Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancers (Basel) ; 16(4)2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38398204

ABSTRACT

From the perspective of health economics, the evaluation of drug-related cost effectiveness and clinical utility is crucial. We conducted a cost-utility analysis of two first-line drugs, tenofovir alafenamide (TAF) and entecavir (ETV), in the treatment of chronic hepatitis B (CHB) patients. We performed inverse probability of treatment weighting (IPTW) to match the independent variables between the two treatment groups. The incremental cost effectiveness ratio (ICER) of the two treatment groups was simulated using a decision tree with the Markov annual-cycle model. A total of 54 patients treated with TAF and 98 with ETV from January 2016 to December 2020 were enrolled. The total medical cost in the TAF group was NT$76,098 less than that in the ETV group, and TAF demonstrated more effectiveness than ETV by 3.19 quality-adjusted life years (QALYs). When the time horizon was set at 30 years, the ICER of the TAF group compared with the ETV group was -NT$23,878 per QALY, suggesting more cost savings for TAF. Additionally, with the application of TAF, over NT$366 million (approximately US$12 million) can be saved annually. TAF demonstrates cheaper medical costs and more favorable clinical QALYs than ETV. To balance health insurance benefits and cost effectiveness, TAF is the optimal treatment for CHB.

4.
11.
Int J Surg ; 109(8): 2527-2528, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37195788
12.
Mol Pharmacol ; 87(6): 996-1005, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25814515

ABSTRACT

COH29 [N-(4-(3,4-dihydroxyphenyl)-5-phenylthiazol-2-yl)-3,4-dihydroxybenzamide], a novel antimetabolite drug developed at City of Hope Cancer Center, has anticancer activity that stems primarily from the inhibition of human ribonucleotide reductase (RNR). This key enzyme in deoxyribonucleotide biosynthesis is the target of established clinical agents such as hydroxyurea and gemcitabine because of its critical role in DNA replication and repair. Herein we report that BRCA-1-defective human breast cancer cells are more sensitive than wild-type BRCA-1 counterparts to COH29 in vitro and in vivo. Microarray gene expression profiling showed that COH29 reduces the expression of DNA repair pathway genes, suggesting that COH29 interferes with these pathways. It is well established that BRCA1 plays a role in DNA damage repair, especially homologous recombination (HR) repair, to maintain genome integrity. In BRCA1-defective HCC1937 breast cancer cells, COH29 induced more double-strand breaks (DSBs) and DNA-damage response than in HCC1937 + BRCA1 cells. By EJ5- and DR-green fluorescent protein (GFP) reporter assay, we found that COH29 could inhibit nonhomologous end joining (NHEJ) efficiency and that no HR activity was detected in HCC1937 cells, suggesting that repression of the NHEJ repair pathway may be involved in COH29-induced DSBs in BRCA1-deficient HCC1937 cells. Furthermore, we observed an accumulation of nuclear Rad51 foci in COH29-treated HCC1937 + BRCA1 cells, suggesting that BRCA1 plays a crucial role in repairing and recovering drug-induced DNA damage by recruiting Rad51 to damage sites. In summary, we describe here additional biologic effects of the RNR inhibitor COH29 that potentially strengthen its use as an anticancer agent.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Benzamides/pharmacology , DNA Repair/drug effects , Ribonucleotide Reductases/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Antimetabolites, Antineoplastic/therapeutic use , BRCA1 Protein/genetics , Benzamides/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA End-Joining Repair/drug effects , Female , Heterografts , Humans , Mice, Inbred NOD , Mutagenicity Tests , Neoplasm Transplantation , Thiazoles/therapeutic use , Zebrafish
13.
Cancer Res ; 73(4): 1298-307, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23393200

ABSTRACT

Fibroblast growth factor (FGF) receptor (FGFR) substrate 2 (FRS2) is an adaptor protein that plays a critical role in FGFR signaling. FRS2 is located on chromosome 12q13-15 that is frequently amplified in liposarcomas. The significance of FRS2 and FGFR signaling in high-grade liposarcomas is unknown. Herein, we first comparatively examined the amplification and expression of FRS2 with CDK4 and MDM2 in dedifferentiated liposarcoma (DDLS) and undifferentiated high-grade pleomorphic sarcoma (UHGPS). Amplification and expression of the three genes were identified in 90% to 100% (9-11 of 11) of DDLS, whereas that of FRS2, CDK4, and MDM2 were observed in 55% (41 of 75), 48% (36 of 75), and 44% (33/75) of clinically diagnosed UHGPS, suggesting that these "UHGPS" may represent DDLS despite lacking histologic evidence of lipoblasts. Immunohistochemical analysis of phosphorylated FRS2 protein indicated that the FGFR/FRS2 signaling axis was generally activated in about 75% of FRS2-positive high-grade liposarcomas. Moreover, we found that FRS2 and FGFRs proteins are highly expressed and functional in three high-grade liposarcoma cell lines: FU-DDLS-1, LiSa-2, and SW872. Importantly, the FGFR selective inhibitor NVP-BGJ-398 significantly inhibited the growth of FU-DDLS-1 and LiSa-2 cells with a concomitant suppression of FGFR signal transduction. Attenuation of FRS2 protein in FU-DDLS-1 and LiSa-2 cell lines decreased the phosphorylated extracellular signal-regulated kinase 1/2 and AKT and repressed cell proliferation. These findings indicate that analysis of FRS2 in combination with CDK4 and MDM2 will more accurately characterize pathologic features of high-grade liposarcomas. Activated FGFR/FRS2 signaling may play a functional role in the development of high-grade liposarcomas, therefore, serve as a potential therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Liposarcoma/genetics , Membrane Proteins/genetics , Receptors, Fibroblast Growth Factor/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Arginine/analogs & derivatives , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Liposarcoma/metabolism , Liposarcoma/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Neoplasm Grading , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrimidines/pharmacology , RNA Interference , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
Anticancer Res ; 32(11): 4685-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23155230

ABSTRACT

BACKGROUND: Ribonucleotide reductase composed of the hRRM1 and hRRM2 subunits catalyzes the conversion of ribonucleotides to their corresponding deoxy forms for DNA replication. Anti-hRRM2 siRNA degrades hRRM2's mRNA and suppresses tumorigenesis. A Phase I clinical trial demonstrated its therapy potential. HN-1 represents a tumor-specifically internalizing peptide for targeted-drug delivery into human head and neck squamous cell carcinoma. MATERIALS AND METHODS: Internalization of peptide was monitored by fluorescence microscopy. The peptide-siRNA conjugate was chemically synthesized. The hRRM2 expression was monitored by western blot analysis. RESULTS: HN-1(TYR) (HN-1 with two N-terminally added tyrosines) was internalized by human head and neck or breast cancer cells. Anti-hRRM2 siRNA(R) (resistant to RNase degradation) was conjugated to HN-1(TYR) without compromising their properties. The treatment with HN-1(TYR)-anti-hRRM2 siRNA(R) partly suppressed the endogenously expressed hRRM2 in human breast cancer cells. CONCLUSION: Our results establish the utility of tumor-specifically internalizing peptides for targeted siRNA delivery into human cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Drug Delivery Systems/methods , Genetic Therapy/methods , Head and Neck Neoplasms/metabolism , Oligopeptides/administration & dosage , RNA, Small Interfering/administration & dosage , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cell Line, Tumor , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/therapy , Humans , Microscopy, Fluorescence , Oligopeptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Ribonucleotide Reductases/biosynthesis , Ribonucleotide Reductases/deficiency , Ribonucleotide Reductases/genetics
15.
Cancer Lett ; 311(1): 20-8, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21764510

ABSTRACT

The present study was performed to investigate the possible role of mTOR inhibitors in restoring chemosensitivity to adriamycin/cisplatin and elucidate the underlying mechanism. Combining adriamycin/cisplatin with torisel synergistically inhibited the cell proliferation in human oropharyngeal carcinoma cell line KB and its multidrug-resistant subclone KB/7D. Combining adriamycin and torisel inhibited the phosphorylation of 4EBP-1 and p70S6K, the proteins involved in mTOR pathway, increased expression of γH2AX indicative of DNA damage, triggered cell cycle arrest at G2/M and apoptosis. We conclude that chromatin decondensation by DNA damage provided an easy access for torisel to block the translation of proteins essential for DNA repair thereby restoring the chemosensitivity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , DNA Topoisomerases, Type II/metabolism , Head and Neck Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Topoisomerase II Inhibitors/pharmacology , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , DNA Damage , DNA Repair , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , Eukaryotic Initiation Factors/metabolism , Everolimus , Female , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism , Topoisomerase II Inhibitors/administration & dosage
16.
Mol Cancer Ther ; 9(6): 1669-79, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20484015

ABSTRACT

Ribonucleotide reductase (RNR) is an enzyme for the de novo conversion of ribonucleotides to deoxyribonucleotides. The two human RNR small subunits hRRM2 and hp53R2 share 83% sequence homology but show distinct expression patterns and function. Structural analyses of the oxidized form of hRRM2 and hp53R2 indicate that both proteins contain a conserved Gln127-hp53R2/Gln165-hRRM2 close to the dinuclear iron center and the essential tyrosine residue Tyr124-hp53R2/Tyr162-hRRM2 forms hydrogen bonds with the tyrosine and iron ligands, implying a critical role for the glutamine residue in assembling the dityrosyl-diiron radical cofactor. The present work also showed that Tyr221 in hRRM2, which is replaced by Phe183 in hp53R2, forms a hydrogen bond with Tyr162 to extend the hydrogen bond network from Gln165-hRRM2. Mutagenesis and spectroscopic experiments suggested that the tyrosine-to-phenylalanine switch at Phe183-hp53R2/Tyr221-hRRM2 could lead to differences in radical generation or enzymatic activity for hp53R2 and hRRM2. This study correlates the distinct catalytic mechanisms of the small subunits hp53R2 and hRRM2 with a hydrogen-bonding network and provides novel directions for designing and developing subunit-specific therapeutic agents for human RNR enzymes.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Free Radicals/metabolism , Iron Compounds/metabolism , Protein Subunits/metabolism , Ribonucleoside Diphosphate Reductase/chemistry , Ribonucleoside Diphosphate Reductase/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Circular Dichroism , Electron Spin Resonance Spectroscopy , Humans , Molecular Sequence Data , Mutagenesis/genetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Subunits/chemistry , Sequence Alignment
17.
Cancer Genomics Proteomics ; 5(3-4): 151-60, 2008.
Article in English | MEDLINE | ID: mdl-18820369

ABSTRACT

BACKGROUND: The expression and activity of ribonucleotide reductase (RR) has been associated with resistance to multiple drugs in human cancer. The use of antisense oligonucleotide drug, GTI-2040, a 20-mer phosphorothioate oligonucleotide complemented to the human RR M2 subunit mRNA, represents an effective strategy for inhibiting RR. The increased specificity due to the anti-resistance effect of GTI-2040 may also lead to a more favorable therapeutic outcome. MATERIALS AND METHODS: To understand the molecular mechanism underlying RR inhibition, patients' blood samples were analyzed using multiple dimensional proteomics technology via matrix-assisted laser desorption and ionization time-of-flight (MALDI-TOF) mass spectrometry. RESULTS: A major difference occurred at 5k m/z in the MALDI profile, which appeared only in the non-responsive group and diminished after GTI-2040 treatment. This specific peptide peak remained at the basal level in responsive patients. The peak was identified to represent the F-box/LLR-repeat protein 17 (FBXL17) through nanoelectrospray ionization liquid chromatography-tandem mass spectrometry (nanoESI LC-MS/MS). Further characterization revealed that FBXL17 [corrected] directly interacts with the human RR M2 (RRM2) subunit to promote hRRM2 overexpression in the breast cancer cell line MCF-7. CONCLUSION: Validation of this protein using real-time RT-PCR indicates the F-box protein 17 (FBXL17) can serve as a therapeutic target and surrogate marker for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , F-Box Proteins/metabolism , Oligodeoxyribonucleotides/therapeutic use , Base Sequence , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromatography, Liquid , DNA Primers , Electrophoresis, Polyacrylamide Gel , Female , Humans , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Biochem Pharmacol ; 73(12): 1927-38, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17428446

ABSTRACT

Mechanistic aberrations leading to Gemcitabine (2',2'-dFdCyd,2,2-difluorodeoxycytidine, Gem) resistance may include alteration in its transport, metabolism and incorporation into DNA. To explore the mechanism of Gem resistance, the restriction fragment differential display PCR (RFDD-PCR) was employed to compare the mRNA expression patterns of KBGem (Gem resistant), KBHURs (hydroxyurea resistant) and KBwt (parental KB cell). Nine gene fragments were overexpressed specifically in the KBGem clone. Sequencing and BLAST results showed that three fragments represent cytochrome C oxidase (CCOX, respiration complex IV) subunit III (CCOX3). The cDNA microarray confirmed that the mRNAs of CCOX and ATP synthase subunits were upregulated in KBGem as compared to KBwt and KBHURs. The increase in CCOX1 protein and activity led to the increase of free ATP concentration, which is consistent with the gene expression profile of KBGem. Furthermore, the sensitivity to Gem could be reversed by sodium azide, a CCOX inhibitor. Following the treatment of sodium azide, the cellular accumulation of [3H]-Gem increased in a dose (of azide)-dependent manner, which is associated with increase of [3H]-Gem incorporation into DNA in KBGem. In summary, an increase of CCOX activity and free ATP level may reduce the transport, metabolism and DNA incorporation of Gem, resulting in Gem resistance.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA/metabolism , Deoxycytidine/analogs & derivatives , Electron Transport Complex IV/metabolism , Adenosine Triphosphate/analysis , Blotting, Western , Clone Cells , DNA/genetics , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , KB Cells , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Sodium Azide/pharmacology , Gemcitabine
19.
Anticancer Drugs ; 18(4): 411-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17351393

ABSTRACT

Cisplatin has been used effectively to treat various human cancer types; yet, the precise mechanism underlying its cytotoxicity remains unknown. In eukaryotes, progression through G1 is monitored by a checkpoint, which executes G1 arrest in the event of DNA damage to allow time for repair before initiating DNA replication. The retinoblastoma tumor suppressor gene is an integral component of the mammalian G1 checkpoint. The utility of the retinoblastoma gene as a therapeutic for human cancers has been investigated. Intriguingly, the cytotoxicity profile of the retinoblastoma gene therapy closely parallels the clinical targets of cisplatin. It prompted an investigation into the potential role of the checkpoint-induced G1 arrest in cisplatin cytotoxicity. Here, the evidence that G1 arrest induction represents a critical step in cisplatin-induced lytic path is presented. First, cisplatin-treated human cancer cells undergo a prolonged G1 arrest before dying. Second, triggering G1 arrest via infection with a recombinant adenovirus expressing the human retinoblastoma gene is sufficient to potentiate lethality in the absence of cisplatin. Third, the extent of the lethality induced correlates with the G1-arresting potential of the ectopically expressed human retinoblastoma polypeptide. Fourth, human cancer cells resistant to cisplatin do not undergo G1 arrest despite cisplatin treatment. The above mechanism may be exploited to develop therapeutics that preserve the efficacy of cisplatin yet bypass its mutagenicity associated with the formation of secondary tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , G1 Phase/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Adenoviridae/genetics , Blotting, Western , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Flow Cytometry , Genetic Vectors , Humans , Retinoblastoma Protein/genetics , Retinoblastoma Protein/physiology , Reverse Transcriptase Polymerase Chain Reaction , TATA-Binding Protein Associated Factors/biosynthesis , TATA-Binding Protein Associated Factors/genetics
20.
Clin Colorectal Cancer ; 6(5): 374-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17311703

ABSTRACT

BACKGROUND: Ribonucleoside diphosphate reductase plays a key role in converting ribonucleoside diphosphate to 2'-deoxyribonucleoside diphosphate, which is necessary for DNA repair and replication. To determine if human ribonucleotide reductase small subunit M2 (hRRM2) and p53-dependent human ribonucleotide reductase small subunit R2 (p53R2) play roles on invasion ability of cancer cells, the gene transferring technique was used to construct stable hRRM2 and p53R2 overexpression transfectants. Increase of hRRM2 dramatically enhanced the cell migration in KB and PC-3 cells, but p53R2 overexpression reduced cellular invasion potential to 50% and 40% in KB and PC-3 cells, respectively. Furthermore, hRRM2 enhanced cancer cells to induce the cell migration of Human Umbilical Vein Endothelial Cells, but p53R2 reduced this ability in transfectants. PATIENTS AND METHODS: To further determine the role of human ribonucleotide reductase subunits on cancer metastasis, a tissue array, including 59 primary and 49 metastatic colon adenocarcinoma samples, was used. Immunohistochemistry was used to evaluate the relationship between human ribonucleotide reductase subunits and metastasis. RESULTS: Univariate and multivariate analysis revealed that p53R2 is negatively related to the metastasis of colon adenocarcinoma samples (odds ratio, 0.23; P < 0.05); hRRM2 increases the risk of metastasis in colon cancer, but did not show significantly. Thus, opposing regulation of hRRM2 and p53R2 in invasion potential might play a critical role in determining the invasion and metastasis phenotype in cancer cells. CONCLUSION: The expression level of ribonucleotide reductase small subunits could serve as biomarkers to predict the malignancy potential of human cancers in the future.


Subject(s)
Biomarkers, Tumor , Colonic Neoplasms/physiopathology , Ribonucleotide Reductases/metabolism , Tumor Suppressor Protein p53 , Adult , Aged , Cell Cycle Proteins , Cell Movement , Cell Survival , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , RNA-Binding Proteins , Ribonucleoside Diphosphate Reductase/metabolism , Risk , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...