Register      Login
Australian Journal of Chemistry Australian Journal of Chemistry Society
An international journal for chemical science
REVIEW

Chemistry, biochemistry and clinical relevance of the glutamine metabolite α-ketoglutaramate/2-hydroxy-5-oxoproline

Travis T. Denton https://orcid.org/0000-0002-1222-2538 A B C * and Arthur J. L. Cooper https://orcid.org/0000-0002-9143-8504 D *
+ Author Affiliations
- Author Affiliations

A Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA, USA.

B Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, USA.

C Steve Gleason Institute for Neuroscience, Washington State University Health Sciences Spokane, Spokane, WA, USA.

D Department of Biochemistry and Molecular Biology, New York Medical College, 15 Dana Road, Valhalla, NY 10595, USA.




Dr. Denton obtained his BS degree in chemistry from Central Washington University and a PhD in chemistry from the University of Montana. Dr Denton performed a postdoctoral fellowship at the Human BioMolecular Research Institute in San Diego, CA, followed by a second postdoctoral fellowship at the Shafizadeh Rocky Mountain Center for Wood and Carbohydrate Chemistry at the University of Montana. He then moved on to become an Assistant Professor of chemistry at Eastern Washington University where he taught organic chemistry and performed medicinal chemistry research. Currently, Dr. Denton is an Assistant Professor at Washington State University Health Sciences Spokane in the College of Pharmacy and Pharmaceutical Sciences. Dr. Denton is an inaugural fellow in the Steve Gleason Institute for Neuroscience at Washington State University and is an affiliate Assistant Professor in the Elson S. Floyd College of Medicine at WSU Health Sciences Spokane. Dr. Denton leads medicinal chemistry/molecular biology efforts in the fields of glutamine addicted cancers, modulation of free radical biology at the level of the TCA cycle, development of small molecules for the treatment of neurological disorders, design and synthesis of small molecule inhibitors of CYP2A6 as smoking cessation agents, and preparation of carbohydrate-based polyhydroxypolyamides (polymers) for biomedical applications.



Dr. Cooper obtained BSc (chemistry and zoology) and DSc (biochemistry) degrees from London University, an MSc degree (Biochemistry) from Imperial College, a PhD degree (biochemistry) from Weill Cornell Medicine (WCM) and a postdoctoral fellowship at Brandeis University (biochemistry). He has held professorships at WCM and New York Medical College (NYMC). He is currently Emeritus Professor of Biochemistry and Molecular Biology at NYMC and an Adjunct Professor at WCM. He is on the editorial board of several journals and is Editor-in-Chief of Analytical Biochemistry. His research interests include pyridoxal 5′-phosphate enzymes, enzyme mechanisms, bioactivation mechanisms, neurochemistry, neurodegenerative diseases, cancer biochemistry, chemoprevention, and 1-C, nitrogen, sulfur and selenium biochemistry.


Handling Editor: Mibel Aguilar

Australian Journal of Chemistry 76(8) 361-371 https://doi.org/10.1071/CH22264
Submitted: 16 December 2022  Accepted: 5 May 2023   Published: 11 July 2023

© 2023 The Author(s) (or their employer(s)). Published by CSIRO Publishing.

Abstract

In the glutaminase II pathway (which we now refer to as the glutamine transaminase-ω-amidase (GTωA) pathway), l-glutamine is transaminated to α-ketoglutaramate (KGM), which, in turn, is hydrolyzed to α-ketoglutarate and ammonia by an enzyme known as ω-amidase. Despite the fact that the GTωA pathway was discovered more than 70 years ago, and is widespread in nature, the pathway has received limited attention. This is partly due to the broad amino acid/α-keto acid specificity of the glutamine transaminases, which has led to confusion over nomenclature and in assigning precise biological roles. Secondly, the α-keto acid product of glutamine transaminases – KGM – has not, until recently, become available in pure form. Here, we briefly discuss the metabolic importance of the GTωA pathway in microorganisms, plants and mammals. We pay special attention to the chemistry of KGM and methods for its synthesis. We discuss the importance of KGM as a biomarker for hyperammonemic diseases. We provide evidence that the GTωA pathway satisfies, in part, ‘glutamine addiction’ in a variety of cancer cells. We show that the anti-cancer drugs 6-diazo-5-oxo-l-norleucine and l-azaserine are transaminase and β-lyase substrates of glutamine transaminase K, respectively. We suggest that there is a pressing need for the development of: (1) inexpensive and scaled-up procedures for the synthesis of KGM to facilitate research on the biological importance of the GTωA pathway in mammalian and human tissues and in agricultural research; and (2) potent and selective inhibitors of ω-amidase, both as anti-cancer agents and as a means for investigating the detailed enzyme mechanism.

Keywords: 2-Hydroxy-5-oxoproline, glutaminase 1 (GLS1), glutaminase 2 (GLS2), glutaminase II pathway, glutamine transaminases, α-ketoglutaramate, α-ketoglutarate, α-keto-γ-methiolbutyrate, ω-amidase.


References

[1]  H Cederkvist, SS Kolan, JA Wik, Z Sener, BS Skålhegg, Identification and characterization of a novel glutaminase inhibitor. FEBS Open Bio 2022, 12, 163.
         | Identification and characterization of a novel glutaminase inhibitor.Crossref | GoogleScholarGoogle Scholar |

[2]  T Dorai, JT Pinto, TT Denton, BF Krasnikov, AJL Cooper, The metabolic importance of the glutaminase II pathway in normal and cancerous cells. Anal Biochem 2022, 644, 114083.
         | The metabolic importance of the glutaminase II pathway in normal and cancerous cells.Crossref | GoogleScholarGoogle Scholar |

[3]  AJL Cooper, YI Shurubor, T Dorai, JT Pinto, EP Isakova, YI Deryabina, TT Denton, BF Krasnikov, ω-Amidase: an underappreciated, but important enzyme in l-glutamine and l-asparagine metabolism; relevance to sulfur and nitrogen metabolism, tumor biology and hyperammonemic diseases. Amino Acids 2015, 48, 1.
         | ω-Amidase: an underappreciated, but important enzyme in l-glutamine and l-asparagine metabolism; relevance to sulfur and nitrogen metabolism, tumor biology and hyperammonemic diseases.Crossref | GoogleScholarGoogle Scholar |

[4]  AJL Cooper, T Dorai, JT Pinto, TT Denton, α-Ketoglutaramate – a key metabolite contributing to glutamine addiction in cancer cells. Front Med 2022, 13, 1035335.
         | α-Ketoglutaramate – a key metabolite contributing to glutamine addiction in cancer cells.Crossref | GoogleScholarGoogle Scholar |

[5]  AJL Cooper, T Dorai, JT Pinto, TT Denton, Metabolic heterogeneity, plasticity and adaptation to ‘glutamine addiction’ in cancer cells: the role of glutaminases [GLS1/GLS2] and the glutaminase II [glutamine transaminase – ω-amidase (GTωA)] pathway. Biology in press

[6]  PE Hanna, MW Anders, The mercapturic acid pathway. Crit Rev Toxicol 2019, 49, 819.
         | The mercapturic acid pathway.Crossref | GoogleScholarGoogle Scholar |

[7]  AJL Cooper, BF Krasnikov, ZV Niatsetskaya, JT Pinto, PS Callery, MT Villar, A Artigues, SA Bruschi, Cysteine S-conjugate β-lyases: important roles in the metabolism of naturally occurring sulfur and selenium-containing compounds, xenobiotics and anticancer agents. Amino Acids 2011, 41, 7.
         | Cysteine S-conjugate β-lyases: important roles in the metabolism of naturally occurring sulfur and selenium-containing compounds, xenobiotics and anticancer agents.Crossref | GoogleScholarGoogle Scholar |

[8]  W Dekant, Biosynthesis of toxic glutathione conjugates from halogenated alkenes. Toxicol Lett 2003, 144, 49.
         | Biosynthesis of toxic glutathione conjugates from halogenated alkenes.Crossref | GoogleScholarGoogle Scholar |

[9]  AS Andrew, M Li, X Shi, JR Rees, KM Craver, JM Petali, Kidney cancer risk associated with historic groundwater trichloroethylene contamination. Int J Environ Res Public Health 2022, 19, 618.
         | Kidney cancer risk associated with historic groundwater trichloroethylene contamination.Crossref | GoogleScholarGoogle Scholar |

[10]  X-Y Zhang, AA Elfarra, Toxicity mechanism-based prodrugs: glutathione-dependent bioactivation as a strategy for anticancer prodrug design. Expert Opin Drug Discov 2018, 13, 815.
         | Toxicity mechanism-based prodrugs: glutathione-dependent bioactivation as a strategy for anticancer prodrug design.Crossref | GoogleScholarGoogle Scholar |

[11]  JNM Commandeur, I Andreadou, M Rooseboom, M Out, LJ de Leur, E Groot, NPE Vermeulen, Bioactivation of selenocysteine Se-conjugates by a highly purified rat renal cysteine conjugate β-lyase/glutamine transaminase K. J Pharmacol Exp Ther 2000, 294, 753.

[12]  YC Tung, ML Tsai, FL Kuo, CS Lai, V Badmaev, CT Ho, MH Pan, Se-Methyl-l-selenocysteine induces apoptosis via endoplasmic reticulum stress and the death receptor pathway in human colon adenocarcinoma COLO 205 Cells. J Agric Food Chem 2015, 63, 5008.
         | Se-Methyl-l-selenocysteine induces apoptosis via endoplasmic reticulum stress and the death receptor pathway in human colon adenocarcinoma COLO 205 Cells.Crossref | GoogleScholarGoogle Scholar |

[13]  AK Selvam, R Jawad, R Gramignoli, A Achour, H Salter, M Björnstedt, Novel mRNA-mediated and microRNA-guided approach to specifically eradicate drug-resistant hepatocellular carcinoma cell lines by Se-methylselenocysteine. Antioxidants (Basel) 2021, 10, 1094.
         | Novel mRNA-mediated and microRNA-guided approach to specifically eradicate drug-resistant hepatocellular carcinoma cell lines by Se-methylselenocysteine.Crossref | GoogleScholarGoogle Scholar |

[14]  J Calderón, E Morett, J Mora, ω-Amidase pathway in the degradation of glutamine in Neurospora crassa. J Bacteriol 1985, 161, 807.
         | ω-Amidase pathway in the degradation of glutamine in Neurospora crassa.Crossref | GoogleScholarGoogle Scholar |

[15]  J Calderón, J Mora, Glutamine assimilation pathways in Neurospora crassa growing on glutamine as sole nitrogen and carbon source. J Gen Microbiol 1989, 135, 2699.
         | Glutamine assimilation pathways in Neurospora crassa growing on glutamine as sole nitrogen and carbon source.Crossref | GoogleScholarGoogle Scholar |

[16]  PJ Unkefer, TJ Knight, RA Martinez, The intermediate in a nitrate-responsive ω-amidase pathway in plants may signal ammonium assimilation status. Plant Physiol 2023, 191, 715.
         | The intermediate in a nitrate-responsive ω-amidase pathway in plants may signal ammonium assimilation status.Crossref | GoogleScholarGoogle Scholar |

[17]  C Cobzaru, P Ganas, M Mihasan, P Schleberger, R Brandsch, Homologous gene clusters of nicotine catabolism, including a new ω-amidase for α-ketoglutaramate, in species of three genera of Gram-positive bacteria. Res Microbiol 2011, 162, 285.
         | Homologous gene clusters of nicotine catabolism, including a new ω-amidase for α-ketoglutaramate, in species of three genera of Gram-positive bacteria.Crossref | GoogleScholarGoogle Scholar |

[18]  CH Chien, QZ Gao, AJL Cooper, JH Lyu, SY Sheu, Structural insights into the catalytic active site and activity of human Nit2/ω-amidase: kinetic assay and molecular dynamics simulation. J Biol Chem 2012, 287, 25715.
         | Structural insights into the catalytic active site and activity of human Nit2/ω-amidase: kinetic assay and molecular dynamics simulation.Crossref | GoogleScholarGoogle Scholar |

[19]  CS Silva Teixeira, SF Sousa, NMFSA Cerqueira, An unsual cys-glu-lys catalytic triad is responsible for the catalytic mechanism of the nitrilase superfamily: a QM/MM study on Nit2. ChemPhysChem 2021, 22, 796.
         | An unsual cys-glu-lys catalytic triad is responsible for the catalytic mechanism of the nitrilase superfamily: a QM/MM study on Nit2.Crossref | GoogleScholarGoogle Scholar |

[20]  F Caligiore, E Zangelmi, C Vetro, T Kentache, JP Dewulf, M Veiga-da-Cunha, E Van Schaftingen, G Bommer, A Peracchi, Human cytosolic transaminases: side activities and patterns of discrimination towards physiologically available alternative substrates. Cell Mol Life Sci 2022, 79, 421.
         | Human cytosolic transaminases: side activities and patterns of discrimination towards physiologically available alternative substrates.Crossref | GoogleScholarGoogle Scholar |

[21]  TE Duffy, AJL Cooper, A Meister, Identification of α-ketoglutaramate in rat liver, kidney, and brain. Relationship to glutamine transaminase and ω-amidase activities. J Biol Chem 1974, 249, 7603.
         | Identification of α-ketoglutaramate in rat liver, kidney, and brain. Relationship to glutamine transaminase and ω-amidase activities.Crossref | GoogleScholarGoogle Scholar |

[22]  YI Shurubor, AJL Cooper, E,P Isakova, YI Deryabina, MF Beal, BF Krasnikov, HPLC determination of α-ketoglutaramate [5-amino-2,5-dioxopentanoate] in biological samples. Anal Biochem 2016, 494, 52.
         | HPLC determination of α-ketoglutaramate [5-amino-2,5-dioxopentanoate] in biological samples.Crossref | GoogleScholarGoogle Scholar |

[23]  TE Duffy, F Vergara, F Plum, Alpha-Ketoglutaramate in hepatic encephalopathy. Res Publ Assoc Res Nerv Ment Dis 1974, 53, 39.

[24]  F Vergara, F Plum, TE Duffy, α-Ketoglutaramate: increased concentrations in the cerebrospinal fluid of patients in hepatic coma. Science 1974, 183, 81.
         | α-Ketoglutaramate: increased concentrations in the cerebrospinal fluid of patients in hepatic coma.Crossref | GoogleScholarGoogle Scholar |

[25]  T Kuhara, Y Inoue, M Ohse, BF Krasnikov, AJL Cooper, Urinary 2-hydroxy-5-oxoproline, the lactam form of α-ketoglutaramate, is markedly increased in urea cycle disorders. Anal Bioanal Chem 2011, 400, 1843.
         | Urinary 2-hydroxy-5-oxoproline, the lactam form of α-ketoglutaramate, is markedly increased in urea cycle disorders.Crossref | GoogleScholarGoogle Scholar |

[26]  T Kuhara, M Ohse, Y Inoue, AJL Cooper, A GC/MS-based metabolomic approach for diagnosing citrin deficiency. Anal Bioanal Chem 2011, 400, 1881.
         | A GC/MS-based metabolomic approach for diagnosing citrin deficiency.Crossref | GoogleScholarGoogle Scholar |

[27]  T Dorai, B Dorai, JT Pinto, M Grasso, AJL Cooper, High Levels of Glutaminase II Pathway Enzymes in Normal and Cancerous Prostate Suggest a Role in ‘Glutamine Addiction’. Biomolecules 2019, 10, 2.
         | High Levels of Glutaminase II Pathway Enzymes in Normal and Cancerous Prostate Suggest a Role in ‘Glutamine Addiction’.Crossref | GoogleScholarGoogle Scholar |

[28]  S Udupa, S Nguyen, G Hoang, T Nguyen, A Quinones, K Pham, R Asaka, K Nguyen, C Zhang, A Elgogary, JG Jung, Q Xu, J Fu, AG Thomas, T Tsukamoto, J Hanes, BS Slusher, AJL Cooper, A Le, Upregulation of the Glutaminase II Pathway Contributes to Glutamate Production upon Glutaminase 1 Inhibition in Pancreatic Cancer. Proteomics 2019, 19, e1800451.
         | Upregulation of the Glutaminase II Pathway Contributes to Glutamate Production upon Glutaminase 1 Inhibition in Pancreatic Cancer.Crossref | GoogleScholarGoogle Scholar |

[29]  K Pham, AR Hanaford, BA Poore, MJ Maxwell, H Sweeney, A Parthasarathy, J Alt, R Rais, BS Slusher, CG Eberhart, EH Raabe, Comprehensive metabolic profiling of myc-amplified medulloblastoma tumors reveals key dependencies on amino acid, tricarboxylic acid and hexosamine pathways. Cancers 2022, 14, 1311.
         | Comprehensive metabolic profiling of myc-amplified medulloblastoma tumors reveals key dependencies on amino acid, tricarboxylic acid and hexosamine pathways.Crossref | GoogleScholarGoogle Scholar |

[30]  YA Shen, CL Chen, YH Huang, EE Evans, CC Cheng, YJ Chuang, C Zhang, A Le, Inhibition of glutaminolysis in combination with other therapies to improve cancer treatment. Curr Opin Chem Biol 2021, 62, 64.
         | Inhibition of glutaminolysis in combination with other therapies to improve cancer treatment.Crossref | GoogleScholarGoogle Scholar |

[31]  Q Qiu, L Yang, Y Feng, Z Zhu, N Li, L Zheng, Y Sun, C Pan, H Qiu, X Cui, W He, F Wang, Y Yi, M Tang, Z Yang, Y Yang, Z Li, L Chen, Y Hu, HDAC I/IIb selective inhibitor purinostat mesylate combined with GLS1 inhibition effectively eliminates CML stem cells. Bioact Mater 2023, 21, 483.
         | HDAC I/IIb selective inhibitor purinostat mesylate combined with GLS1 inhibition effectively eliminates CML stem cells.Crossref | GoogleScholarGoogle Scholar |

[32]  KR Jacobs, G Castellano-Gonzalez, GJ Guillemin, DB Lovejoy, Major developments in the design of inhibitors along the kynurenine pathway. Curr Med Chem 2017, 24, 2471.
         | Major developments in the design of inhibitors along the kynurenine pathway.Crossref | GoogleScholarGoogle Scholar |

[33]  A Nematollahi, G Sun, GS Jayawickrama, WB Church, Kynurenine aminotransferase isozyme inhibitors: a review. Int J Mol Sci 2016, 17, 946.
         | Kynurenine aminotransferase isozyme inhibitors: a review.Crossref | GoogleScholarGoogle Scholar |

[34]  R Schwarcz, TW Stone, The kynurenine pathway and the brain: challenges, controversies and promises. Neuropharmacology 2017, 112, 237.
         | The kynurenine pathway and the brain: challenges, controversies and promises.Crossref | GoogleScholarGoogle Scholar |

[35]  A Meister, Preparation of enzymatic reactions of the keto analogues of asparagine and glutamine. J Biol Chem 1953, 200, 571.
         | Preparation of enzymatic reactions of the keto analogues of asparagine and glutamine.Crossref | GoogleScholarGoogle Scholar |

[36]  TT Otani, A Meister, ω-Amide and ω-amino acid derivatives of α-ketoglutaric and oxalacetic acids. J Biol Chem 1957, 224, 137.
         | ω-Amide and ω-amino acid derivatives of α-ketoglutaric and oxalacetic acids.Crossref | GoogleScholarGoogle Scholar |

[37]  LB Hersh, Rat liver ω-amidase. Purification and properties. Biochemistry 1971, 10, 2884.
         | Rat liver ω-amidase. Purification and properties.Crossref | GoogleScholarGoogle Scholar |

[38]  AJL Copper, A Meister, Isolation and properties of highly purified glutamine transaminase. Biochemistry 1972, 11, 661.
         | Isolation and properties of highly purified glutamine transaminase.Crossref | GoogleScholarGoogle Scholar |

[39]  AJL Cooper, Spot test for the detection of α-ketoglutaramic acid in human cerebrospinal fluid. Anal Biochem 1978, 90, 444.
         | Spot test for the detection of α-ketoglutaramic acid in human cerebrospinal fluid.Crossref | GoogleScholarGoogle Scholar |

[40]  BF Krasnikov, R Nostramo, JT Pinto, AJL Cooper, Assay and purification of ω-amidase/Nit2, a ubiquitously expressed putative tumor suppressor that catalyzes the deamidation of the α-keto acid analogues of glutamine and asparagine. Anal Biochem 2009, 391, 144.
         | Assay and purification of ω-amidase/Nit2, a ubiquitously expressed putative tumor suppressor that catalyzes the deamidation of the α-keto acid analogues of glutamine and asparagine.Crossref | GoogleScholarGoogle Scholar |

[41]  M Nikulin, V Drobot, V Švedas, BF Krasnikov, Preparative biocatalytic synthesis of α-ketoglutaramate. Int J Mol Sci 2021, 22, 12748.
         | Preparative biocatalytic synthesis of α-ketoglutaramate.Crossref | GoogleScholarGoogle Scholar |

[42]  D Shen, L Kruger, T Deatherage, TT Denton, Synthesis of α-ketoglutaramic acid. Anal Biochem 2020, 607, 113862.
         | Synthesis of α-ketoglutaramic acid.Crossref | GoogleScholarGoogle Scholar |

[43]  VA Hariharan, TT Denton, S Paraszcszak, K McEvoy, TM Jeitner, BF Krasnikov, AJL Cooper, The enzymology of 2-hydroxyglutarate, 2-hydroxyglutaramate and 2-hydroxysuccinamate and their relationship to oncometabolites. Biology (Basel) 2017, 6, 24.
         | The enzymology of 2-hydroxyglutarate, 2-hydroxyglutaramate and 2-hydroxysuccinamate and their relationship to oncometabolites.Crossref | GoogleScholarGoogle Scholar |

[44]  Martinez RA, Unkefer PJ. Preparation of 2-hydroxy-5-oxoproline and analogs thereof (U.S. Patent No. 6288240B1); 2001. Available at https://patents.google.com/patent/US6288240B1/en

[45]  L Deng, ZH Zhou, Spontaneous conversions of glutamine, histidine and arginine into α-hydroxycarboxylates with NH4VO3 or V2O5. Dalton Trans 2020, 49, 11921.
         | Spontaneous conversions of glutamine, histidine and arginine into α-hydroxycarboxylates with NH4VO3 or V2O5.Crossref | GoogleScholarGoogle Scholar |

[46]  AJL Cooper, Asparagine transaminase from rat liver. J Biol Chem 1977, 252, 2032.
         | Asparagine transaminase from rat liver.Crossref | GoogleScholarGoogle Scholar |

[47]  KM Lemberg, JJ Vornov, R Rais, BS Slusher, We’re not ‘DON’ yet: optimal dosing and prodrug delivery of 6-diazo-5-oxo-L-norleucine. Mol Cancer Ther 2018, 17, 1824.
         | We’re not ‘DON’ yet: optimal dosing and prodrug delivery of 6-diazo-5-oxo-L-norleucine.Crossref | GoogleScholarGoogle Scholar |

[48]  Q Han, J Li, J Li, pH dependence, substrate specificity and inhibition of human kynurenine aminotransferase I. Eur J Biochem 2004, 271, 4804.
         | pH dependence, substrate specificity and inhibition of human kynurenine aminotransferase I.Crossref | GoogleScholarGoogle Scholar |

[49]  JT Pinto, BF Krasnikov, S Alcutt, ME Jones, T Dorai, MT Villar, A Artigues, J Li, AJL Cooper, Kynurenine aminotransferase III and glutamine transaminase L are identical enzymes that have cysteine S-conjugate β-lyase activity and can transaminate l-selenomethionine. J Biol Chem 2014, 289, 30950.
         | Kynurenine aminotransferase III and glutamine transaminase L are identical enzymes that have cysteine S-conjugate β-lyase activity and can transaminate l-selenomethionine.Crossref | GoogleScholarGoogle Scholar |

[50]  JA Jacquez, JH Sherman, Enzymatic degradation of azaserine. Cancer Res 1962, 22, 56.

[51]  H Rosenkrantz, R Sprague, U Schaeppi, RF Pittillo, DA Cooney, RD Davis, The stoichiometric fate of azaserine metabolized in vitro by tissues from azaserine-treated dogs and mice. Toxicol Appl Pharmacol 1972, 22, 607.
         | The stoichiometric fate of azaserine metabolized in vitro by tissues from azaserine-treated dogs and mice.Crossref | GoogleScholarGoogle Scholar |

[52]  SM Geisen, CMN Aloisi, SM Huber, ES Sandell, NA Escher, SJ Sturla, Direct alkylation of deoxyguanosine by azaserine leads to O6-carboxymethyldeoxyguanosine. Chem Res Toxicol 2021, 34, 1518.
         | Direct alkylation of deoxyguanosine by azaserine leads to O6-carboxymethyldeoxyguanosine.Crossref | GoogleScholarGoogle Scholar |