Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
2.
Article in English | MEDLINE | ID: mdl-35732105

ABSTRACT

The majority of women have health problems that require medication after giving birth. Complications such as allergies, postpartum depression, and diabetes are often treated with drugs such as cetirizine, venlafaxine, and metformin, respectively. These treatments are considered safe during lactation, but information of the transfer of drugs to breast milk and possible effects on the infant is scarce. Therefore, this needs to be systematically investigated in larger populations. To enable the determination of drug transfer, we here describe the validation of two rapid, sensitive, and high-throughput analysis methods for 1) simultaneous quantification of cetirizine, venlafaxine, and O-desmethylvenlafaxine in human breast milk, and 2) metformin in human breast milk and plasma. In both methods, a simple protein precipitation protocol with acetonitrile and benchtop-centrifugation was used prior to compound analysis with liquid-chromatography tandem mass spectrometry. The methods had linear ranges between 0.39 - 194.5 ng/mL for cetirizine, 0.28 - 138.7 ng/mL for venlafaxine, 0.26 - 131.7 ng/mL for O-desmethylvenlafaxine, in milk, and 0.65 - 193.7 ng/mL for metformin in both milk and plasma. Intra-run and inter-run precision and accuracy were ≤ 9% for cetirizine, venlafaxine, and O-desmethylvenlafaxine in milk, and ≤ 7% for metformin in milk and plasma. Cetirizine was measured to median milk concentrations of 13 ng/mL (range: 0.65 - 65 ng/mL) in 228 donor samples from breast-feeding women.


Subject(s)
Metformin , Tandem Mass Spectrometry , Cetirizine , Chromatography, High Pressure Liquid/methods , Desvenlafaxine Succinate , Female , Humans , Infant , Milk, Human , Pregnancy , Reproducibility of Results , Tandem Mass Spectrometry/methods , Venlafaxine Hydrochloride
3.
J Med Chem ; 63(8): 3915-3934, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32212728

ABSTRACT

Human dihydroorotate dehydrogenase (DHODH), an enzyme in the de novo pyrimidine synthesis pathway, is a target for the treatment of rheumatoid arthritis and multiple sclerosis and is re-emerging as an attractive target for cancer therapy. Here we describe the optimization of recently identified tetrahydroindazoles (HZ) as DHODH inhibitors. Several of the HZ analogues synthesized in this study are highly potent inhibitors of DHODH in an enzymatic assay, while also inhibiting cancer cell growth and viability and activating p53-dependent transcription factor activity in a reporter cell assay. Furthermore, we demonstrate the specificity of the compounds toward the de novo pyrimidine synthesis pathway through supplementation with an excess of uridine. We also show that induction of the DNA damage marker γ-H2AX after DHODH inhibition is preventable by cotreatment with the pan-caspase inhibitor Z-VAD-FMK. Additional solubility and in vitro metabolic stability profiling revealed compound 51 as a favorable candidate for preclinical efficacy studies.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Indazoles/chemistry , Indazoles/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Animals , Cell Survival/drug effects , Cell Survival/physiology , Dihydroorotate Dehydrogenase , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Female , Humans , Indazoles/pharmacology , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism
4.
Nat Commun ; 9(1): 2071, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29789663

ABSTRACT

The original PDF version of this Article listed the authors as "Marcus J.G.W. Ladds," where it should have read "Marcus J. G. W. Ladds, Ingeborg M. M. van Leeuwen, Catherine J. Drummond et al.#".Also in the PDF version, it was incorrectly stated that "Correspondence and requests for materials should be addressed to S. Lín.", instead of the correct "Correspondence and requests for materials should be addressed to S. Laín."This has been corrected in the PDF version of the Article. The HTML version was correct from the time of publication.

5.
Nat Commun ; 9(1): 1107, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549331

ABSTRACT

The development of non-genotoxic therapies that activate wild-type p53 in tumors is of great interest since the discovery of p53 as a tumor suppressor. Here we report the identification of over 100 small-molecules activating p53 in cells. We elucidate the mechanism of action of a chiral tetrahydroindazole (HZ00), and through target deconvolution, we deduce that its active enantiomer (R)-HZ00, inhibits dihydroorotate dehydrogenase (DHODH). The chiral specificity of HZ05, a more potent analog, is revealed by the crystal structure of the (R)-HZ05/DHODH complex. Twelve other DHODH inhibitor chemotypes are detailed among the p53 activators, which identifies DHODH as a frequent target for structurally diverse compounds. We observe that HZ compounds accumulate cancer cells in S-phase, increase p53 synthesis, and synergize with an inhibitor of p53 degradation to reduce tumor growth in vivo. We, therefore, propose a strategy to promote cancer cell killing by p53 instead of its reversible cell cycle arresting effect.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Neoplasms/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dihydroorotate Dehydrogenase , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Proteolysis/drug effects , Tumor Suppressor Protein p53/genetics
7.
J Pharm Biomed Anal ; 104: 1-11, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25459754

ABSTRACT

MTH1 is a protein that is required for cancer cell survival and is overexpressed in cancer cells. TH588 and TH287 are two new compounds that inhibit the MTH1 protein. The inhibitors were tested in pharmacokinetic studies on mice. A bioanalytical method was developed and validated for determination in mice plasma. The method was based on protein precipitation followed by LC-MS/MS analysis. The separation was performed on an Ascentis Express RP-Amide C18 column. The mass spectrometer was operated in positive electrospray ionization mode and the analytes were determined with multiple reaction monitoring (MRM). Abundant monoisotopic fragments were used for quantification. Two additional fragments were used for conformational analysis. The recovery of the compounds in plasma varied between 61 and 91% and the matrix effects were low and ranged between -3% and +2%. The method showed to be selective, linear, accurate and precise, and applicable for preclinical pharmacokinetic studies of TH588 and TH287 in mouse plasma. Half-life (T1/2) was ≤3.5h and maximum concentration (Cmax) ranged between 0.82 and 338µM for the different administration routes and compounds.


Subject(s)
Antineoplastic Agents/blood , Phosphoric Monoester Hydrolases/blood , Pyrimidines/blood , Animals , Antineoplastic Agents/pharmacokinetics , Female , Gas Chromatography-Mass Spectrometry , Half-Life , Mice , Mice, Inbred C57BL , Pyrimidines/pharmacokinetics , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization
8.
Nature ; 508(7495): 215-21, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24695224

ABSTRACT

Cancers have dysfunctional redox regulation resulting in reactive oxygen species production, damaging both DNA and free dNTPs. The MTH1 protein sanitizes oxidized dNTP pools to prevent incorporation of damaged bases during DNA replication. Although MTH1 is non-essential in normal cells, we show that cancer cells require MTH1 activity to avoid incorporation of oxidized dNTPs, resulting in DNA damage and cell death. We validate MTH1 as an anticancer target in vivo and describe small molecules TH287 and TH588 as first-in-class nudix hydrolase family inhibitors that potently and selectively engage and inhibit the MTH1 protein in cells. Protein co-crystal structures demonstrate that the inhibitors bind in the active site of MTH1. The inhibitors cause incorporation of oxidized dNTPs in cancer cells, leading to DNA damage, cytotoxicity and therapeutic responses in patient-derived mouse xenografts. This study exemplifies the non-oncogene addiction concept for anticancer treatment and validates MTH1 as being cancer phenotypic lethal.


Subject(s)
DNA Repair Enzymes/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Nucleotides/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Animals , Catalytic Domain , Cell Death/drug effects , Cell Survival/drug effects , Crystallization , DNA Damage , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/metabolism , Deoxyguanine Nucleotides/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Models, Molecular , Molecular Conformation , Molecular Targeted Therapy , Neoplasms/pathology , Oxidation-Reduction/drug effects , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrophosphatases/antagonists & inhibitors , Reproducibility of Results , Xenograft Model Antitumor Assays , Nudix Hydrolases
9.
Article in English | MEDLINE | ID: mdl-23153637

ABSTRACT

In this study a rapid liquid chromatography-time-of-flight mass spectrometry method was developed, validated and applied in order to evaluate the potential of this technique for routine urine drug testing. Approximately 800 authentic patient samples were analyzed for amphetamines (amphetamine and methamphetamine), opiates (morphine, morphine-3-glucuronide, morphine-6-glucuronide, codeine and codeine-6-glucuronide) and buprenorphines (buprenorphine and buprenorphine-glucuronide) using immunochemical screening assays and mass spectrometry confirmation methods for comparison. The chromatographic application utilized a rapid gradient with high flow and a reversed phase column with 1.8 µm particles. Total analysis time was 4 min. The mass spectrometer operated with an electrospray interface in positive mode with a resolution power of >10,000 at m/z 956. The applied reporting limits were 100 ng/mL for amphetamines and opiates, and 5 ng/mL for buprenorphines, with lower limits of quantification were 2.8-41 ng/mL. Calibration curves showed a linear response with coefficients of correlation of 0.97-0.99. The intra- and interday imprecision in quantification at the reporting limits were <10% for all analytes but for buprenorphines <20%. Method validation data met performance criteria for a qualitative and quantitative method. The liquid chromatography-time-of-flight mass spectrometry method was found to be more selective than the immunochemical method by producing lower rates of false positives (0% for amphetamines and opiates; 3.2% for buprenorphines) and negatives (1.8% for amphetamines; 0.6% for opiates; 0% for buprenorphines). The overall agreement between the two screening methods was between 94.2 and 97.4%. Comparison of data with the confirmation (LC-MS) results for all individual 9 analytes showed that most deviating results were produced in samples with low levels of analytes. False negatives were mainly related to failure of detected peak to meet mass accuracy criteria (±20 mDa). False positives was related to presence of interfering peaks meeting mass accuracy and retention time criteria and occurred mainly at low levels. It is concluded that liquid chromatography-time-of-flight mass spectrometry has potential both as a complement and as replacement of immunochemical screening assays.


Subject(s)
Chromatography, Liquid/methods , Illicit Drugs/urine , Spectrometry, Mass, Electrospray Ionization/methods , Substance Abuse Detection/methods , Urinalysis/methods , Amphetamines/chemistry , Amphetamines/urine , Buprenorphine/chemistry , Buprenorphine/urine , Codeine/analogs & derivatives , Codeine/chemistry , Codeine/urine , High-Throughput Screening Assays/methods , Humans , Illicit Drugs/chemistry , Linear Models , Morphine/chemistry , Morphine/urine , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...