effects of chemotherapy. Hence, MGMT can play an important role in both the prevention and treatment of cancer; thus, it can be considered as a double-edged sword. From a clinical perspective, MGMT is a therapeutic target, and it is important to explore the rational development of its clinical exploitation.
Collapse
Affiliation(s)
- Peiying Bai
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China; Department of Medical Technology, Beijing Pharmaceutical University of Staff and Workers, Beijing 100079, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China.
| | - Xin Wang
- Department of Clinical Trials Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
4
|
Fillion A, Franco Pinto J, Granzhan A. Harnessing an emissive guanine surrogate to design small-molecule fluorescent chemosensors of O6-methylguanine-DNA-methyltransferase (MGMT). Org Biomol Chem 2022; 20:1888-1892. [PMID: 35174383 DOI: 10.1039/d2ob00208f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The fluorescence properties of an emissive guanine surrogate, thienoguanine (thGN, 2-aminothieno[3,4-d]pyrimidin-4(3H)-one), were exploited to design two real-time chemosensors of O6-methylguanine-DNA-methyltransferase (MGMT), a key DNA repair enzyme involved in the resistance to DNA-alkylating anti-cancer drugs though direct reversal of O6-alkylated guanine adducts.
Collapse
Affiliation(s)
- Alexandra Fillion
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405 Orsay, France.
- CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405 Orsay, France
| | - Jaime Franco Pinto
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405 Orsay, France.
- CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405 Orsay, France
| | - Anton Granzhan
- CNRS UMR9187, Inserm U1196, Institut Curie, PSL Research University, 91405 Orsay, France.
- CNRS UMR9187, Inserm U1196, Université Paris Saclay, 91405 Orsay, France
| |
Collapse
|
5
|
Kurosawa T, Tega Y, Sako D, Mochizuki T, Yamaguchi T, Kawabata K, Inoue K, Ito N, Kusuhara H, Deguchi Y. Transport Characteristics of 6-Mercaptopurine in Brain Microvascular Endothelial Cells Derived From Human Induced Pluripotent Stem Cells. J Pharm Sci 2021; 110:3484-3490. [PMID: 34102205 DOI: 10.1016/j.xphs.2021.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 01/25/2023]
Abstract
The likelihood of reoccurrence of acute lymphoblastic leukemia is influenced by the cerebral concentration of the therapeutic agent 6-mercaptopurine (6-MP) during treatment. Therefore, it is important to understand the blood-brain barrier (BBB) transport mechanism of 6-MP. The purpose of this study was to characterize this mechanism using human induced pluripotent stem cell-derived microvascular endothelial cells (hiPS-BMECs). The permeability coefficient of 6-MP across hiPS-BMECs monolayer in the basal-to-apical direction (B-to-A) was significantly greater than that in the opposite direction (A-to-B). The inhibition profiles of 6-MP transport in the A-to-B direction were different from those in the B-to-A direction. Transport in the A-to-B direction was mainly inhibited by adenine (an inhibitor of equilibrative nucleobase transporter 1; ENBT1), while transport in the B-to-A direction was significantly reduced by inhibitors of multidrug resistance-associated proteins (MRPs), especially zaprinast (an MRP5 inhibitor). Immunocytochemical analyses demonstrated the expression of ENBT1 and MRP5 proteins in hiPS-BMECs. We confirmed that the cellular uptake of 6-MP is decreased by ENBT1 inhibitors in hiPS-BMECs and by knockdown of ENBT1 in hCMEC/D3 cells. These results suggest that ENBT1 and MRP5 make substantial contributions to the transport of 6-MP in hiPS-BMECs and hCMEC/D3 cells.
Collapse
Affiliation(s)
- Toshiki Kurosawa
- Laboratory of Drug Disposition and Pharmacokinetics, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Yuma Tega
- Laboratory of Drug Disposition and Pharmacokinetics, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Daiki Sako
- Laboratory of Drug Disposition and Pharmacokinetics, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Tatsuki Mochizuki
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Naoki Ito
- Department of Pediatrics, School of Medicine, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshiharu Deguchi
- Laboratory of Drug Disposition and Pharmacokinetics, Faculty of Pharma-Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan.
| |
Collapse
|
6
|
Nguyen VT, Tran TTN, Van TK, Tran T. DNA-Templated Silver Nanoclusters Used as a Label-Free Fluorescent Probe for the Detection of O6-Methyltransferase Activity. JOURNAL OF ANALYTICAL CHEMISTRY 2021. [DOI: 10.1134/s1061934821050130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
7
|
Penketh PG, Williamson HS, Baumann RP, Shyam K. Design Strategy for the EPR Tumor-Targeting of 1,2-Bis(sulfonyl)-1-alkylhydrazines. Molecules 2021; 26:molecules26020259. [PMID: 33419160 PMCID: PMC7825511 DOI: 10.3390/molecules26020259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/30/2020] [Accepted: 01/01/2021] [Indexed: 02/04/2023] Open
Abstract
A design strategy for macromolecular prodrugs is described, that are expected to exhibit robust activity against most solid tumor types while resulting in minimal toxicities to normal tissues. This approach exploits the enhanced permeability, and retention (EPR) effect, and utilizes carefully engineered rate constants to selectively target tumor tissue with short-lived cytotoxic moieties. EPR based tumor accumulation (half-life ~ 15 h) is dependent upon the ubiquitous abnormal solid tumor capillary morphology and is expected to be independent of individual tumor cell genetic variability that leads to resistance to molecularly targeted agents. The macromolecular sulfonylhydrazine-based prodrugs hydrolyze spontaneously with long half-life values (~10 h to >300 h dependent upon their structure) resulting in the majority of the 1,2-bis(sulfonyl)-1-alkylhydrazines (BSHs) cytotoxic warhead being released only after tumor sequestration. The very short half-life (seconds) of the finally liberated BSHs localizes the cytotoxic stress to the tumor target site by allowing insufficient time for escape. Thus, short lifespan anticancer species are liberated, and exhibit their activity largely within the tumor target. The abnormal tumor cell membrane pH gradients favor the uptake of BSHs compared to that of normal cells, further enhancing their selectivity. The reliance on physicochemical/chemical kinetic parameters and the EPR effect is expected to reduce response variability, and the acquisition of resistance.
Collapse
Affiliation(s)
- Philip G. Penketh
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
- Correspondence:
| | | | - Raymond P. Baumann
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
| |
Collapse
|
8
|
MGMT promoter methylation in triple negative breast cancer of the GeparSixto trial. PLoS One 2020; 15:e0238021. [PMID: 32841306 PMCID: PMC7446962 DOI: 10.1371/journal.pone.0238021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is typically treated with chemotherapeutic agents, including carboplatin (Cb), an DNA platinating agent. The O6-methylguanine-DNA-methyltransferase gene (MGMT) encodes for the protein O6-alkylguanine-DNA-alkyltransferase (MGMT protein). MGMT protein is involved in DNA repair mechanisms to remove mutagenic and cytotoxic adducts from O6-guanine in DNA. In glioblastoma multiforme, MGMT methylation status is a predictive biomarker for increased response to temozolomide therapy. It has been suggested, that MGMT protein may have relevance for cellular adaptation and could have an influence on resistance to carboplatin therapy. We investigated the influence of MGMT promoter methylation on pathologic complete response and survival of patients with TNBC treated in the neoadjuvant GeparSixto trial. In 174 of 210 available TNBC tumors a valid MGMT promoter methylation status was determined by pyrosequencing of 5 CpG islands. In 21.8%, we detected a mean MGMT promoter methylation >10%. Overall, MGMT promoter methylation was not significantly associated with pathological complete response (pCR) rate. After stratification for the two therapy arms with and without Cb no statistically significant differences in therapy response rates between the two MGMT promoter methylation groups could be observed. Our results show that different MGMT promoter methylation status is not related to different chemotherapy response rates in the TNBC setting in GeparSixto.
Collapse
|
9
|
Le DV, Jiang JH. Fluorescence determination of the activity of O 6-methylguanine-DNA methyltransferase based on the activation of restriction endonuclease and the use of graphene oxide. Mikrochim Acta 2020; 187:300. [PMID: 32347374 DOI: 10.1007/s00604-020-04280-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
A fluorescence method is described for the determination of the activity of O6-methylguanine-DNA methyltransferase (MGMT). It is based on the activation of restriction endonuclease PvuII and the adsorbing a fluorophore-labelled DNA onto the surface of graphene oxide (GO). MGMT activity removes the methyl group from O6-methylguanine (O6MeG) in the fluorophore-labelled DNA to unblock the specific recognition site for further hydrolysis reaction of restriction endonuclease PvuII. The endonuclease catalytic reaction releases fluorophores (5-carboxyfluorescein) from fluorophore-labelled DNA, which can avoid fluorescence quenching by GO, creating an abundance of the fluorescence signal. The fluorescence increase in the assay is thus directly dependent on the MGMT activity. Under the optimal conditions with the emission wavelength of 519 nm (exitation at 494 nm), the activity of the MGMT can be determined in the range 0.5 to 35 ng mL-1 with a detection limit of 0.15 ng mL-1. This is extremely sensitive for the determination of MGMT. The short time of analysis (2 h) is superior to many reported strategies. The method can also be extended for the rapid and sensitive activity assay of other DNA repair enzymes by designing a proper substrate DNA. Conceivably, the technique represents a powerful tool for diagnosis and drug exploitation. Graphical abstract Schematic representation of the fluorescence method for MGMT activity assay.
Collapse
Affiliation(s)
- Dinh-Vu Le
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, 12 Nguyen Van Bao St. Go Vap, Ho Chi Minh, 70000, Viet Nam.
| | - Jian-Hui Jiang
- State Key Laboratory for Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, Hunan, China
| |
Collapse
|
10
|
Penketh P, Williamson H, Shyam K. Physicochemical Considerations of Tumor Selective Drug Delivery and Activity Confinement with Particular Reference to 1,2-Bis(Sulfonyl)-1- Alkylhydrazines Delivery. Curr Drug Deliv 2020; 17:362-374. [PMID: 32342817 PMCID: PMC7499353 DOI: 10.2174/1567201817666200427215044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/21/2019] [Accepted: 03/12/2020] [Indexed: 02/04/2023]
Abstract
Hypoxic tumor cell sub-populations are highly resistant to radiotherapy and their presence frequently causes disease recurrence and death. Here, we described the physicochemical properties required to develop superior tumor-targeted hypoxia-activated modular prodrugs that liberate extremely short-lived bis(sulfonyl)hydrazines (BSHs) as reactive cytotoxins, thereby precisely focusing cytotoxic stress on these radio-resistant hypoxic sub-populations. Therefore, cytotoxic stress will be focused on radiation resistant areas and thus strongly synergizing with radiotherapy.
Collapse
Affiliation(s)
- Philip Penketh
- Address correspondence to this author at the Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA;
Tel: 2032309516; E-mail:
| | | | | |
Collapse
|
11
|
Mattossovich R, Merlo R, Fontana A, d'Ippolito G, Terns MP, Watts EA, Valenti A, Perugino G. A journey down to hell: new thermostable protein-tags for biotechnology at high temperatures. Extremophiles 2019; 24:81-91. [PMID: 31555904 DOI: 10.1007/s00792-019-01134-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/13/2019] [Indexed: 12/14/2022]
Abstract
The specific labelling of proteins in recent years has made use of self-labelling proteins, such as the SNAP-tag® and the Halotag®. These enzymes, by their nature or suitably engineered, have the ability to specifically react with their respective substrates, but covalently retaining a part of them in the catalytic site upon reaction. This led to the synthesis of substrates conjugated with, e.g., fluorophores (proposing them as alternatives to fluorescent proteins), but also with others chemical groups, for numerous biotechnological applications. Recently, a mutant of the OGT from Saccharolobus solfataricus (H5) very stable to high temperatures and in the presence of physical and chemical denaturing agents has been proposed as a thermostable SNAP-tag® for in vivo and in vitro harsh reaction conditions. Here, we show two new thermostable OGTs from Thermotoga neapolitana and Pyrococcus furiosus, which, respectively, display a higher catalytic activity and thermostability respect to H5, proposing them as alternatives for in vivo studies in these extreme model organisms.
Collapse
Affiliation(s)
- Rosanna Mattossovich
- Institute of Biosciences and BioResources, National Council of Research of Italy, Via P. Castellino 111, 80131, Naples, Italy
| | - Rosa Merlo
- Institute of Biosciences and BioResources, National Council of Research of Italy, Via P. Castellino 111, 80131, Naples, Italy
| | - Angelo Fontana
- Institute of Biomolecular Chemistry, National Council of Research of Italy, Via Campi Flegrei, 34, 80078, Pozzuoli, NA, Italy
| | - Giuliana d'Ippolito
- Institute of Biomolecular Chemistry, National Council of Research of Italy, Via Campi Flegrei, 34, 80078, Pozzuoli, NA, Italy
| | - Michael P Terns
- Departments of Biochemistry and Molecular Biology, Genetics, and Microbiology, University of Georgia, Athens, GA, USA
| | - Elizabeth A Watts
- Departments of Biochemistry and Molecular Biology, Genetics, and Microbiology, University of Georgia, Athens, GA, USA
| | - Anna Valenti
- Institute of Biosciences and BioResources, National Council of Research of Italy, Via P. Castellino 111, 80131, Naples, Italy
| | - Giuseppe Perugino
- Institute of Biosciences and BioResources, National Council of Research of Italy, Via P. Castellino 111, 80131, Naples, Italy.
| |
Collapse
|
12
|
Miggiano R, Valenti A, Rossi F, Rizzi M, Perugino G, Ciaramella M. Every OGT Is Illuminated … by Fluorescent and Synchrotron Lights. Int J Mol Sci 2017; 18:ijms18122613. [PMID: 29206193 PMCID: PMC5751216 DOI: 10.3390/ijms18122613] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 12/23/2022] Open
Abstract
O6-DNA-alkyl-guanine-DNA-alkyl-transferases (OGTs) are evolutionarily conserved, unique proteins that repair alkylation lesions in DNA in a single step reaction. Alkylating agents are environmental pollutants as well as by-products of cellular reactions, but are also very effective chemotherapeutic drugs. OGTs are major players in counteracting the effects of such agents, thus their action in turn affects genome integrity, survival of organisms under challenging conditions and response to chemotherapy. Numerous studies on OGTs from eukaryotes, bacteria and archaea have been reported, highlighting amazing features that make OGTs unique proteins in their reaction mechanism as well as post-reaction fate. This review reports recent functional and structural data on two prokaryotic OGTs, from the pathogenic bacterium Mycobacterium tuberculosis and the hyperthermophilic archaeon Sulfolobus solfataricus, respectively. These studies provided insight in the role of OGTs in the biology of these microorganisms, but also important hints useful to understand the general properties of this class of proteins.
Collapse
Affiliation(s)
- Riccardo Miggiano
- DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio 6, 28100 Novara, Italy.
| | - Anna Valenti
- Institute of Biosciences and BioResources, National Research Council of Italy, Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Franca Rossi
- DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio 6, 28100 Novara, Italy.
| | - Menico Rizzi
- DSF-Dipartimento di Scienze del Farmaco, University of Piemonte Orientale, Via Bovio 6, 28100 Novara, Italy.
| | - Giuseppe Perugino
- Institute of Biosciences and BioResources, National Research Council of Italy, Via Pietro Castellino 111, 80131 Naples, Italy.
| | - Maria Ciaramella
- Institute of Biosciences and BioResources, National Research Council of Italy, Via Pietro Castellino 111, 80131 Naples, Italy.
| |
Collapse
|
13
|
Penketh PG, Finch RA, Sauro R, Baumann RP, Ratner ES, Shyam K. pH-dependent general base catalyzed activation rather than isocyanate liberation may explain the superior anticancer efficacy of laromustine compared to related 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine prodrugs. Chem Biol Drug Des 2017. [PMID: 28636806 DOI: 10.1111/cbdd.13057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Laromustine (also known as cloretazine, onrigin, VNP40101M, 101M) is a prodrug of 90CE, a short-lived chloroethylating agent with anticancer activity. The short half-life of 90CE necessitates the use of latentiated prodrug forms for in vivo treatments. Alkylaminocarbonyl-based prodrugs such as laromustine exhibit significantly superior in vivo activity in several murine tumor models compared to analogs utilizing acyl, and alkoxycarbonyl latentiating groups. The alkylaminocarbonyl prodrugs possess two exclusive characteristics: (i) They are primarily unmasked by spontaneous base catalyzed elimination; and (ii) they liberate a reactive carbamoylating species. Previous speculations as to the therapeutic superiority of laromustine have focused upon the inhibition of enzymes by carbamoylation. We have investigated the therapeutic interactions of analogs with segregated chloroethylating and carbamoylating activities (singly and in combination) in the in vivo murine L1210 leukemia model. The combined treatment with chloroethylating and carbamoylating prodrugs failed to result in any synergism and produced a reduction in the therapeutic efficacy compared to the chloroethylating prodrug alone. Evidence supporting an alternative explanation for the superior tumor selectivity of laromustine is presented that is centered upon the high pH sensitivity of its base catalyzed activation, and the more alkaline intracellular pH values commonly found within tumor cells.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Finch
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Rachel Sauro
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Raymond P Baumann
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Elena S Ratner
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Measurement of O 6 -alkylguanine-DNA alkyltransferase activity in tumour cells using stable isotope dilution HPLC-ESIMS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1033-1034:138-146. [PMID: 27544051 DOI: 10.1016/j.jchromb.2016.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/01/2016] [Accepted: 08/06/2016] [Indexed: 11/21/2022]
|
15
|
Lai WY, Tan KT. Environment-sensitive Fluorescent Turn-on Chemical Probe for the Specific Detection of O-Methylguanine-DNA Methyltransferase (MGMT) in Living Cells. J CHIN CHEM SOC-TAIP 2016. [DOI: 10.1002/jccs.201600015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
16
|
Wang C, Abegg D, Hoch DG, Adibekian A. Chemoproteomics-Enabled Discovery of a Potent and Selective Inhibitor of the DNA Repair Protein MGMT. Angew Chem Int Ed Engl 2016; 55:2911-5. [DOI: 10.1002/anie.201511301] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Chao Wang
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Switzerland
| | - Daniel Abegg
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Switzerland
| | - Dominic G. Hoch
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Switzerland
| | - Alexander Adibekian
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Switzerland
| |
Collapse
|
17
|
Wang C, Abegg D, Hoch DG, Adibekian A. Chemoproteomik-vermittelte Entdeckung eines potenten und selektiven Inhibitors des DNA-Reparaturproteins MGMT. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201511301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Chao Wang
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Schweiz
| | - Daniel Abegg
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Schweiz
| | - Dominic G. Hoch
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Schweiz
| | - Alexander Adibekian
- School of Chemistry and Biochemistry; NCCR Chemical Biology; University of Geneva; 30 quai Ernest-Ansermet Geneva Schweiz
| |
Collapse
|
18
|
Yu WT, Wu TW, Huang CL, Chen IC, Tan KT. Protein sensing in living cells by molecular rotor-based fluorescence-switchable chemical probes. Chem Sci 2016; 7:301-307. [PMID: 28758005 PMCID: PMC5515057 DOI: 10.1039/c5sc02808f] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 12/19/2022] Open
Abstract
In this paper, we introduce a general design to construct fluorescence-switching probes by using conjugates of a fluorescent molecular rotor and protein specific ligands for the selective protein detection and real-time tracking of protein degradation in living cells. Upon the interaction of the ligand with the protein ligand-binding domain, the crowded surroundings restrict the bond rotation of the fluorescent molecular rotor to trigger the emission of a strong fluorescence signal, which is reduced upon the addition of a competitive ligand or after protein degradation. With this probe design, two fluorescent probes for MGMT and hCAII proteins were constructed and applied for detecting the endogenous proteins in living cells. In addition, real-time degradation kinetics of the alkylated-MGMT at the single living cell level were revealed for the first time. We believe that this fluorescence-switching probe design can possibly be extended for the analysis of other proteins, for which there are still no effective tools to visualize them in living cells.
Collapse
Affiliation(s)
- Wan-Ting Yu
- Department of Chemistry , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China . ; Tel: +886-3-5715131
| | - Ting-Wei Wu
- Department of Chemistry , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China . ; Tel: +886-3-5715131
| | - Chi-Ling Huang
- Department of Chemistry , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China . ; Tel: +886-3-5715131
| | - I-Chia Chen
- Department of Chemistry , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China . ; Tel: +886-3-5715131
- Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China
| | - Kui-Thong Tan
- Department of Chemistry , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China . ; Tel: +886-3-5715131
- Frontier Research Center on Fundamental and Applied Sciences of Matters , National Tsing Hua University , 101 Sec. 2, Kuang Fu Rd , Hsinchu 30013 , Taiwan , Republic of China
| |
Collapse
|
19
|
Wang CW, Yu WT, Lai HP, Lee BY, Gao RC, Tan KT. Steric-dependent label-free and washing-free enzyme amplified protein detection with dual-functional synthetic probes. Anal Chem 2015; 87:4231-6. [PMID: 25811916 DOI: 10.1021/ac504398g] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Enzyme-catalyzed signal amplification with an antibody-enzyme conjugate is commonly employed in many bioanalytical methods to increase assay sensitivity. However, covalent labeling of the enzyme to the antibody, laborious operating procedures, and extensive washing steps are necessary for protein recognition and signal amplification. Herein, we describe a novel label-free and washing-free enzyme-amplified protein detection method by using dual-functional synthetic molecules to impose steric effects upon protein binding. In our approach, protein recognition and signal amplification are modulated by a simple dual-functional synthetic probe which consists of a protein ligand and an inhibitor. In the absence of the target protein, the inhibitor from the dual-functional probe would inhibit the enzyme activity. In contrast, binding of the target protein to the ligand perturbs this enzyme-inhibitor affinity due to the generation of steric effects caused by the close proximity between the target protein and the enzyme, thereby activating the enzyme to initiate signal amplification. With this strategy, the fluorescence signal can be amplified to as high as 70-fold. The generality and versatility of this strategy are demonstrated by the rapid, selective, and sensitive detection of four different proteins, avidin, O6-methylguanine DNA methyltransferase (MGMT), SNAP-tag, and lactoferrin, with four different probes.
Collapse
Affiliation(s)
- Chia-Wen Wang
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| | - Wan-Ting Yu
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| | - Hsiu-Ping Lai
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| | - Bing-Yuan Lee
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| | - Ruo-Cing Gao
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| | - Kui-Thong Tan
- †Department of Chemistry, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC).,‡Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, 101 Sec. 2, Kuang Fu Rd., Hsinchu 30013, Taiwan (ROC)
| |
Collapse
|
20
|
Shyam K, Penketh PG, Baumann RP, Finch RA, Zhu R, Zhu YL, Sartorelli AC. Antitumor sulfonylhydrazines: design, structure-activity relationships, resistance mechanisms, and strategies for improving therapeutic utility. J Med Chem 2015; 58:3639-71. [PMID: 25612194 DOI: 10.1021/jm501459c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
1,2-Bis(sulfonyl)-1-alkylhydrazines (BSHs) were conceived as more specific DNA guanine O-6 methylating and chloroethylating agents lacking many of the undesirable toxicophores contained in antitumor nitrosoureas. O(6)-Alkylguanine-DNA alkyltransferase (MGMT) is the sole repair protein for O(6)-alkylguanine lesions in DNA and has been reported to be absent in 5-20% of most tumor types. Many BSHs exhibit highly selective cytotoxicity toward cells deficient in MGMT activity. The development of clinically useful MGMT assays should permit the identification of tumors with this vulnerability and allow for the preselection of patient subpopulations with a high probability of responding. The BSH system is highly versatile, permitting the synthesis of many prodrug types with the ability to incorporate an additional level of tumor-targeting due to preferential activation by tumor cells. Furthermore, it may be possible to expand the spectrum of activity of these agents to include tumors with MGMT activity by combining them with tumor-targeted MGMT inhibitors.
Collapse
Affiliation(s)
- Krishnamurthy Shyam
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Philip G Penketh
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Raymond P Baumann
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Rick A Finch
- ‡Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, 650 Cool Water Drive, Bastrop, Texas 78602, United States
| | - Rui Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Yong-Lian Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Alan C Sartorelli
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| |
Collapse
|
21
|
Target-mediated consecutive endonuclease reactions for specific and sensitive homogeneous fluorescence assay of O6-methylguanine-DNA methyltransferase. Anal Chim Acta 2013; 804:252-7. [PMID: 24267090 DOI: 10.1016/j.aca.2013.10.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 10/11/2013] [Accepted: 10/16/2013] [Indexed: 11/23/2022]
Abstract
O(6)-Methylguanine-DNA methyltransferase (MGMT) is one of the most important DNA-repair enzymes. Herein, a simple, sensitive and selective homogeneous fluorescence assay strategy is developed for the detection of MGMT on the basis of target-mediated two consecutive endonuclease reactions. The activity assay of MGMT is firstly accomplished using a hairpin-structured DNA substrate to offer a specific recognition site on the substrate DNA for restriction endonuclease PvuII, and thus to initiate the first endonuclease reaction. The product which activates the second endonuclease reaction allows an efficient amplification approach to create an abundance of fluorescence signal reporters. The first endonuclease reaction offers the method high specificity and the second one furnishes the assay improved sensitivity. The results reveal that the MGMT assay strategy shows dynamic responses in the concentration range from 1 to 120 ng mL(-1) with a detection limit of 0.5 ng mL(-1). By simply altering the alkylated bases, this strategy can also be extended for the detection of other alkyltransferases. Therefore, the developed strategy might provide an intrinsically convenient, sensitive and specific platform for alkyltransferase activate assay and related biochemical studies due to its label-free, homogeneous, and fluorescence-based detection format.
Collapse
|
22
|
Zhu R, Baumann RP, Penketh PG, Shyam K, Sartorelli AC. Hypoxia-selective O6-alkylguanine-DNA alkyltransferase inhibitors: design, synthesis, and evaluation of 6-(benzyloxy)-2-(aryldiazenyl)-9H-purines as prodrugs of O6-benzylguanine. J Med Chem 2013; 56:1355-9. [PMID: 23311288 PMCID: PMC3722860 DOI: 10.1021/jm301804p] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) is a DNA repair protein which removes alkyl groups from the O-6 position of guanine, thereby providing strong resistance to anticancer agents which alkylate this position. The clinical usefulness of these anticancer agents would be substantially augmented if AGT could be selectively inhibited in tumor tissue, without a corresponding depletion in normal tissue. We report the synthesis of a new AGT inhibitor (5c) which selectively depletes AGT in hypoxic tumor cells.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Raymond P. Baumann
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Philip G. Penketh
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Alan C. Sartorelli
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
23
|
Chloroethylating and methylating dual function antineoplastic agents display superior cytotoxicity against repair proficient tumor cells. Bioorg Med Chem Lett 2013; 23:1853-9. [PMID: 23395657 DOI: 10.1016/j.bmcl.2013.01.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 12/20/2012] [Accepted: 01/02/2013] [Indexed: 11/20/2022]
Abstract
Two new agents based upon the structure of the clinically active prodrug laromustine were synthesized. These agents, 2-(2-chloroethyl)-N-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (1) and N-(2-chloroethyl)-2-methyl-1,2-bis(methylsulfonyl)-N-nitrosohydrazinecarboxamide (2), were designed to retain the potent chloroethylating and DNA cross-linking functions of laromustine, and gain the ability to methylate DNA at the O-6 position of guanine, while lacking the carbamoylating activity of laromustine. The methylating arm was introduced with the intent of depleting the DNA repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT). Compound 1 is markedly more cytotoxic than laromustine in both AGT minus EMT6 mouse mammary carcinoma cells and high AGT expressing DU145 human prostate carcinoma cells. DNA cross-linking studies indicated that its cross-linking efficiency is nearly identical to its predicted active decomposition product, 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which is also produced by laromustine. AGT ablation studies in DU145 cells demonstrated that 1 can efficiently deplete AGT. Studies assaying methanol and 2-chloroethanol production as a consequence of the methylation and chloroethylation of water by 1 and 2 confirmed their ability to function as methylating and chloroethylating agents and provided insights into the superior activity of 1.
Collapse
|
24
|
Ishiguro K, Shyam K, Penketh PG, Baumann RP, Sartorelli AC, Rutherford TJ, Ratner ES. Expression of O6-Methylguanine-DNA Methyltransferase Examined by Alkyl-Transfer Assays, Methylation-Specific PCR and Western Blots in Tumors and Matched Normal Tissue. ACTA ACUST UNITED AC 2013; 4:919-931. [PMID: 23946891 DOI: 10.4236/jct.2013.44103] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The tumor selectivity of alkylating agents that produce guanine O6-chloroethyl (laromustine and carmustine) and O6-methyl (temozolomide) lesions, depends upon O6-methylguanine-DNA methyltransferase (MGMT) activity being lower in tumor than in host tissue. Despite the established role of MGMT as a tumor resistance factor, consensus on how to assess MGMT expression in clinical samples is unsettled. The aim of this study is to examine the relationship between the values derived from distinctive MGMT measurements in 13, 12, 6 and 2 pairs of human tumors and matched normal adjacent tissue from the colon, kidney, lung and liver, respectively, and in human cell lines. The MGMT measurements included (a) alkyl-transfer assays using [benzene-3H]O6-benzylguanine as a substrate to assess functional MGMT activity, (b) methylation-specific PCR (MSP) to probe MGMT gene promoter CpG methylations as a measure of gene silencing, and (c) western immunoblots to analyze the MGMT protein. In human cell lines, a strict negative correlation existed between MGMT activity and the extent of promoter methylation. In tissue specimens, by contrast, the correlation between these two variables was low. Moreover, alkyl-transfer assays identified 3 pairs of tumors and normal tissue with tumor-selective reduction in MGMT activity in the absence of promoter methylation. Cell line MGMT migrated as a single band in western analyses, whereas tissue MGMT was heterogeneous around its molecular size and at much higher molecular masses, indicative of multi-layered post-translational modifications. Malignancy is occasionally associated with a mobility shift in MGMT. Contrary to the prevalent expectation that MGMT expression is governed at the level of gene silencing, these data suggest that other mechanisms that can lead to tumor-selective reduction in MGMT activity exist in human tissue.
Collapse
Affiliation(s)
- Kimiko Ishiguro
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Lamb KL, Liu Y, Ishiguro K, Kwon Y, Paquet N, Sartorelli AC, Sung P, Rockwell S, Sweasy JB. Tumor-associated mutations in O⁶ -methylguanine DNA-methyltransferase (MGMT) reduce DNA repair functionality. Mol Carcinog 2012; 53:201-10. [PMID: 23065697 DOI: 10.1002/mc.21964] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 08/27/2012] [Accepted: 08/29/2012] [Indexed: 11/10/2022]
Abstract
MGMT is the primary vehicle for cellular removal of alkyl lesions from the O-6 position of guanine and the O-4 position of thymine. While key to the maintenance of genomic integrity, MGMT also removes damage induced by alkylating chemotherapies, inhibiting the efficacy of cancer treatment. Germline variants of human MGMT are well-characterized, but somatic variants found in tumors were, prior to this work, uncharacterized. We found that MGMT G132R, from a human esophageal tumor, and MGMT G156C, from a human colorectal cancer cell line, are unable to rescue methyltransferase-deficient Escherichia coli as well as wild type (WT) human MGMT after treatment with a methylating agent. Using pre-steady state kinetics, we biochemically characterized these variants as having a reduced rate constant. G132R binds DNA containing an O⁶ -methylguanine lesion half as tightly as WT MGMT, while G156C has a 40-fold decrease in binding affinity for the same damaged DNA versus WT. Mammalian cells expressing either G132R or G156C are more sensitive to methylating agents than mammalian cells expressing WT MGMT. G132R is slightly resistant to O⁶ -benzylguanine, an inhibitor of MGMT in clinical trials, while G156C is almost completely resistant to this inhibitor. The impared functionality of expressed variants G132R and G156C suggests that the presence of somatic variants of MGMT in a tumor could impact chemotherapeutic outcomes.
Collapse
Affiliation(s)
- Kristy L Lamb
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut; Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhu R, Seow HA, Baumann RP, Ishiguro K, Penketh PG, Shyam K, Sartorelli AC. Design of a hypoxia-activated prodrug inhibitor of O6-alkylguanine-DNA alkyltransferase. Bioorg Med Chem Lett 2012; 22:6242-7. [PMID: 22932317 DOI: 10.1016/j.bmcl.2012.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/24/2012] [Accepted: 08/01/2012] [Indexed: 11/15/2022]
Abstract
The efficacy of agents that alkylate the O-6 position of guanine is inhibited by O(6)-alkylguanine-DNA alkyltransferase (AGT) which removes these lesions from the tumor DNA. To increase differential toxicity, inhibitors must selectively deplete AGT in tumors, while sparing normal tissues where this protein serves a protective function. A newly synthesized prodrug of the AGT inhibitor O(6)-benzylguanine (O(6)-BG) with an α,α-dimethyl-4-nitrobenzyloxycarbonyl moiety masking the essential 2-amino group has demonstrated the feasibility of targeting hypoxic regions that are unique to solid tumors, for drug delivery. However, these modifications resulted in greatly decreased solubility. Recently, new potent global AGT inhibitors with improved formulatability such as O(6)-[(3-aminomethyl)benzylguanine (1) have been developed. However, acetylamino (N-(3-(((2-amino-9H-purin-6-yl)oxy)methyl)benzyl)acetamide) (2) exhibits a pronounced decrease in activity. Thus, 1 would be inactivated by N-acetylation and probably N-glucuronidation. To combat potential conjugational inactivation while retaining favorable solubility, we synthesized 6-((3-((dimethylamino)methyl)benzyl)oxy)-9H-purin-2-amine (3) in which the 3-aminomethyl moiety is protected by methylation; and to impart tumor selectivity we synthesized 2-(4-nitrophenyl)propan-2-yl(6-((3-((dimethylamino)methyl)benzyl)oxy)-9H-purin-2-yl)carbamate (7), a hypoxia targeted prodrug of 3 utilizing an α,α-dimethyl-4-nitrobenzyloxycarbonyl moiety. Consistent with this design, 7 demonstrates both hypoxia selective conversion by EMT6 cells of 7 to 3 and hypoxic sensitization of AGT containing DU145 cells to the cytotoxic actions of laromustine, while exhibiting improved solubility.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8066, United States
| | | | | | | | | | | | | |
Collapse
|
27
|
Penketh PG, Shyam K, Baumann RP, Ishiguro K, Patridge EV, Zhu R, Sartorelli AC. A strategy for selective O(6)-alkylguanine-DNA alkyltransferase depletion under hypoxic conditions. Chem Biol Drug Des 2012; 80:279-90. [PMID: 22553921 DOI: 10.1111/j.1747-0285.2012.01401.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cellular resistance to chemotherapeutics that alkylate the O-6 position of guanine residues in DNA correlates with their O(6)-alkylguanine-DNA alkyltransferase activity. In normal cells high [O(6)-alkylguanine-DNA alkyltransferase] is beneficial, sparing the host from toxicity, whereas in tumor cells high [O(6)-alkylguanine-DNA alkyltransferase] prevents chemotherapeutic response. Therefore, it is necessary to selectively inactivate O(6)-alkylguanine-DNA alkyltransferase in tumors. The oxygen-deficient compartment unique to solid tumors is conducive to reduction, and could be utilized to provide this selectivity. Therefore, we synthesized 2-nitro-6-benzyloxypurine, an analog of O(6)-benzylguanine in which the essential 2-amino group is replaced by a nitro moiety, and 2-nitro-6-benzyloxypurine is >2000-fold weaker than O(6)-benzylguanine as an O(6)-alkylguanine-DNA alkyltransferase inhibitor. We demonstrate oxygen concentration sensitive net reduction of 2-nitro-6-benzyloxypurine by cytochrome P450 reductase, xanthine oxidase, and EMT6, DU145, and HL-60 cells to yield O(6)-benzylguanine. We show that 2-nitro-6-benzyloxypurine treatment depletes O(6)-alkylguanine-DNA alkyltransferase in intact cells under oxygen-deficient conditions and selectively sensitizes cells to laromustine (an agent that chloroethylates the O-6 position of guanine) under oxygen-deficient but not normoxic conditions. 2-Nitro-6-benzyloxypurine represents a proof of concept lead compound; however, its facile reduction (E(1/2) - 177 mV versus Ag/AgCl) may result in excessive oxidative stress and/or the generation of O(6)-alkylguanine-DNA alkyltransferase inhibitors in normoxic regions in vivo.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | |
Collapse
|
28
|
Li X, Qian S, Zheng L, Yang B, He Q, Hu Y. A mechanism-based fluorescent probe for labeling O6-methylguanine-DNA methyltransferase in live cells. Org Biomol Chem 2012; 10:3189-91. [DOI: 10.1039/c2ob25231g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Perugino G, Vettone A, Illiano G, Valenti A, Ferrara MC, Rossi M, Ciaramella M. Activity and regulation of archaeal DNA alkyltransferase: conserved protein involved in repair of DNA alkylation damage. J Biol Chem 2011; 287:4222-31. [PMID: 22167184 DOI: 10.1074/jbc.m111.308320] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Agents that form methylation adducts in DNA are highly mutagenic and carcinogenic, and organisms have evolved specialized cellular pathways devoted to their repair, including DNA alkyltransferases. These are proteins conserved in eucarya, bacteria and archaea, acting by a unique reaction mechanism, which leads to direct repair of DNA alkylation damage and irreversible protein alkylation. The alkylated form of DNA alkyltransferases is inactive, and in eukaryotes, it is rapidly directed to degradation. We report here in vitro and in vivo studies on the DNA alkyltransferase from the thermophilic archaeon Sulfolobus solfataricus (SsOGT). The development of a novel, simple, and sensitive fluorescence-based assay allowed a careful characterization of the SsOGT biochemical and DNA binding activities. In addition, transcriptional and post-translational regulation of SsOGT by DNA damage was studied. We show that although the gene transcription is induced by alkylating agent treatment, the protein is degraded in vivo by an alkylation-dependent mechanism. These experiments suggest a striking conservation, from archaea to humans, of this important pathway safeguarding genome stability.
Collapse
Affiliation(s)
- Giuseppe Perugino
- Institute of Protein Biochemistry, Consiglio Nazionale delle Ricerche, Via P. Castellino 111, 80131 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
30
|
KS900: A hypoxia-directed, reductively activated methylating antitumor prodrug that selectively ablates O(6)-alkylguanine-DNA alkyltransferase in neoplastic cells. Biochem Pharmacol 2011; 81:1201-10. [PMID: 21396917 DOI: 10.1016/j.bcp.2011.02.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 11/22/2022]
Abstract
To most effectively treat cancer it may be necessary to preferentially destroy tumor tissue while sparing normal tissues. One strategy to accomplish this is to selectively cripple the involved tumor resistance mechanisms, thereby allowing the affected anticancer drugs to gain therapeutic efficacy. Such an approach is exemplified by our design and synthesis of the intracellular hypoxic cell activated methylating agent, 1,2-bis(methylsulfonyl)-1-methyl-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS900) that targets the O-6 position of guanine in DNA. KS900 is markedly more cytotoxic in clonogenic experiments under conditions of oxygen deficiency than the non-intracellularly activated agents KS90, and 90M, when tested in O(6)-alkylguanine-DNA alkyltransferase (AGT) non-expressing cells (EMT6 mouse mammary carcinoma, CHO/AA8 hamster ovary, and U251 human glioma), and than temozolomide when tested in AGT expressing cells (DU145 human prostate carcinoma). Furthermore, KS900 more efficiently ablates AGT in HL-60 human leukemia and DU145 cells than the spontaneous globally activated methylating agent KS90, with an IC(50) value over 9-fold lower than KS90. Finally, KS900 under oxygen-deficient conditions selectively sensitizes DU145 cells to the chloroethylating agent, onrigin, through the ablation of the resistance protein AGT. Thus, under hypoxia, KS900 is more cytotoxic at substantially lower concentrations than methylating agents such as temozolomide that are not preferentially activated in neoplastic cells by intracellular reductase catalysts. The necessity for intracellular activation of KS900 permits substantially greater cytotoxic activity against cells containing the resistance protein O(6)-alkylguanine-DNA alkyltransferase (AGT) than agents such as temozolomide. Furthermore, the hypoxia-directed intracellular activation of KS900 allows it to preferentially ablate AGT pools under the oxygen-deficient conditions that are present in malignant tissue.
Collapse
|
31
|
Ishiguro K, Zhu YL, Shyam K, Penketh PG, Baumann RP, Sartorelli AC. Quantitative relationship between guanine O(6)-alkyl lesions produced by Onrigin™ and tumor resistance by O(6)-alkylguanine-DNA alkyltransferase. Biochem Pharmacol 2010; 80:1317-25. [PMID: 20654586 DOI: 10.1016/j.bcp.2010.07.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 11/16/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) mediates tumor resistance to alkylating agents that generate guanine O(6)-chloroethyl (Onrigin™ and carmustine) and O(6)-methyl (temozolomide) lesions; however, the relative efficiency of AGT protection against these lesions and the degree of resistance to these agents that a given number of AGT molecules produces are unclear. Measured from differential cytotoxicity in AGT-ablated and AGT-intact HL-60 cells containing 17,000 AGT molecules/cell, AGT produced 12- and 24-fold resistance to chloroethylating (90CE) and methylating (KS90) analogs of Onrigin™, respectively. For 50% growth inhibition, KS90 and 90CE generated 5,600 O(6)-methylguanines/cell and ∼300 O(6)-chloroethylguanines/cell, respectively. AGT repaired O(6)-methylguanines until the AGT pool was exhausted, while its repair of O(6)-chloroethylguanines was incomplete due to progression of the lesions to AGT-irreparable interstrand DNA cross-links. Thus, the smaller number of O(6)-chloroethylguanine lesions needed for cytotoxicity accounted for the marked degree of resistance (12-fold) to 90CE produced by AGT. Transfection of human or murine AGT into AGT deficient transplantable tumor cells (i.e., EMT6, M109 and U251) generated transfectants expressing AGT ranging from 4,000 to 700,000 molecules/cell. In vitro growth inhibition assays using these transfectants treated with 90CE revealed that AGT caused a concentration dependent resistance up to a level of ∼10,000 AGT molecules/cell. This finding was corroborated by in vivo studies where expression of 4,000 and 10,000 murine AGT molecules/cell rendered EMT6 tumors partially and completely resistant to Onrigin™, respectively. These studies imply that the antitumor activity of Onrigin™ stems from guanine O(6)-chloroethylation and define the threshold concentration of AGT that negates its antineoplastic activity.
Collapse
Affiliation(s)
- Kimiko Ishiguro
- Department of Pharmacology, Cancer Center, Yale University School of Medicine, New Haven, CT 06520, United States
| | | | | | | | | | | |
Collapse
|
32
|
Schiller GJ, O'Brien SM, Pigneux A, DeAngelo DJ, Vey N, Kell J, Solomon S, Stuart RK, Karsten V, Cahill AL, Albitar MX, Giles FJ. Single-Agent Laromustine, A Novel Alkylating Agent, Has Significant Activity in Older Patients With Previously Untreated Poor-Risk Acute Myeloid Leukemia. J Clin Oncol 2010; 28:815-21. [DOI: 10.1200/jco.2009.24.2008] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose An international phase II study of laromustine (VNP40101M), a sulfonylhydrazine alkylating agent, was conducted in patients age 60 years or older with previously untreated poor-risk acute myeloid leukemia (AML). Patients and Methods Laromustine 600 mg/m2 was administered as a single 60-minute intravenous infusion. Patients were age 70 years or older or 60 years or older with at least one additional risk factor—unfavorable AML karyotype, Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 2, and/or cardiac, pulmonary, or hepatic comorbidities. Results Eighty-five patients (median age, 72 years; range, 60 to 87 years) were treated. Poor-risk features included age 70 years or older, 78%; adverse karyotype, 47%; PS of 2, 41%; pulmonary disease, 77%; cardiac disease, 73%; and hepatic disease, 3%. Ninety-six percent of patients had at least two risk factors, and 39% had at least four risk factors. The overall response rate (ORR) was 32%, with 20 patients (23%) achieving complete response (CR) and seven (8%) achieving CR with incomplete platelet recovery (CRp). ORR was 20% in patients with adverse cytogenetics; 32% in those age 70 years or older; 32% in those with PS of 2; 32% in patients with baseline pulmonary dysfunction; 34% in patients with baseline cardiac dysfunction; and 27% in 33 patients with at least four risk factors. Twelve (14%) patients died within 30 days of receiving laromustine therapy. Median overall survival was 3.2 months, with a 1-year survival of 21%; the median duration of survival for those who achieved CR/CRp was 12.4 months, with a 1-year survival of 52%. Conclusion Laromustine has significant single-agent activity in elderly patients with poor-risk AML. Adverse events are predominantly myelosuppressive or respiratory. Response rates are consistent across a spectrum of poor-risk features.
Collapse
Affiliation(s)
- Gary J. Schiller
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Susan M. O'Brien
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Arnaud Pigneux
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Daniel J. DeAngelo
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Norbert Vey
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Jonathan Kell
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Scott Solomon
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Robert K. Stuart
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Verena Karsten
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Ann L. Cahill
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Maher X. Albitar
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Francis J. Giles
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| |
Collapse
|
33
|
Baumann RP, Penketh PG, Ishiguro K, Shyam K, Zhu YL, Sartorelli AC. Reductive activation of the prodrug 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119) selectively occurs in oxygen-deficient cells and overcomes O(6)-alkylguanine-DNA alkyltransferase mediated KS119 tumor cell resistance. Biochem Pharmacol 2009; 79:1553-61. [PMID: 20005211 DOI: 10.1016/j.bcp.2009.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 12/01/2009] [Accepted: 12/03/2009] [Indexed: 12/23/2022]
Abstract
1,2-Bis(methylsulfonyl)-1-(2-chloroethyl)-2-[[1-(4-nitrophenyl)ethoxy]carbonyl]hydrazine (KS119) is a prodrug of the 1,2-bis(sulfonyl)hydrazine class of antineoplastic agents designed to exploit the oxygen-deficient regions of cancerous tissue. Thus, under reductive conditions in hypoxic cells this agent decomposes to produce the reactive intermediate 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE), which in turn generates products that alkylate the O(6)-position of guanine in DNA. Comparison of the cytotoxicity of KS119 in cultured cells lacking O(6)-alkylguanine-DNA alkyltransferase (AGT) to an agent such as Onrigin, which through base catalyzed activation produces the same critical DNA G-C cross-link lesions by the generation of 90CE, indicates that KS119 is substantially more potent than Onrigin under conditions of oxygen deficiency, despite being incompletely activated. In cell lines expressing relatively large amounts of AGT, the design of the prodrug KS119, which requires intracellular activation by reductase enzymes to produce a cytotoxic effect, results in an ability to overcome resistance derived from the expression of AGT. This appears to derive from the ability of a small portion of the chloroethylating species produced by the activation of KS119 to slip through the cellular protection afforded by AGT to generate the few DNA G-C cross-links that are required for tumor cell lethality. The findings also demonstrate that activation of KS119 under oxygen-deficient conditions is ubiquitous, occurring in all of the cell lines tested thus far, suggesting that the enzymes required for reductive activation of this agent are widely distributed in many different tumor types.
Collapse
Affiliation(s)
- Raymond P Baumann
- Department of Pharmacology and Cancer Center, Yale University School of Medicine, New Haven, CT 06520, United States
| | | | | | | | | | | |
Collapse
|
34
|
Phase 3 randomized, placebo-controlled, double-blind study of high-dose continuous infusion cytarabine alone or with laromustine (VNP40101M) in patients with acute myeloid leukemia in first relapse. Blood 2009; 114:4027-33. [PMID: 19710500 DOI: 10.1182/blood-2009-06-229351] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Laromustine is a sulfonylhdrazine alkylator with significant antileukemia activity. An international, randomized (2:1), double-blind, placebo-controlled study was conducted to compare complete remission (CR) rates and overall survival (OS) in patients with first relapse acute myeloid leukemia (AML) treated with laromustine and high-dose cytarabine (HDAC) versus HDAC/placebo. Patients received 1.5 g/m(2) per day cytarabine continuous infusion for 3 days and laromustine 600 mg/m(2) (n = 177) or placebo (n = 86) on day 2. Patients in CR received consolidation with laromustine/HDAC or HDAC/placebo as per initial randomization. After interim analysis at 50% enrollment, the Data Safety Monitoring Board (DSMB) expressed concern that any advantage in CR would be compromised by the observed on-study mortality, and enrollment was held. The CR rate was significantly higher for the laromustine/HDAC group (35% vs 19%, P = .005). However, the 30-day mortality rate and median progression-free survival were significantly worse in this group compared with HDAC/placebo (11% vs 2%; P = .016; 54 days vs 34; P = .002). OS and median response durations were similar in both groups. Laromustine/HDAC induced significantly more CR than HDAC/placebo, but OS was not improved due to mortality associated with myelosuppression and its sequelae. The DSMB subsequently approved a revised protocol with laromustine dose reduction and recombinant growth factor support. The study was registered as NCT00112554 at http://www.clinicaltrials.gov.
Collapse
|