Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Med Oncol ; 40(8): 213, 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37358701

ABSTRACT

Cancer cells require continuous synthesis of nucleotides for their uncontrolled proliferation. Deoxy thymidylate kinase (DTYMK) belongs to the thymidylate kinase family and is concerned with pyrimidine metabolism. DTYMK catalyzes the ATP-based conversion of deoxy-TMP to deoxy-TDP in both de novo and salvage pathways. Different studies demonstrated that DTYMK was increased in various types of cancers such as hepatocellular carcinoma, colon cancer, lung cancer, etc. Increased level of DTYMK was associated with poorer survival and prognosis, stage, grade and size of tumor, cell proliferation, colony formation, enhanced sensitivity to chemotherapy drugs, migration. Some studies were showed that knockdown of DTYMK reduced the signaling pathway of PI3K/AKT and downregulated expression of CART, MAPKAPK2, AKT1 and NRF1. Moreover, some microRNAs could suppress DTYMK expressions. On the other hand based on the TIMER database, the infiltration of macrophages, dendritic cells, neutrophils, B cells, CD4+ T cell and CD8+ T cell is affected by DTYMK. In the present review, we describe the genomic location, protein structure and isoforms of DTYMK and focus on its role in cancer development.


Subject(s)
Lung Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/therapeutic use , Lung Neoplasms/pathology , Signal Transduction
2.
Crit Rev Eukaryot Gene Expr ; 26(1): 11-7, 2016.
Article in English | MEDLINE | ID: mdl-27278881

ABSTRACT

Molecular targeted therapy is an important, novel approach in the treatment of cancer because it interferes with certain molecules involved in carcinogenesis and tumor growth. Examples include monoclonal antibodies, microvesicles, and suicide genes. Several studies have focused on targeted therapies in prostate cancer, which is a serious cause of cancer death in men. We hypothesize that antibody-coated microvesicles can deliver thymidylate kinase, a suicide protein, to prostate cancer cells, potentiating them to death following azidothymidine (AZT) treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Molecular Targeted Therapy/methods , Nucleoside-Phosphate Kinase/administration & dosage , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , Male , Nucleoside-Phosphate Kinase/pharmacology , Nucleoside-Phosphate Kinase/therapeutic use
3.
Methods Mol Biol ; 1317: 55-67, 2015.
Article in English | MEDLINE | ID: mdl-26072401

ABSTRACT

Suicide gene therapy of cancer (SGTC) entails the introduction of a cDNA sequence into tumor cells whose polypeptide product is capable of either directly activating apoptotic pathways itself or facilitating the activation of pharmacologic agents that do so. The latter class of SGTC approaches is of the greater utility in cancer therapy owing to the ability of some small, activated cytotoxic compounds to diffuse from their site of activation into neighboring malignant cells, where they can also mediate destruction. This phenomenon, termed "bystander killing", can be highly advantageous in driving significant tumor regression in vivo without the requirement of transduction of each and every tumor cell with the suicide gene. We have developed a robust suicide gene therapy enzyme/prodrug system based on an engineered variant of the human thymidylate kinase (TMPK), which has been endowed with the ability to drive azidothymidine (AZT) activation. Delivery of this suicide gene sequence into tumors by means of recombinant lentivirus-mediated transduction embodies an SGTC strategy that successfully employs bystander cell killing as a mechanism to achieve significant ablation of solid tumors in vivo. Thus, this engineered TMPK/AZT suicide gene therapy axis holds great promise for clinical application in the treatment of inoperable solid tumors in the neoadjuvant setting. Here we present detailed procedures for the preparation of recombinant TMPK-based lentivirus, transduction of target cells, and various approaches for the evaluation of bystander cell killing effects in SGCT in both in vitro and in vivo models.


Subject(s)
Bystander Effect/drug effects , Genes, Transgenic, Suicide , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Nucleoside-Phosphate Kinase/genetics , Prodrugs/pharmacology , Zidovudine/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation , Colorimetry , Gene Expression , Genetic Engineering , HEK293 Cells , Humans , Lentivirus/metabolism , Male , Mice, Inbred NOD , Mice, SCID , Nucleoside-Phosphate Kinase/therapeutic use , Recombinant Proteins/metabolism , Transduction, Genetic , Transgenes
4.
Neoplasia ; 11(7): 637-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19568409

ABSTRACT

Gemcitabine is a first-line agent for advanced pancreatic cancer therapy. However, its efficacy is often limited by its poor intracellular metabolism and chemoresistance. To exert its antitumor activity, gemcitabine requires to be converted to its active triphosphate form. Thus, our aim was to improve gemcitabine activation using gene-directed enzyme prodrug therapy based on gemcitabine association with the deoxycytidine kinase::uridine monophosphate kinase fusion gene (dCK::UMK) and small interference RNA directed against ribonucleotide reductase (RRM2) and thymidylate synthase (TS). In vitro, cytotoxicity was assessed by 3-[4,5-dimethylthiazol-2-yl]-3,5-diphenyl tetrazolium bromide and [(3)H]thymidine assays. Apoptosis-related gene expression and activity were analyzed by reverse transcription-polymerase chain reaction, Western blot, and ELISA. For in vivo studies, the treatment efficacy was evaluated on subcutaneous and orthotopic pancreatic tumor models. Our data indicated that cell exposure to gemcitabine induced a down-regulation of dCK expression and up-regulation of TS and RR expression in Panc1-resistant cells when compared with BxPc3- and HA-hpc2-sensitive cells. The combination of TS/RRM2 small interference RNA with Ad-dCK::UMK induced a 40-fold decrease of gemcitabine IC(50) in Panc1 cells. This strong sensitization was associated to apoptosis induction with a remarkable increase in TRAIL expression and a diminution of gemcitabine-induced nuclear factor-kappaB activity. In vivo, the gemcitabine-based tritherapy strongly reduced tumor volumes and significantly prolonged mice survival. Moreover, we observed an obvious increase of apoptosis and decrease of cell proliferation in tumors receiving the tritherapy regimens. Together, these findings suggest that simultaneous TS/RRM2-gene silencing and dCK::UMK gene overexpression markedly improved gemcitabine's therapeutic activity. Clearly, this combined strategy warrants further investigation.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Deoxycytidine/analogs & derivatives , Genetic Therapy/methods , Pancreatic Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Adenoviridae/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine Kinase/genetics , Deoxycytidine Kinase/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression , Gene Expression Profiling , Genetic Vectors , Humans , Mice , Mice, Nude , Nucleoside-Phosphate Kinase/genetics , Nucleoside-Phosphate Kinase/therapeutic use , Prodrugs/administration & dosage , RNA Interference , RNA, Small Interfering , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase/genetics , Thymidylate Synthase/genetics , Xenograft Model Antitumor Assays , Gemcitabine
5.
Chembiochem ; 4(2-3): 143-6, 2003 Mar 03.
Article in English | MEDLINE | ID: mdl-12616626

ABSTRACT

The use of nucleoside analogues in anticancer and antiviral treatments is often impaired by the slow intracellular activation of these drugs. This problem can be addressed by the modulation of rate-limiting enzymes in the activation pathways of the nucleoside analogues. Therapeutic strategies based on the combination of optimized activating enzymes and established nucleoside drugs promise significant improvements to traditional chemotherapy.


Subject(s)
Nucleosides/chemical synthesis , Nucleosides/therapeutic use , Phosphotransferases/chemical synthesis , Prodrugs/chemical synthesis , Acquired Immunodeficiency Syndrome/drug therapy , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Neoplasms/drug therapy , Nucleoside-Phosphate Kinase/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Protein Engineering/trends , Structure-Activity Relationship , Zidovudine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...