Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nature ; 524(7563): 114-8, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26200337

ABSTRACT

Cells require nucleotides to support DNA replication and repair damaged DNA. In addition to de novo synthesis, cells recycle nucleotides from the DNA of dying cells or from cellular material ingested through the diet. Salvaged nucleosides come with the complication that they can contain epigenetic modifications. Because epigenetic inheritance of DNA methylation mainly relies on copying of the modification pattern from parental strands, random incorporation of pre-modified bases during replication could have profound implications for epigenome fidelity and yield adverse cellular phenotypes. Although the salvage mechanism of 5-methyl-2'deoxycytidine (5mdC) has been investigated before, it remains unknown how cells deal with the recently identified oxidized forms of 5mdC: 5-hydroxymethyl-2'deoxycytidine (5hmdC), 5-formy-2'deoxycytidine (5fdC) and 5-carboxyl-2'deoxycytidine (5cadC). Here we show that enzymes of the nucleotide salvage pathway display substrate selectivity, effectively protecting newly synthesized DNA from the incorporation of epigenetically modified forms of cytosine. Thus, cell lines and animals can tolerate high doses of these modified cytidines without any deleterious effects on physiology. Notably, by screening cancer cell lines for growth defects after exposure to 5hmdC, we unexpectedly identify a subset of cell lines in which 5hmdC or 5fdC administration leads to cell lethality. Using genomic approaches, we show that the susceptible cell lines overexpress cytidine deaminase (CDA). CDA converts 5hmdC and 5fdC into variants of uridine that are incorporated into DNA, resulting in accumulation of DNA damage, and ultimately, cell death. Our observations extend current knowledge of the nucleotide salvage pathway by revealing the metabolism of oxidized epigenetic bases, and suggest a new therapeutic option for cancers, such as pancreatic cancer, that have CDA overexpression and are resistant to treatment with other cytidine analogues.


Subject(s)
Cytidine Deaminase/metabolism , Cytidine/analogs & derivatives , Cytidine/metabolism , Cytosine/metabolism , Cytosine/pharmacology , Epigenesis, Genetic , Neoplasms/drug therapy , 5-Methylcytosine/metabolism , 5-Methylcytosine/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cytidine/chemistry , Cytidine/pharmacology , Cytidine Deaminase/genetics , Cytosine/analogs & derivatives , Cytosine/chemistry , DNA/biosynthesis , DNA/chemistry , DNA Damage/drug effects , DNA-Directed DNA Polymerase/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Deoxycytidine/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasms/genetics , Neoplasms/metabolism , Nucleotides/chemistry , Nucleotides/metabolism , Nucleotides/pharmacology , Oxidation-Reduction , Phosphotransferases/metabolism , Substrate Specificity , Up-Regulation , Uridine/analogs & derivatives , Uridine/chemistry , Uridine/metabolism
2.
Angew Chem Int Ed Engl ; 58(4): 1007-1012, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30589164

ABSTRACT

Bromodomain-containing proteins are epigenetic modulators involved in a wide range of cellular processes, from recruitment of transcription factors to pathological disruption of gene regulation and cancer development. Since the druggability of these acetyl-lysine reader domains was established, efforts were made to develop potent and selective inhibitors across the entire family. Here we report the development of a small molecule-based approach to covalently modify recombinant and endogenous bromodomain-containing proteins by targeting a conserved lysine and a tyrosine residue in the variable ZA or BC loops. Moreover, the addition of a reporter tag allowed in-gel visualization and pull-down of the desired bromodomains.


Subject(s)
Carbamates/chemistry , Histones/chemistry , Lysine/chemistry , Protein Domains , Pyridazines/chemistry , Triazoles/chemistry , Acetylation , Amino Acid Sequence , Binding Sites , Conserved Sequence , Molecular Docking Simulation , Protein Binding
3.
Mol Cancer Ther ; 22(2): 254-263, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36722141

ABSTRACT

Antibody-drug conjugates (ADC) delivering pyrrolobenzodiazepine (PBD) DNA cross-linkers are currently being evaluated in clinical trials, with encouraging results in Hodgkin and non-Hodgkin lymphomas. The first example of an ADC delivering a PBD DNA cross-linker (loncastuximab tesirine) has been recently approved by the FDA for the treatment of relapsed and refractory diffuse large B-cell lymphoma. There has also been considerable interest in mono-alkylating PBD analogs. We conducted a head-to-head comparison of a conventional PBD bis-imine and a novel PBD mono-imine. Key Mitsunobu chemistry allowed clean and convenient access to the mono-imine class. Extensive DNA-binding studies revealed that the mono-imine mediated a type of DNA interaction that is described as "pseudo cross-linking," as well as alkylation. The PBD mono-imine ADC demonstrated robust antitumor activity in mice bearing human tumor xenografts at doses 3-fold higher than those that were efficacious for the PBD bis-imine ADC. A single-dose toxicology study in rats demonstrated that the MTD of the PBD mono-alkylator ADC was approximately 3-fold higher than that of the ADC bearing a bis-imine payload, suggesting a comparable therapeutic index for this molecule. However, although both ADCs caused myelosuppression, renal toxicity was observed only for the bis-imine, indicating possible differences in toxicologic profiles that could influence tolerability and therapeutic index. These data show that mono-amine PBDs have physicochemical and pharmacotoxicologic properties distinct from their cross-linking analogs and support their potential utility as a novel class of ADC payload.


Subject(s)
Immunoconjugates , Lymphoma, Non-Hodgkin , Humans , Animals , Mice , Rats , Alkylation , DNA , Imines , Immunoconjugates/pharmacology
4.
J Pharm Biomed Anal ; 205: 114287, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34385015

ABSTRACT

Antibody-drug conjugates (ADCs) are an emerging class of oncology treatments combining the unique specificity of monoclonal antibodies with the highly cytotoxic properties of small molecule compounds. Pyrrolobenzodiazepines (PBDs) are highly potent agents capable of inhibiting cellular DNA replication which leads to apoptosis. To ensure efficacy and patient safety upon administration of such toxic and heterogeneous molecules, their structure and quality attributes must be closely monitored. Size exclusion chromatography (SEC) is a powerful, fast and robust tool for the separation of compounds varying in molecular weight. When using volatile components in the chromatographic mobile phase, SEC has also been shown to be amenable for interfacing to mass spectrometry, providing potential for reliable identification of protein isoforms across the size variants present. Here, we present a SEC-MS method developed for the characterisation of PBD-based ADCs on the intact molecular level. We demonstrate that information on ADC monomers such as the glycoform distribution and the average drug-antibody ratio (DAR) can be obtained in 15 minutes of analysis time. Qualitative and quantitative information on low and high molecular weight impurities such as aggregates and fragments, fundamental for critical quality attribute analysis of biopharmaceuticals, can be generated simultaneously. SEC-MS enables the characterisation of multiple product quality attributes of complex biotherapeutics at the same time.


Subject(s)
Immunoconjugates , Antibodies, Monoclonal , Benzodiazepines , Chromatography, Gel , Humans , Immunoconjugates/analysis , Mass Spectrometry , Pyrroles
5.
Nat Commun ; 11(1): 2743, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488087

ABSTRACT

Concerted multidisciplinary efforts have led to the development of Cyclin-Dependent Kinase inhibitors (CDKi's) as small molecule drugs and chemical probes of intracellular CDK function. However, conflicting data has been reported on the inhibitory potency of CDKi's and a systematic characterization of affinity and selectivity against intracellular CDKs is lacking. We have developed a panel of cell-permeable energy transfer probes to quantify target occupancy for all 21 human CDKs in live cells, and present a comprehensive evaluation of intracellular isozyme potency and selectivity for a collection of 46 clinically-advanced CDKi's and tool molecules. We observed unexpected intracellular activity profiles for a number of CDKi's, offering avenues for repurposing of highly potent molecules as probes for previously unreported targets. Overall, we provide a broadly applicable method for evaluating the selectivity of CDK inhibitors in living cells, and present a refined set of tool molecules to study CDK function.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor Proteins/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , CDC2 Protein Kinase , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase 9 , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Phosphorylation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL