1
|
Bacolod MD, Barany F. MGMT Epigenetics: The Influence of Gene Body Methylation and Other Insights Derived from Integrated Methylomic, Transcriptomic, and Chromatin Analyses in Various Cancer Types. Curr Cancer Drug Targets 2021; 21:360-374. [PMID: 33535955 DOI: 10.2174/1568009621666210203111620] [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: 08/14/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND MGMT (O6-methylguanine-DNA methyltransferase) is primarily responsible for limiting the activity of some widely used chemotherapeutic agents, including temozolomide (TMZ) and carmustine (BCNU). The gene encoding this protein is epigenetically regulated, and assessment of methylation at its promoter region is used to predict glioma patients' response to TMZ. METHODS In this report, we employed a bioinformatic approach to elucidate MGMT's epigenetic regulation. Integrated for the analysis were genome-wide methylation and transcription datasets for > 8,600 human tissue (representing 31 distinct cancer types) and 500 human cancer cell line samples. Also crucial to the interpretation of results were publicly available data from the ENCODE Project: tracks for histone modifications (via ChIP-seq) and DNase I hypersensitivity (via DNaseseq), as well as methylation and transcription data for representative cell lines (HeLa-S3, HMEC, K562). RESULTS AND DISCUSSION We were able to validate (perhaps more comprehensively) the contrasting influences of CpG methylation at promoter region and at gene body on MGMT transcription. While the MGMT promoter is populated by CpG sites whose methylation levels displayed high negative correlation (R) with MGMT mRNA counts, the gene body harbors CpG sites exhibiting high positive R values. The promoter CpG sites with very high negative R's across cancer types include cg12981137, cg12434587, and cg00618725. Among the notable gene body CpG sites (high positive R's across cancer types) are cg00198994 (Intron 1), cg04473030 (Intron 2), and cg07367735 (Intron 4). For certain cancer types, such as melanoma, gene body methylation appears to be a better predictor of MGMT transcription (compared to promoter methylation). In general, the CpG methylation v. MGMT expression R values are higher in cell lines relative to tissues. Also, these correlations are noticeably more prominent in certain cancer types such as colorectal, adrenocortical, esophageal, skin, and head and neck cancers, as well as glioblastoma. As expected, hypomethylation at the promoter region is associated with more open chromatin, and enrichment of histone marks H3K4m1, H3K4m2, H3K4m3, and H3K9ac. CONCLUSION Overall, our analysis illustrated the contrasting influence of promoter and gene body methylation on MGMT expression. These observations may help improve diagnostic assays for MGMT.
Collapse
Affiliation(s)
- Manny D Bacolod
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, United States
| | - Francis Barany
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065, United States
| |
Collapse
|
2
|
Povedano E, Vargas E, Montiel VRV, Torrente-Rodríguez RM, Pedrero M, Barderas R, Segundo-Acosta PS, Peláez-García A, Mendiola M, Hardisson D, Campuzano S, Pingarrón JM. Electrochemical affinity biosensors for fast detection of gene-specific methylations with no need for bisulfite and amplification treatments. Sci Rep 2018; 8:6418. [PMID: 29686400 PMCID: PMC5913137 DOI: 10.1038/s41598-018-24902-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
This paper describes two different electrochemical affinity biosensing approaches for the simple, fast and bisulfite and PCR-free quantification of 5-methylated cytosines (5-mC) in DNA using the anti-5-mC antibody as biorecognition element. One of the biosensing approaches used the anti-5-mC as capture bioreceptor and a sandwich type immunoassay, while the other one involved the use of a specific DNA probe and the anti-5-mC as a detector bioreceptor of the captured methylated DNA. Both strategies, named for simplicity in the text as immunosensor and DNA sensor, respectively, were implemented on the surface of magnetic microparticles and the transduction was accomplished by amperometry at screen-printed carbon electrodes by means of the hydrogen peroxide/hydroquinone system. The resulting amperometric biosensors demonstrated reproducibility throughout the entire protocol, sensitive determination with no need for using amplification strategies, and competitiveness with the conventional enzyme-linked immunosorbent assay methodology and the few electrochemical biosensors reported so far in terms of simplicity, sensitivity and assay time. The DNA sensor exhibited higher sensitivity and allowed the detection of the gene-specific methylations conversely to the immunosensor, which detected global DNA methylation. In addition, the DNA sensor demonstrated successful applicability for 1 h-analysis of specific methylation in two relevant tumor suppressor genes in spiked biological fluids and in genomic DNA extracted from human glioblastoma cells.
Collapse
Affiliation(s)
- Eloy Povedano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Eva Vargas
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | | | - Rebeca M Torrente-Rodríguez
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - María Pedrero
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain
| | - Rodrigo Barderas
- Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Pablo San Segundo-Acosta
- Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Alberto Peláez-García
- Department of Pathology, Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz IdiPAZ, Madrid, Spain
| | - Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group and Molecular Pathology Section, INGEMM, Hospital Universitario La Paz IdiPAZ, Madrid, Spain
| | - David Hardisson
- Department of Pathology, Molecular Pathology and Therapeutic Targets Group, Hospital Universitario La Paz IdiPAZ, Madrid, Spain.,Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain
| | - Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain.
| | - José M Pingarrón
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, E-28040, Madrid, Spain.
| |
Collapse
|
3
|
Kotsarenko K, Lylo V, Ruban T, Macewicz L, Lukash L. Effects of Some Growth Factors and Cytokines on the Expression of the Repair Enzyme MGMT and Protein MARP in Human Cells In Vitro : Effect of Some Growth Factors and Cytokines. Biochem Genet 2018; 56:459-477. [PMID: 29589213 DOI: 10.1007/s10528-018-9854-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/21/2018] [Indexed: 11/28/2022]
Abstract
The inducible repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) eliminates O6-methylguanine adducts in DNA and protects the cells from damaging effects of alkylating agents. We have found that anti-MGMT antibodies recognize both the MGMT protein with a mol. weight ~ 24 kDa and a protein with a mol. weight ~ 48 kDa, which was named MARP (anti-methyltransferase antibody recognizable protein). A number of growth factors and cytokines were shown to regulate the expression of MGMT and MARP proteins. The ranges of concentrations of several growth factors and cytokines that caused increasing or decreasing protein amounts in human cell cultures were determined. The results of special biological experiments have allowed us to assume a possible role of MARP in the repair of alkyl adducts in human cells.
Collapse
Affiliation(s)
- Kateryna Kotsarenko
- University of South Bohemia in České Budějovice, Branisovska 1760, 37005, Ceske Budejovice, Czech Republic.
| | - Valentyna Lylo
- Institute of Molecular Biology and Genetics of NAS of Ukraine, Kiev, 03680, Ukraine
| | - Tetiana Ruban
- Institute of Molecular Biology and Genetics of NAS of Ukraine, Kiev, 03680, Ukraine
| | - Larysa Macewicz
- Institute of Molecular Biology and Genetics of NAS of Ukraine, Kiev, 03680, Ukraine
| | - Lyubov Lukash
- Institute of Molecular Biology and Genetics of NAS of Ukraine, Kiev, 03680, Ukraine
| |
Collapse
|
4
|
Chen L, Wang Y, Liu F, Xu L, Peng F, Zhao N, Fu B, Zhu Z, Shi Y, Liu J, Wu R, Wang C, Yao S, Li Y. A systematic review and meta-analysis: Association between MGMT hypermethylation and the clinicopathological characteristics of non-small-cell lung carcinoma. Sci Rep 2018; 8:1439. [PMID: 29362385 PMCID: PMC5780517 DOI: 10.1038/s41598-018-19949-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 01/10/2018] [Indexed: 12/21/2022] Open
Abstract
The relationship between O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation and clinicopathological characteristics of non-small-cell lung carcinoma (NSCLC) has remained controversial and unclear. Therefore, in this study we have undertaken a systematic review and meta-analysis of relevant studies to quantitatively investigate this association. We identified 30 eligible studies investigating 2714 NSCLC patients. The relationship between MGMT hypermethylation and NSCLC was identified based on 20 studies, including 1539 NSCLC patient tissue and 1052 normal and adjacent tissue samples (OR = 4.60, 95% CI = 3.46~6.11, p < 0.00001). MGMT methylation varied with ethnicity (caucasian: OR = 4.56, 95% CI = 2.63~7.92, p < 0.00001; asian: OR = 5.18, 95% CI = 2.03~13.22, p = 0.0006) and control style (autologous: OR = 4.44, 95% CI = 3.32~5.92, p < 0.00001; heterogeneous: OR = 9.05, 95% CI = 1.79~45.71, p = 0.008). In addition, MGMT methylation was observed to be specifically associated with NSCLC clinical stage, and not with age, sex, smoking, pathological types, and differentiation status. Also MGMT methylation did not impact NSCLC patients survival (HR = 1.32, 95% CI = 0.77~2.28, p = 0.31). Our study provided clear evidence about the association of MGMT hypermethylation with increased risk of NSCLC.
Collapse
Affiliation(s)
- Lin Chen
- Department of Internal Neurology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Yong Wang
- Department of Medical Oncology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Fen Liu
- Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Liyao Xu
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Feifei Peng
- Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Ning Zhao
- Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Biqi Fu
- Department of Rheumatology, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Zijie Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Yu Shi
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China
| | - Jiansheng Liu
- Department of Medical Oncology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Renrui Wu
- Department of Internal Neurology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Chen Wang
- Department of Internal Neurology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Shengmin Yao
- Department of Internal Neurology, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou People's Hospital), Ganzhou, Jiangxi, 341000, China
| | - Yong Li
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchan, Jiangxi, 330000, China.
| |
Collapse
|
5
|
Zhou X, Zhao M, Duan X, Guo B, Cheng W, Ding S, Ju H. Collapse of DNA Tetrahedron Nanostructure for "Off-On" Fluorescence Detection of DNA Methyltransferase Activity. ACS APPLIED MATERIALS & INTERFACES 2017; 9:40087-40093. [PMID: 29111659 DOI: 10.1021/acsami.7b13551] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
As a potential detection technique, highly rigid and versatile functionality of DNA tetrahedron nanostructures is often used in biosensing systems. In this work, a novel multifunctional nanostructure has been developed as an "off-on" fluorescent probe for detection of target methyltransferase by integrating the elements of DNA tetrahedron, target recognition, and dual-labeled reporter. This sensing system is initially in an "OFF" state owing to the close proximity of fluorophores and quenchers. After the substrate is recognized by target methyltransferase, the DNA tetrahedron can be methylated to produce methylated DNA sites. These sites can be recognized and cut by the restriction endonuclease DpnI to bring about the collapse of the DNA tetrahedron, which leads to the separation of the dual-labeled reporters from the quenchers, and thus the recovery of fluorescence signal to produce an "ON" state. The proposed DNA tetrahedron-based sensing method can detect Dam methyltransferase in the range of 0.1-90 U mL-1 with a detection limit of 0.045 U mL-1 and shows good specificity and reproducibility for detection of Dam methyltransferase in a real sample. It has been successfully applied for screening various methylation inhibitors. Thus, this work possesses a promising prospect for detection of DNA methyltransfrase in the field of clinical diagnostics.
Collapse
Affiliation(s)
- Xiaoyan Zhou
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
- Department of Clinical Laboratory, The Affiliated Hospital of Medical College, Qingdao University , Qingdao 266101, China
| | - Min Zhao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
| | - Xiaolei Duan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
| | - Bin Guo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University , Chongqing 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
| | - Huangxian Ju
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University , Chongqing 400016, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University , Nanjing 210023, China
| |
Collapse
|
6
|
Fluorogenic Real-Time Reporters of DNA Repair by MGMT, a Clinical Predictor of Antitumor Drug Response. PLoS One 2016; 11:e0152684. [PMID: 27035132 PMCID: PMC4818092 DOI: 10.1371/journal.pone.0152684] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/17/2016] [Indexed: 01/26/2023] Open
Abstract
Common alkylating antitumor drugs, such as temozolomide, trigger their cytotoxicity by methylating the O6-position of guanosine in DNA. However, the therapeutic effect of these drugs is dampened by elevated levels of the DNA repair enzyme, O6-methylguanine DNA methyltransferase (MGMT), which directly reverses this alkylation. As a result, assessing MGMT levels in patient samples provides an important predictor of therapeutic response; however, current methods available to measure this protein are indirect, complex and slow. Here we describe the design and synthesis of fluorescent chemosensors that report directly on MGMT activity in a single step within minutes. The chemosensors incorporate a fluorophore and quencher pair, which become separated by the MGMT dealkylation reaction, yielding light-up responses of up to 55-fold, directly reflecting repair activity. Experiments show that the best-performing probe retains near-native activity at mid-nanomolar concentrations. A nuclease-protected probe, NR-1, was prepared and tested in tumor cell lysates, demonstrating an ability to evaluate relative levels of MGMT repair activity in twenty minutes. In addition, a probe was employed to evaluate inhibitors of MGMT, suggesting utility for discovering new inhibitors in a high-throughput manner. Probe designs such as that of NR-1 may prove valuable to clinicians in selection of patients for alkylating drug therapies and in assessing resistance that arises during treatment.
Collapse
|
7
|
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: 1.1] [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
|
8
|
Penketh PG, Patridge E, Shyam K, Baumann RP, Zhu R, Ishiguro K, Sartorelli AC. Influence of glutathione and glutathione S-transferases on DNA interstrand cross-link formation by 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine, the active anticancer moiety generated by laromustine. Chem Res Toxicol 2014; 27:1440-9. [PMID: 25012050 PMCID: PMC4137992 DOI: 10.1021/tx500197t] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
Prodrugs
of 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine
(90CE) are promising anticancer agents. The 90CE moiety is a readily
latentiated, short-lived (t1/2 ∼
30 s) chloroethylating agent that can generate high yields of oxophilic
electrophiles responsible for the chloroethylation of the O-6 position
of guanine in DNA. These guanine O-6 alkylations are believed to be
responsible for the therapeutic effects of 90CE and its prodrugs.
Thus, 90CE demonstrates high selectivity toward tumors with diminished
levels of O6-alkylguanine-DNA alkyltransferase
(MGMT), the resistance protein responsible for O6-alkylguanine repair. The formation of O6-(2-chloroethyl)guanine lesions ultimately leads to the generation
of highly cytotoxic 1-(N3-cytosinyl),-2-(N1-guaninyl)ethane DNA interstrand cross-links
via N1,O6-ethanoguanine
intermediates. The anticancer activity arising from this sequence
of reactions is thus identical to this component of the anticancer
activity of the clinically used chloroethylnitrosoureas. Herein, we
evaluate the ability of glutathione (GSH) and other low molecular
weight thiols, as well as GSH coupled with various glutathione S-transferase enzymes (GSTs) to attenuate the final yields
of cross-links generated by 90CE when added prior to or immediately
following the initial chloroethylation step to determine the major
point(s) of interaction. In contrast to studies utilizing BCNU as
a chloroethylating agent by others, GSH (or GSH/GST) did not appreciably
quench DNA interstrand cross-link precursors. While thiols alone offered
little protection at either alkylation step, the GSH/GST couple was
able to diminish the initial yields of cross-link precursors. 90CE
exhibited a very different GST isoenzyme susceptibility to that reported
for BCNU, this could have important implications in the relative resistance
of tumor cells to these agents. The protection afforded by GSH/GST
was compared to that produced by MGMT.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology and Yale Cancer Center, Yale University School of Medicine , New Haven, Connecticut 06520, United States
| | | | | | | | | | | | | |
Collapse
|
9
|
Oliver JA, Ortiz R, Melguizo C, Álvarez PJ, Gómez-Millán J, Prados J. Prognostic impact of MGMT promoter methylation and MGMT and CD133 expression in colorectal adenocarcinoma. BMC Cancer 2014; 14:511. [PMID: 25015560 PMCID: PMC4227111 DOI: 10.1186/1471-2407-14-511] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/07/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND New biomarkers are needed for the prognosis of advanced colorectal cancer, which remains incurable by conventional treatments. O6-methylguanine DNA methyltransferase (MGMT) methylation and protein expression have been related to colorectal cancer treatment failure and tumor progression. Moreover, the presence in these tumors of cancer stem cells, which are characterized by CD133 expression, has been associated with chemoresistance, radioresistance, metastasis, and local recurrence. The objective of this study was to determine the prognostic value of CD133 and MGMT and their possible interaction in colorectal cancer patients. METHODS MGMT and CD133 expression was analyzed by immunohistochemistry in 123 paraffin-embedded colorectal adenocarcinoma samples, obtaining the percentage staining and intensity. MGMT promoter methylation status was obtained by using bisulfite modification and methylation-specific PCR (MSP). These values were correlated with clinical data, including overall survival (OS), disease-free survival (DFS), tumor stage, and differentiation grade. RESULTS Low MGMT expression intensity was significantly correlated with shorter OS and was a prognostic factor independently of treatment and histopathological variables. High percentage of CD133 expression was significantly correlated with shorter DFS but was not an independent factor. Patients with low-intensity MGMT expression and ≥50% CD133 expression had the poorest DFS and OS outcomes. CONCLUSIONS Our results support the hypothesis that MGMT expression may be an OS biomarker as useful as tumor stage or differentiation grade and that CD133 expression may be a predictive biomarker of DFS. Thus, MGMT and CD133 may both be useful for determining the prognosis of colorectal cancer patients and to identify those requiring more aggressive adjuvant therapies. Future studies will be necessary to determine its clinical utility.
Collapse
Affiliation(s)
- Jaime Antonio Oliver
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18100, Spain
| | - Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18100, Spain
- Department of Health Sciences, University of Jaén, Jaén 23071, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18100, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada 18012, Spain
| | - Pablo Juan Álvarez
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18100, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, Granada, Spain
| | - Jaime Gómez-Millán
- Radiation Oncology Department, Hospital Clinico Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, Granada 18100, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-Universidad de Granada, Granada, Spain
- Department of Anatomy and Embryology, University of Granada, Granada 18012, Spain
| |
Collapse
|
10
|
Nagel ZD, Chaim IA, Samson LD. Inter-individual variation in DNA repair capacity: a need for multi-pathway functional assays to promote translational DNA repair research. DNA Repair (Amst) 2014; 19:199-213. [PMID: 24780560 DOI: 10.1016/j.dnarep.2014.03.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Why does a constant barrage of DNA damage lead to disease in some individuals, while others remain healthy? This article surveys current work addressing the implications of inter-individual variation in DNA repair capacity for human health, and discusses the status of DNA repair assays as potential clinical tools for personalized prevention or treatment of disease. In particular, we highlight research showing that there are significant inter-individual variations in DNA repair capacity (DRC), and that measuring these differences provides important biological insight regarding disease susceptibility and cancer treatment efficacy. We emphasize work showing that it is important to measure repair capacity in multiple pathways, and that functional assays are required to fill a gap left by genome wide association studies, global gene expression and proteomics. Finally, we discuss research that will be needed to overcome barriers that currently limit the use of DNA repair assays in the clinic.
Collapse
Affiliation(s)
- Zachary D Nagel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Isaac A Chaim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leona D Samson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
11
|
Penketh PG, Shyam K, Zhu R, Baumann RP, Ishiguro K, Sartorelli AC. Influence of phosphate and phosphoesters on the decomposition pathway of 1,2-bis(methylsulfonyl)-1-(2-chloroethyhydrazine (90CE), the active anticancer moiety generated by Laromustine, KS119, and KS119W. Chem Res Toxicol 2014; 27:818-33. [PMID: 24618018 PMCID: PMC4033638 DOI: 10.1021/tx500004y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Prodrugs of the short-lived chloroethylating
agent 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine
(90CE) and its methylating analogue 1,2-bis(methylsulfonyl)-1-(methyl)hydrazine
(KS90) are potentially useful anticancer agents. This class of agents
frequently yields higher ratios of therapeutically active oxophilic
electrophiles responsible for DNA O6-guanine
alkylations to other electrophiles with lower therapeutic relevance
than the nitrosoureas. This results in improved selectivity toward
tumors with diminished levels of O6-alkylguanine-DNA
alkyltransferase (MGMT), the resistance protein responsible for O6-alkylguanine repair. The formation of O6-(2-chloroethyl)guanine, which leads to the
formation of a DNA–DNA interstrand cross-link, accounts for
the bulk of the anticancer activity of 90CE prodrugs. Herein, we describe
a new decomposition pathway that is available to 90CE but not to its
methylating counterpart. This pathway appears to be subject to general/acid
base catalysis with phosphate (Pi), phosphomonoesters, and phosphodiesters,
being particularly effective. This pathway does not yield a chloroethylating
species and results in a major change in nucleophile preference since
thiophilic rather than oxophilic electrophiles are produced. Thus,
a Pi concentration dependent decrease in DNA–DNA interstand
cross-link formation was observed. Changes in 90CE decomposition products
but not alkylation kinetics occurred in the presence of Pi since the
prebranch point elimination of the N-1 methanesulfinate moiety remained
the rate-limiting step. The Pi catalyzed route is expected to dominate
at Pi and phosphoester concentrations totaling >25–35 mM.
In
view of the abundance of Pi and phosphoesters in cells, this pathway
may have important effects on agent toxicity, tumor selectivity, and
resistance to prodrugs of 90CE. Furthermore, it may be possible to
design analogues that diminish this thiophile-generating pathway,
which is likely superfluous at best and potentially detrimental to
the targeting of hypoxic regions where Pi concentrations can be significantly
elevated.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology and Yale Cancer Center, Yale University School of Medicine , 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | | | | | | | | | | |
Collapse
|