Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Yakugaku Zasshi ; 142(2): 189-193, 2022.
Article in Japanese | MEDLINE | ID: mdl-35110455

ABSTRACT

We report a rare case of suppurative thrombophlebitis of the posterior neck caused by Streptococcus constellatus. A 69-year-old female patient was admitted to the hospital with neck pain and fever, which had persisted for 16 days prior to hospitalization. On day 1 (day of admission), blood cultures (later identifying S. constellatus) were performed, and ceftriaxone (CTRX) IV (2 g SID) was started. On day 3, suppurative thrombophlebitis of the posterior neck was diagnosed by CT scan. The antimicrobials were changed from CTRX to ampicillin/sulbactam IV (12 g QID) to guard against the possibility of complicated infection with Fusobacterium spp. or Prevotella spp. On day 17, a CT scan revealed that the thrombus remained. Therefore, oral edoxaban (30 mg SID) was started. On day 27, the patient was discharged after her medication was changed to oral amoxicillin/clavulanate (1500 mg/375 mg TID). On day 33, the amoxicillin/clavulanate was changed to oral cefaclor (1500 mg TID) and edoxaban was discontinued due to itching. On day 45, the course of cefaclor was completed. The patient went on to follow an uneventful course with no relapses or complications for two years since the conclusion of treatment. These results suggest that when a patient presents with persistent neck pain accompanied by fever, suppurative thrombophlebitis of the posterior neck should be considered. In antimicrobial therapy, the treatment could be switched from intravenous to oral. In addition, direct-acting oral anticoagulants may be an alternative to other forms of anticoagulants.


Subject(s)
Amoxicillin-Potassium Clavulanate Combination/administration & dosage , Anti-Bacterial Agents/administration & dosage , Cefaclor/administration & dosage , Neck , Streptococcal Infections , Streptococcus constellatus/pathogenicity , Thrombophlebitis/drug therapy , Thrombophlebitis/microbiology , Administration, Oral , Aged , Ampicillin/administration & dosage , Deoxyuridine/administration & dosage , Deoxyuridine/adverse effects , Deoxyuridine/analogs & derivatives , Drug Substitution , Female , Humans , Infusions, Intravenous , Streptococcus constellatus/isolation & purification , Sulbactam/administration & dosage , Suppuration , Thrombophlebitis/diagnosis , Thrombophlebitis/pathology , Treatment Outcome
2.
Histochem Cell Biol ; 157(2): 239-250, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34757474

ABSTRACT

Detection of synthetic thymidine analogues after their incorporation into replicating DNA during the S-phase of the cell cycle is a widely exploited methodology for evaluating proliferative activity, tracing dividing and post-mitotic cells, and determining cell-cycle parameters both in vitro and in vivo. To produce valid quantitative readouts for in vivo experiments with single intraperitoneal delivery of a particular nucleotide, it is necessary to determine the time interval during which a synthetic thymidine analogue can be incorporated into newly synthesized DNA, and the time by which the nucleotide is cleared from the blood serum. To date, using a variety of methods, only the bioavailability time of tritiated thymidine and 5-bromo-2'-deoxyuridine (BrdU) have been evaluated. Recent advances in double- and triple-S-phase labeling using 5-iodo-2'-deoxyuridine (IdU), 5-chloro-2'-deoxyuridine (CldU), and 5-ethynyl-2'-deoxyuridine (EdU) have raised the question of the bioavailability time of these modified nucleotides. Here, we examined their labeling kinetics in vivo and evaluated label clearance from blood serum after single intraperitoneal delivery to mice at doses equimolar to the saturation dose of BrdU (150 mg/kg). We found that under these conditions, all the examined thymidine analogues exhibit similar labeling kinetics and clearance rates from the blood serum. Our results indicate that all thymidine analogues delivered at the indicated doses have similar bioavailability times (approximately 1 h). Our findings are significant for the practical use of multiple S-phase labeling with any combinations of BrdU, IdU, CldU, and EdU and for obtaining valid labeling readouts.


Subject(s)
Bromodeoxyuridine/metabolism , Deoxyuridine/analogs & derivatives , Glyburide/analogs & derivatives , Thymidine/metabolism , Animals , Biological Availability , Bromodeoxyuridine/administration & dosage , Bromodeoxyuridine/blood , Dentate Gyrus/metabolism , Deoxyuridine/administration & dosage , Deoxyuridine/blood , Deoxyuridine/metabolism , Glyburide/administration & dosage , Glyburide/blood , Glyburide/metabolism , Injections, Intraperitoneal , Kinetics , Mice , Mice, Inbred C57BL , Thymidine/administration & dosage , Thymidine/analogs & derivatives
3.
Mol Pharm ; 18(5): 2053-2065, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33886324

ABSTRACT

We fabricated bilirubin-bovine serum albumin (BR-BSA) nanocomplexes as candidates for the delivery of 5-fluoro-2-deoxyuridine (5FUdr) against experimental murine lymphoma. BR was attached to 5FUdr via acid-labile ester bonds mimicking small-molecule drug conjugates. The construct was self-assembled with BSA through strong noncovalent interactions with high drug occupancy in the core and labeled with folic acid (FA) to target cancer cells. The BR-5FUdr-BSA-FA nanoconstruct exhibits excellent biocompatibility, prevents nephrotoxicity, and is tolerated by red blood cells and mononuclear cells. The construct also showed increased accumulation in lymph nodes and tumor cells. BR-5FUdr-BSA-FA caused prolonged growth inhibition and apoptosis, enhanced mitochondrial reactive oxygen species generation, and minimized the viability of parental and doxorubicin-resistant Dalton's lymphoma cells. Treatment of tumor-bearing mice with BR-5FUdr-BSA-FA significantly increased the life span of the animals, improved their histopathological parameters, and downregulated PD-1 expression, suggesting the potential of the construct for 5FUdr delivery to treat lymphoma.


Subject(s)
Deoxyuridine/analogs & derivatives , Drug Carriers/chemistry , Lymphoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bilirubin/chemistry , Biomimetic Materials/chemistry , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Deoxyuridine/administration & dosage , Deoxyuridine/pharmacokinetics , Disease Models, Animal , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma/pathology , Mice , Programmed Cell Death 1 Receptor/metabolism , Serum Albumin, Bovine/chemistry
4.
Aging (Albany NY) ; 12(12): 12268-12284, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516128

ABSTRACT

Caenorhabditis elegans is widely used for aging studies. 5-Fluoro-2´-deoxyuridine (FUdR) is commonly used to control offspring. While larvae are stopped from further development, also mitochondrial DNA and function may be affected. Since mitochondria and longevity are closely related, the use of FUdR may falsify possible studies. PX627, an auxin inducible infertility strain to control offspring, allows mitochondrial investigations during senescence without FUdR toxicity.Longevity and health parameters were assessed in 2- and 10-day old nematodes wild-type N2 and PX627 treated with FUdR or auxin, respectively. Mitochondrial membrane potential, energetic metabolites and reactive oxygen species levels, were determined. mRNA expression levels of key genes involved were quantified using quantitative real-time PCR.FUdR significantly increased lifespan and health parameters, as well as, mitochondrial function compared to untreated controls and auxin treated PX627. Although a decrease in all parameters could be observed in aged nematodes, this was less severe after FUdR exposure. Glycolysis was significantly up-regulated in aged PX627 compared to N2. Expression levels of daf-16, sir-2.1, aak-2, skn-1, atp-2 and atfs-1 were regulated accordingly.Hence, auxin in PX627 might be a good alternative to control progeny, for mitochondrial- and longevity-related investigations in nematodes.


Subject(s)
Aging/drug effects , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/drug effects , Indoleacetic Acids/administration & dosage , Models, Animal , Aging/physiology , Animals , Caenorhabditis elegans/physiology , DNA, Mitochondrial/drug effects , Deoxyuridine/administration & dosage , Deoxyuridine/adverse effects , Deoxyuridine/analogs & derivatives , Fertility/drug effects , Indoleacetic Acids/adverse effects , Longevity/drug effects , Longevity/physiology , Mitochondria/drug effects , Mitochondria/metabolism , Proteolysis/drug effects
5.
Bioconjug Chem ; 30(11): 2958-2966, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31638370

ABSTRACT

In the present study, we used a nucleoside derivative 5-vinyluridine (VrU) for labeling during cell division and for tumor imaging in living mice. We demonstrated that the functional nucleoside bearing a 5-vinyl group is metabolically incorporated into cellular RNA and can be used to image RNA using a Diels-Alder reaction. The reagent allows for simultaneous and clear imaging of DNA and RNA in mammalian cells at single-cell resolution. We extended this approach to observe DNA and RNA behaviors in several basic stages of cell division. We further demonstrated that the derivative can be used for fluorescence imaging of tumor in live mice.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Deoxyuridine/analogs & derivatives , Molecular Imaging/methods , RNA, Neoplasm/metabolism , Animals , Deoxyuridine/administration & dosage , Deoxyuridine/chemistry , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Neoplasm/analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Elife ; 72018 08 02.
Article in English | MEDLINE | ID: mdl-30070633

ABSTRACT

The mechanisms underlying biological aging are becoming recognized as therapeutic targets to delay the onset of multiple age-related morbidities. Even greater health benefits can potentially be achieved by halting or reversing age-associated changes. C. elegans restore their tissues and normal longevity upon exit from prolonged adult reproductive diapause, but the mechanisms underlying this phenomenon remain unknown. Here, we focused on the mechanisms controlling recovery from adult diapause. Here, we show that functional improvement of post-mitotic somatic tissues does not require germline signaling, germline stem cells, or replication of nuclear or mitochondrial DNA. Instead a large expansion of the somatic RNA pool is necessary for restoration of youthful function and longevity. Treating animals with the drug 5-fluoro-2'-deoxyuridine prevents this restoration by blocking reactivation of RNA metabolism. These observations define a critical early step during exit from adult reproductive diapause that is required for somatic rejuvenation of an adult metazoan animal.


Subject(s)
Diapause/genetics , Longevity/genetics , RNA/genetics , Reproduction/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Germ Cells/drug effects , Germ Cells/growth & development , Longevity/drug effects , RNA/drug effects , Reproduction/drug effects , Stem Cells/drug effects
8.
J Immunol Methods ; 457: 82-86, 2018 06.
Article in English | MEDLINE | ID: mdl-29605230

ABSTRACT

The study of organogenesis, tissue-homeostasis and regeneration requires the precise assessment of in vivo cell proliferation. To this end a host of methods have been developed to detect and quantify DNA synthesis in proliferating cells. These include cell labeling with various nucleotide analogues and fluorescence reporter-based animal models with each method presenting its idiosyncratic shortcomings. Quantitative assessment of epithelial cell turnover has been partly hampered due to their variable and limited in vivo accessibility and the requirement for harsher isolation procedures to procure single cells for FACS analysis. Here, we report a reliable protocol to study in vivo cell proliferation of epithelial cells in mice by repeatedly injecting EdU intravenously for an extended 12-day period. EdU incorporation was quantitated ex vivo by FACS after tissue dissociation in order to obtain single epithelial cell suspensions. As a lead population, we analyzed thymic epithelial cells (TECs), where we were able to label compartmentalized TEC subsets to saturation without apparent toxic effects on the thymus architecture or stress-sensitive TEC lineage differentiation. The data is in concordance with the prevailing model of medullary TEC terminal differentiation that includes the post-Aire stage. The same protocol was successfully applied to epithelial cells of various other organs - skin, lymph node, kidney and small intestine - tissues with widely varying frequencies and rates of proliferating epithelial cells.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells/cytology , Staining and Labeling/methods , Administration, Intravenous , Animals , Cell Count , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Flow Cytometry , Mice , Mice, Inbred C57BL , Thymus Gland/cytology
9.
J Surg Oncol ; 117(4): 634-643, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29165816

ABSTRACT

BACKGROUND/OBJECTIVES: Combination hepatic artery infusion (HAI) and systemic (SYS) chemotherapy for unresectable CRLM results in high tumor-response rates. This study represents an update of long-term survival and conversion to resectability in patients with unresectable CRLM treated with HAI and SYS chemotherapy in a phase II study. METHOD: The primary endpoint was complete resection. Multivariate and landmark analysis assessed the effect of complete resection on progression-free (PFS) and overall survival (OS). RESULTS: From 2007 to 2012, 64 patients with median of 13 tumors were enrolled; 67% had prior chemotherapy. 33 patients (52%) were converted to resection. Median follow-up among survivors was 81 months. Median PFS and OS were 13 and 38 months, respectively, with 5-year-OS of 36%. Chemotherapy-naïve patients had 5-year-OS of 51%. Conversion to resection was the only independent factor prognostic of improved PFS and OS. Nine of 64 patients (14%) are NED (five since initial resection, three after resection of recurrent disease, one from chemotherapy alone) at median follow-up of 86 months from treatment initiation, and 72 months from last operative intervention. CONCLUSION: Combination HAI and SYS is an effective therapy for high-volume unresectable CRLM, resulting in a high rate of resection, long-term survival, and the potential for cure.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Bevacizumab/administration & dosage , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Colorectal Neoplasms/surgery , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Disease-Free Survival , Female , Fluorouracil/administration & dosage , Hepatic Artery , Humans , Infusions, Intra-Arterial , Irinotecan , Leucovorin/administration & dosage , Liver Neoplasms/surgery , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prospective Studies , Survival Rate , Treatment Outcome
10.
J Med Chem ; 59(8): 3661-70, 2016 04 28.
Article in English | MEDLINE | ID: mdl-27022837

ABSTRACT

We report a series of novel O-(substituted benzyl) phosphoramidate prodrugs of 5-fluoro-2'-deoxyuridine for the treatment of hepatocellular carcinoma. Through structure optimization, the o-methylbenzyl analog (1t) was identified as an orally bioavailable and liver-targeted lead compound. This lead prodrug is well-tolerated at a dose up to 3 g/kg in Kuming mice via oral administration. An efficacy study demonstrated that it possesses good inhibitory effect (61.67% and 72.50%, respectively) on tumor growth in a mouse xenograft model. A metabolism study in Sprague-Dawley rats suggested that 1t can release the desired 5'-monophosphate in the liver with high liver-targeting index.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Deoxyuridine/analogs & derivatives , Liver Neoplasms, Experimental/drug therapy , Liver/drug effects , Prodrugs/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Area Under Curve , Deoxyuridine/administration & dosage , Deoxyuridine/pharmacokinetics , Deoxyuridine/pharmacology , Deoxyuridine/therapeutic use , Drug Discovery , Mice , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/therapeutic use , Rats , Rats, Sprague-Dawley , Tissue Distribution
11.
Invest New Drugs ; 33(6): 1206-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26377590

ABSTRACT

BACKGROUND: This Phase-I-study aimed to determine the recommended Phase-II-dosing-schedule of LY2334737, an oral gemcitabine prodrug, in patients with advanced/metastatic solid tumors. Pharmacokinetics, cytokeratin-18 (CK18) levels, genetic polymorphisms, and antitumor activity were additionally evaluated. METHODS: Patients received escalating doses of LY2334737 either every other day for 21 days (d) followed by 7 days-drug-free period (QoD-arm) or once daily for 7 days every other week (QD-arm). The 28 days-cycles were repeated until disease progression or unacceptable toxicity. Standard 3 + 3 dose-escalation was succeeded by a dose-confirmation phase (12 additional patients to be enrolled on the maximum tolerated dose [MTD]). RESULTS: Forty-one patients received QoD- (40-100 mg) and 32 QD-dosing (40-90 mg). On QoD, 3/9 patients experienced dose-limiting toxicities (DLTs) on the 100 mg dose (2 × G3 diarrhea, 1 × G3 transaminase increase); 1 additional DLT (G3 diarrhea) occurred during dose confirmation at 90 mg (12 patients). On QD, 1 patient each experienced DLTs on 60 mg (G3 transaminase increase) and 80 mg (G3 prolonged QTcF-interval); 2/7 patients had 3 DLTs on the 90 mg dose (diarrhea, edema, liver-failure; all G3). The MTD was established at 90 mg for the QoD-arm. Seven patients on QoD and 4 on QD achieved SD (no CR + PR). Pharmacokinetics showed a dose-proportional increase in exposure of LY2334737 and dFdC without accumulation after repeated dosing. Significant increases in CK18 levels were observed. Genetic polymorphism of the cytidine deaminase gene (rs818202) could be associated with ≥ G3 hepatotoxicity. CONCLUSIONS: Both schedules displayed linear pharmacokinetics and acceptable safety profiles. The recommended dose and schedule of LY2334737 for subsequent Phase-II-studies is 90 mg given QoD for 21 day.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxyuridine/analogs & derivatives , Neoplasms/drug therapy , Prodrugs/administration & dosage , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Deoxyuridine/administration & dosage , Deoxyuridine/pharmacokinetics , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Neoplasms/diagnosis , Neoplasms/metabolism , Prodrugs/pharmacokinetics , Gemcitabine
12.
Oncol Rep ; 34(3): 1487-93, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134602

ABSTRACT

Heterologous expression of the Drosophila melanogaster multi-substrate deoxyribonucleoside kinase (Dm-dNK) increases the sensitivity of cancer cells to several cytotoxic nucleoside analogs. Thus, it may be used as a suicide gene in combined gene/chemotherapy treatment of cancer. To further characterize this potential suicide gene, we constructed two retroviral vectors that enabled the expression of Dm-dNK in cancer cells. One vector harbored the wild­type enzyme that localized to the nucleus. The other vector harbored a mitochondrial localized mutant enzyme that was constructed by deleting the nuclear localization signal and fusing it to a mitochondrial import signal of cytochrome c oxidase. A thymidine kinase-deficient osteosarcoma cell line was transduced with the recombinant viruses. The sensitivity and bystander cell killing in the presence of pyrimidine nucleoside analogs (E)-5-(2-bromovinyl)­2'­deoxyuridine and 1-ß-D-arabinofuranosylthymine were investigated. Tanshinone IIA is a constituent of Danshen; a traditional Chinese medicine used in the treatment of cardiovascular diseases. This study also looked at the influence of Tanshinone IIA on the bystander effect and the underlying mechanisms. We showed that sensitivity of the osteosarcoma cell line to the nucleoside analogs and the efficiency of bystander cell killing were independent of the subcellular localization of Dm-dNK. The enhanced effect of tanshinone IIA on the bystander effect was related to the increased expression of Cx43 and Cx26.


Subject(s)
Abietanes/administration & dosage , Genetic Therapy , Osteosarcoma/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Bystander Effect , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Connexin 26 , Connexins , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Galactose/administration & dosage , Galactose/analogs & derivatives , Genetic Vectors , Humans , Mitochondria/drug effects , Mitochondria/genetics , Nuclear Localization Signals/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Retroviridae/genetics , Thymine/administration & dosage , Thymine/analogs & derivatives
13.
Am J Clin Nutr ; 101(4): 860-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25833982

ABSTRACT

BACKGROUND: Folic acid prevents neural tube closure defects (NTDs), but the causal metabolic pathways have not been established. Serine hydroxymethyltransferase 1 (SHMT1) is an essential scaffold protein in folate-dependent de novo thymidylate synthesis in the nucleus. SHMT1-deficient mice provide a model to investigate folic acid-responsive NTDs wherein disruption of de novo thymidylate synthesis impairs neural tube closure. OBJECTIVE: We examined the effects of maternal supplementation with the pyrimidine nucleosides uridine, thymidine, or deoxyuridine with and without folate deficiency on NTD incidence in the Shmt1 mouse model. DESIGN: Shmt1(+/+) and Shmt1(-/-) female mice fed folate-replete or folate-deficient diets and supplemented with uridine, thymidine, or deoxyuridine were bred, and litters (n = 10-23 per group) were examined for the presence of NTDs. Biomarkers of impaired folate status and metabolism were measured, including plasma nucleosides, hepatic uracil content, maternal plasma folate concentrations, and incorporation of nucleoside precursors into DNA. RESULTS: Shmt1(+/-) and Shmt1(-/-) embryos from dams fed the folate-deficient diet were susceptible to NTDs. No NTDs were observed in litters from dams fed the folate-deficient diet supplemented with deoxyuridine. Surprisingly, uridine supplementation increased NTD incidence, independent of embryo genotype and dietary folic acid. These dietary nucleosides did not affect maternal hepatic uracil accumulation in DNA but did affect plasma folate concentrations. CONCLUSIONS: Maternal deoxyuridine supplementation prevented NTDs in dams fed the folate-deficient diet, whereas maternal uridine supplementation increased NTD incidence, independent of folate and embryo genotype. These findings provide new insights into the metabolic impairments and mechanisms of folate-responsive NTDs resulting from decreased Shmt1 expression.


Subject(s)
Deoxyuridine/administration & dosage , Folic Acid/administration & dosage , Neural Tube Defects/drug therapy , Uridine/administration & dosage , Uridine/adverse effects , Animals , Deoxyuridine/blood , Disease Models, Animal , Female , Folic Acid/blood , Folic Acid Deficiency/drug therapy , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , HeLa Cells , Humans , Maternal Nutritional Physiological Phenomena , Mice , Neural Tube/drug effects , Neural Tube Defects/blood , Neural Tube Defects/etiology , Pregnancy , Thymidine/administration & dosage , Thymidine/adverse effects , Thymidine/blood , Uracil/metabolism , Uridine/blood
14.
Org Biomol Chem ; 13(15): 4589-95, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25777799

ABSTRACT

The representative DNA-labeling agent 5-ethynyl-2'-deoxyuridine (EdU) was chemically modified to improve its function. Chemical monophosphorylation was expected to enhance the efficiency of the substrate in DNA polymerization by circumventing the enzymatic monophosphorylation step that consumes energy. In addition, to enhance cell permeability, the phosphates were protected with bis-pivaloyloxymethyl that is stable in buffer and plasma, and degradable inside various cell types. The phosphorylated EdU (PEdU) was less toxic than EdU, and had the same or a slightly higher DNA-labeling ability in vitro. PEdU was also successfully applied to DNA labeling in vivo. In conclusion, PEdU can be used as a less toxic DNA-labeling agent for studies that require long-term cell survival or very sensitive cell lines.


Subject(s)
DNA/analysis , Deoxyuridine/analogs & derivatives , 3T3 Cells , Animals , DNA/metabolism , Deoxyuridine/administration & dosage , Deoxyuridine/chemistry , Deoxyuridine/metabolism , HeLa Cells , Humans , Mice , Phosphorylation , Staining and Labeling/methods
15.
Invest New Drugs ; 33(2): 432-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640850

ABSTRACT

Background This Phase 1b study aimed to determine the recommended Phase 2 dose of LY2334737, an oral pro-drug of gemcitabine, in combination with capecitabine, an oral pro-drug of 5-fluorouracil, in patients with advanced solid tumors. In addition, pharmacokinetics (PK) and tumor response were evaluated. Patients and methods Patients with advanced/metastatic solid tumors received 650 mg/m(2) capecitabine twice daily (BID) and escalating doses of LY2334737 once daily (QD; initial dose 10 mg/day), both for 14 days followed by 7-day drug holiday. Cycles were repeated until progressive disease (PD) or unacceptable toxicity. Results Fifteen patients received a median of 2 (range 1-7) treatment cycles; 14 patients discontinued due to PD, 1 due to toxicity (pyrexia). LY2334737 doses up to 40 mg/day were explored. Three dose-limiting toxicities were reported by 2 patients (fatigue, diarrhea, hyponatremia; all Grade 3). Seven patients achieved stable disease. Enrollment was stopped after unexpected hepatic toxicities were observed with LY2334737 QD in a study of Japanese patients. PK parameters for LY2334737 were consistent with the first-in-human study of LY2334737; PK data after 14 day combination treatment revealed no drug-drug interactions between LY2334737 and capecitabine. Conclusions No drug interactions or unexpected toxicities were observed in US patients when LY2334737 at doses up to 40 mg/day was administered QD in combination with capecitabine BID; the maximum tolerated dose was not reached.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Deoxyuridine/analogs & derivatives , Neoplasms/drug therapy , Prodrugs/pharmacokinetics , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols , Capecitabine/therapeutic use , Deoxyuridine/administration & dosage , Deoxyuridine/adverse effects , Deoxyuridine/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Prodrugs/administration & dosage , Prodrugs/adverse effects
16.
Oncotarget ; 5(14): 5483-93, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-24978848

ABSTRACT

The existence of therapy resistant glioma stem cells is responsible for the high recurrence rate and incurability of glioblastomas. The Hedgehog pathway activity plays an essential role for self-renewal capacity and survival of glioma stem cells. We examined the potential of the Sonic hedgehog ligand for sensitizing of glioma stem cells to endogenous nano-irradiation. We demonstrate that the Sonic hedgehog ligand preferentially and efficiently activated glioma stem cells to enter the radiation sensitive G2/M phase. Concomitant inhibition of de novo thymidine synthesis with fluorodeoxyuridine and treatment with the Auger electron emitting thymidine analogue 5-[I-125]-Iodo-4'-thio-2'-deoxyuridine ([I-125]ITdU) leads to a fatal nano-irradiation in sensitized glioma stem cells. Targeting of proliferating glioma stem cells with DNA-incorporated [I-125]ITdU efficiently invokes the intrinsic apoptotic pathway despite active DNA repair mechanisms. Further, [I-125]ITdU completely inhibits survival of glioma stem cells in vitro. Analysis of non-stem glioblastoma cells and normal human astrocytes reveals that glioma stem cells differentially respond to Sonic hedgehog ligand. These data demonstrate a highly efficient and controllable single-cell kill therapeutic model for targeting glioma stem cells.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/radiation effects , Apoptosis/radiation effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/radiation effects , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Glioblastoma/pathology , Humans , Iodine Radioisotopes/administration & dosage , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation Tolerance , Radiopharmaceuticals/administration & dosage , Signal Transduction/radiation effects
17.
J Vis Exp ; (87)2014 May 07.
Article in English | MEDLINE | ID: mdl-24837791

ABSTRACT

Neurons of the cerebral cortex are generated during brain development from different types of neural stem and progenitor cells (NSPC), which form a pseudostratified epithelium lining the lateral ventricles of the embryonic brain. Genotoxic stresses, such as ionizing radiation, have highly deleterious effects on the developing brain related to the high sensitivity of NSPC. Elucidation of the cellular and molecular mechanisms involved depends on the characterization of the DNA damage response of these particular types of cells, which requires an accurate method to determine NSPC progression through the cell cycle in the damaged tissue. Here is shown a method based on successive intraperitoneal injections of EdU and BrdU in pregnant mice and further detection of these two thymidine analogues in coronal sections of the embryonic brain. EdU and BrdU are both incorporated in DNA of replicating cells during S phase and are detected by two different techniques (azide or a specific antibody, respectively), which facilitate their simultaneous detection. EdU and BrdU staining are then determined for each NSPC nucleus in function of its distance from the ventricular margin in a standard region of the dorsal telencephalon. Thus this dual labeling technique allows distinguishing cells that progressed through the cell cycle from those that have activated a cell cycle checkpoint leading to cell cycle arrest in response to DNA damage. An example of experiment is presented, in which EdU was injected before irradiation and BrdU immediately after and analyzes performed within the 4 hr following irradiation. This protocol provides an accurate analysis of the acute DNA damage response of NSPC in function of the phase of the cell cycle at which they have been irradiated. This method is easily transposable to many other systems in order to determine the impact of a particular treatment on cell cycle progression in living tissues.


Subject(s)
Brain/embryology , Bromodeoxyuridine/chemistry , DNA Damage , Deoxyuridine/analogs & derivatives , Microscopy, Fluorescence/methods , Neural Stem Cells/physiology , Stem Cells/physiology , Animals , Brain/cytology , Brain/radiation effects , Bromodeoxyuridine/administration & dosage , Cell Cycle/genetics , Cell Cycle/radiation effects , Deoxyuridine/administration & dosage , Deoxyuridine/chemistry , Female , G1 Phase Cell Cycle Checkpoints/genetics , G1 Phase Cell Cycle Checkpoints/radiation effects , Mice , Neural Stem Cells/cytology , Pregnancy , S Phase Cell Cycle Checkpoints/genetics , S Phase Cell Cycle Checkpoints/radiation effects , Stem Cells/cytology , Stem Cells/radiation effects , Whole-Body Irradiation
18.
Cancer Chemother Pharmacol ; 73(6): 1205-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24744161

ABSTRACT

PURPOSE: This Phase 1 study aimed to determine the recommended Phase 2 dose of LY2334737, an oral gemcitabine prodrug, when combined with standard dose docetaxel treatment in patients with advanced solid tumors. Pharmacokinetics (PK) and antitumor activity were additionally evaluated. METHODS: Patients with advanced/metastatic solid tumors received escalating doses of LY2334737 once daily (QD) for 14 days, followed by a 7-day drug-free period. Docetaxel was given at 75 mg/m(2) every 3 weeks (q3w). Cycles were repeated until progressive disease (PD) or unacceptable toxicity. RESULTS: Of 22 patients recruited, all Caucasian, 7 received an LY2334737 dose of 10 mg/day, 10 received 20 mg/day, 5 received 30 mg/day. Nineteen patients discontinued due to PD, 2 due to adverse events, 1 due to investigator decision. Dose-limiting toxicities: 2× febrile neutropenia (G3), 2× fatigue (1× G2, 1× G3), 1× neutropenia (G4). The maximum tolerated dose (MTD) was identified to be 10 mg/day. Two patients achieved partial response, 10 patients stable disease. Enrollment was stopped after unexpected hepatic toxicities were observed with LY2334737 QD for 14 days per cycle in another study of Japanese patients. PK data were consistent with the first-in-man study of LY2334737 and did not reveal any drug-drug interaction between LY2334737 and docetaxel. CONCLUSIONS: Combination of LY2334737 at doses up to 30 mg/day QD for 14 days per cycle with docetaxel 75 mg/m(2) q3w resulted in an undesirable toxicity profile and a low MTD of 10 mg/day. Alternative treatment schedules of LY2334737 should be explored.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Neoplasms/drug therapy , Neoplasms/metabolism , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Deoxyuridine/administration & dosage , Deoxyuridine/adverse effects , Deoxyuridine/analogs & derivatives , Deoxyuridine/pharmacokinetics , Docetaxel , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Neoplasms/pathology , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Taxoids/administration & dosage , Taxoids/adverse effects , Taxoids/pharmacokinetics , Treatment Outcome , Gemcitabine
19.
Neurosci Lett ; 563: 90-5, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24486842

ABSTRACT

Olfactory ensheathing cells (OECs) play an important role in the continuous regeneration of the primary olfactory nervous system throughout life and for regeneration of olfactory neurons after injury. While it is known that several individual OEC subpopulations with distinct properties exist in different anatomical locations, it remains unclear how these different subpopulations respond to a major injury. We have examined the proliferation of OECs from one distinct location, the peripheral accessory olfactory nervous system, following large-scale injury (bulbectomy) in mice. We used crosses of two transgenic reporter mouse lines, S100ß-DsRed and OMP-ZsGreen, to visualise OECs, and main/accessory olfactory neurons, respectively. We surgically removed one olfactory bulb including the accessory olfactory bulb to induce degeneration, and found that accessory OECs in the nerve bundles that terminate in the accessory olfactory bulb responded by increased proliferation with a peak occurring 2 days after the injury. To label proliferating cells we used the thymidine analogue ethynyl deoxyuridine (EdU) using intranasal delivery instead of intraperitoneal injection. We compared and quantified the number of proliferating cells at different regions at one and four days after EdU labelling by the two different methods and found that intranasal delivery method was as effective as intraperitoneal injection. We demonstrated that accessory OECs actively respond to widespread degeneration of accessory olfactory axons by proliferating. These results have important implications for selecting the source of OECs for neural regeneration therapies and show that intranasal delivery of EdU is an efficient and reliable method for assessing proliferation of olfactory glia.


Subject(s)
Deoxyuridine/analogs & derivatives , Neuroglia/pathology , Olfactory Bulb/pathology , Administration, Intranasal , Animals , Cell Proliferation , Deoxyuridine/administration & dosage , Indicators and Reagents , Mice, Transgenic , Olfactory Bulb/injuries , Staining and Labeling
20.
Mol Cancer Ther ; 13(1): 144-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24174494

ABSTRACT

In multiple myeloma, the presence of highly resistant cancer stem cells (CSC) that are responsible for tumor metastasis and relapse has been proven. Evidently, for achieving complete response, new therapeutic paradigms that effectively eradicate both, CSCs and bulk cancer populations, need to be developed. For achieving that goal, an innovative two-step treatment combining targeting of thymidine de novo synthesis pathway and a nanoirradiation by the Auger electron emitting thymidine analogue (123/125)I-5-iodo-4'-thio-2'-deoxyuridine ((123/125)I-ITdU) could be a promising approach. The pretreatment with thymidylate synthase inhibitor 5-fluoro-2'-deoxyuridine (FdUrd, 1 µmol/L for 1 hour) efficiently induced proliferation and terminal differentiation of isolated myeloma stem-like cells. Moreover, FdUrd stimulation led to a decreased activity of a functional CSC marker, aldehyde dehydrogenase (ALDH). The metabolic conditioning by FdUrd emerged to be essential for enhanced incorporation of (125)I-ITdU (incubation with 50 kBq/2 × 10(4) cells for 4 days) and, consequently, for the induction of irreparable DNA damage. (125)I-ITdU showed a pronounced antimyeloma effect on isolated tumor stem-like cells. More than 85% of the treated cells were apoptotic, despite activation of DNA repair mechanisms. Most important, exposure of metabolically conditioned cells to (125)I-ITdU resulted in a complete inhibition of clonogenic recovery. This is the first report showing that pretreatment with FdUrd sensitizes the stem-like cell compartment in multiple myeloma to apoptosis induced by (125)I-ITdU-mediated nanoirradiation of DNA.


Subject(s)
Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplastic Stem Cells/drug effects , Thymidylate Synthase/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , DNA Damage/drug effects , Deoxyuridine/administration & dosage , Deoxyuridine/analogs & derivatives , Enzyme Inhibitors/administration & dosage , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Thymidylate Synthase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...