Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Eur J Nucl Med Mol Imaging ; 34(9): 1339-47, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17333178

ABSTRACT

PURPOSE: 3'-deoxy-3'-[18F]fluorothymidine positron emission tomography ([18F]FLT-PET) has been developed for imaging cell proliferation and findings correlate strongly with the Ki-67 labelling index in breast cancer. The aims of this pilot study were to define objective criteria for [18F]FLT response and to examine whether [18F]FLT-PET can be used to quantify early response of breast cancer to chemotherapy. METHODS: Seventeen discrete lesions in 13 patients with stage II-IV breast cancer were scanned prior to and at 1 week after treatment with combination 5-fluorouracil, epirubicin and cyclophosphamide (FEC) chemotherapy. The uptake at 90 min (SUV90) and irreversible trapping (Ki) of [18F]FLT were calculated for each tumour. The reproducibility of [18F]FLT-PET was determined in nine discrete lesions from eight patients who were scanned twice before chemotherapy. Clinical response was assessed at 60 days after commencing FEC. RESULTS: All tumours showed [18F]FLT uptake and this was reproducible in serial measurements (SD of mean % difference=10.5% and 15.1%, for SUV90 and Ki, respectively; test-retest correlation coefficient>or=0.97). Six patients had a significant clinical response (complete or partial) at day 60; these patients also had a significant reduction in [18F]FLT uptake at 1 week. Decreases in Ki and SUV90 at 1 week discriminated between clinical response and stable disease (p=0.022 for both parameters). In three patients with multiple lesions there was a mixed [18F]FLT response in primary tumours and metastases. [18F]FLT response generally preceded tumour size changes. CONCLUSION: [18F]FLT-PET can detect changes in breast cancer proliferation at 1 week after FEC chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dideoxynucleosides/pharmacology , Positron-Emission Tomography/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/diagnosis , Cell Proliferation , Cyclophosphamide/administration & dosage , Epirubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Ki-67 Antigen/biosynthesis , Kinetics , Middle Aged , Neoplasm Metastasis , Treatment Outcome
2.
Bioorg Med Chem Lett ; 17(1): 136-41, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17046252

ABSTRACT

Syntheses of aryloxyalkanoic acid hydroxyamides are described, all of which are potent inhibitors of histone deacetylase, some being more potent in vitro than trichostatin A (IC(50)=3 nM). Variation of the substituents on the benzene ring as well as fusion of a second ring have marked effects on potency, in vitro IC(50) values down to 1 nM being obtained.


Subject(s)
Amides/chemistry , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Histone Deacetylase Inhibitors , Amides/chemical synthesis , Amides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Histone Deacetylases/chemistry , Humans , Protein Conformation , Structure-Activity Relationship
3.
Cancer Res ; 66(15): 7621-9, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16885362

ABSTRACT

Histone deacetylase inhibitors (HDACI) are emerging as growth inhibitory compounds that modulate gene expression and inhibit tumor cell proliferation. We assessed whether 3'-deoxy-3'-[(18)F]fluorothymidine-positron emission tomography ([18F]FLT-PET) could be used to noninvasively measure the biological activity of a novel HDACI LAQ824 in vivo. We initially showed that thymidine kinase 1 (TK1; EC2.7.1.21), the enzyme responsible for [18F]FLT retention in cells, was regulated by LAQ824 in a drug concentration-dependent manner in vitro. In HCT116 colon carcinoma xenograft-bearing mice, LAQ824 significantly decreased tumor [18F]FLT uptake in a dose-dependent manner. At day 4 of treatment, [18F]FLT tumor-to-heart ratios at 60 minutes (NUV60) were 2.16 +/- 0.15, 1.86 +/- 0.13, and 1.45 +/- 0.20 in vehicle, and 5 and 25 mg/kg LAQ824 treatment groups, respectively (P < or = 0.05). LAQ825 at 5 mg/kg also significantly reduced both TK1 levels and [18F]FLT uptake at day 10 but not at day 2 (P < or = 0.05). [18F]FLT NUV60 correlated significantly with cellular proliferation (r = 0.68; P = 0.0019) and was associated with drug-induced histone H4 hyperacetylation. Of interest to [18F]FLT-PET imaging, both TK1 mRNA copy numbers and protein levels decreased in the order vehicle >5 mg/kg LAQ824 > 25 mg/kg LAQ824, providing a rationale for the use of [18F]FLT-PET in this setting. We also observed increases in Rb hypophosphorylation and p21 levels, factors that could have contributed to the alteration in TK1 transcription in vivo. In conclusion, we have shown the utility of [18F]FLT-PET for monitoring the biological activity of the HDACI, LAQ824. Drug-induced changes in tumor [18F]FLT uptake were due, at least in part, to reductions in TK1 transcription and translation.


Subject(s)
Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/drug therapy , Dideoxynucleosides , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Radiopharmaceuticals , Animals , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Dideoxynucleosides/pharmacokinetics , Drug Interactions , Female , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Positron-Emission Tomography , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiopharmaceuticals/pharmacokinetics , Random Allocation , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 12(5): 1585-92, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16533785

ABSTRACT

PURPOSE: Inhibition of steroid sulfatase (STS), the enzyme responsible for the hydrolysis of steroid sulfates, represents a potential novel treatment for postmenopausal women with hormone-dependent breast cancer. Estrone and DHEA are formed by this sulfatase pathway and can be converted to steroids (estradiol and androstenediol, respectively), which have potent estrogenic properties. EXPERIMENTAL DESIGN: STX64 (667 Coumate), a tricylic coumarin-based sulfamate that irreversibly inhibits STS activity, was selected for entry into the first phase I trial of a STS inhibitor in postmenopausal women with breast cancer. STX64 was administered orally (nine patients at 5 mg and five patients at 20 mg) as an initial dose followed 1 week later by 3 x 2 weekly cycles, with each cycle comprising daily dosing for 5 days followed by 9 days off treatment. Blood and tumor tissue samples were collected for the assessment of STS activity and serum was obtained for steroid hormone measurements before and after treatment. RESULTS: The median inhibition of STS activity by STX64 was 98% in peripheral blood lymphocytes (PBL) and 99% in breast tumor tissue at the end of the 5-day dosing period. As expected, serum concentrations of estrone, estradiol, androstenediol, and DHEA all decreased significantly from pretreatment levels. Unexpectedly, androstenedione and testosterone concentrations also decreased. Four patients, all of whom had previously progressed on aromatase inhibitors, showed evidence of stable disease for 2.75 to 7 months. The drug was well tolerated with only minor drug-related adverse events recorded. CONCLUSION: STX64 is a potent, well-tolerated STS inhibitor. It inhibits STS activity in PBLs and tumor tissues and causes significant decreases in serum concentrations of steroids with estrogenic properties.


Subject(s)
Breast Neoplasms/drug therapy , Coumarins/therapeutic use , Steryl-Sulfatase/antagonists & inhibitors , Sulfonamides/therapeutic use , Administration, Oral , Adult , Aged , Aged, 80 and over , Androstenediol/blood , Breast Neoplasms/enzymology , Dose-Response Relationship, Drug , Estradiol/blood , Estrone/blood , Female , Humans , Maximum Tolerated Dose , Middle Aged , Neoplasm Metastasis/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Steryl-Sulfatase/metabolism , Sulfonic Acids , Testosterone/blood
5.
Mol Cancer ; 5: 7, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16503970

ABSTRACT

BACKGROUND: The cyclin D1 proto-oncogene is an important regulator of G1 to S-phase transition and an important cofactor for several transcription factors in numerous cell types. Studies on neonatal cardiomyocytes and postmitotic neurons indicate that the activity of cyclin D1 may be regulated through its cytoplasmic sequestration. We have demonstrated previously, that TSA induces the ubiquitin-dependent degradation of cyclin D1 in MCF-7 breast cancer cells. Additional studies were initiated in order to further investigate the effect of TSA on cyclin D1 regulation using sub-cellular fractionation techniques. RESULTS: Our studies revealed cyclin D1 to be localized predominantly within the cytoplasmic fraction of all cell lines tested. These observations were confirmed by confocal microscopy. GSK3beta was found to be localized within both the nucleus and cytoplasm throughout the cell cycle. Inhibition of GSK3beta or CRM1-dependent nuclear export resulted in only modest nuclear accumulation, suggesting that the cytoplasmic localization of cyclin D1 results from the inhibition of its nuclear import. CONCLUSION: We have shown by several different experimental approaches, that cyclin D1 is in fact a predominantly cytoplasmic protein in mammalian cancer cell lines. Recent studies have shown that the cytoplasmic sequestration of cyclin D1 prevents apoptosis in neuronal cells. Our results suggest that cytoplasmic sequestration may additionally serve to regulate cyclin D1 activity in mammalian cancer cells.


Subject(s)
Cell Nucleus/metabolism , Cyclin D1/metabolism , Cytoplasm/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Cycle/drug effects , Cyclin D1/genetics , Cycloheximide/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , HeLa Cells , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Karyopherins/metabolism , Leupeptins/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Mas , Receptors, Cytoplasmic and Nuclear/metabolism , Transfection , Exportin 1 Protein
6.
Mol Cancer ; 5: 8, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16504004

ABSTRACT

BACKGROUND: Cyclin D1 is an important regulator of G1-S phase cell cycle transition and has been shown to be important for breast cancer development. GSK3beta phosphorylates cyclin D1 on Thr-286, resulting in enhanced ubiquitylation, nuclear export and degradation of the cyclin in the cytoplasm. Recent findings suggest that the development of small-molecule cyclin D1 ablative agents is of clinical relevance. We have previously shown that the histone deacetylase inhibitor trichostatin A (TSA) induces the rapid ubiquitin-dependent degradation of cyclin D1 in MCF-7 breast cancer cells prior to repression of cyclin D1 gene (CCND1) transcription. TSA treatment also resulted in accumulation of polyubiquitylated GFP-cyclin D1 species and reduced levels of the recombinant protein within the nucleus. RESULTS: Here we provide further evidence for TSA-induced ubiquitin-dependent degradation of cyclin D1 and demonstrate that GSK3beta-mediated nuclear export facilitates this activity. Our observations suggest that TSA treatment results in enhanced cyclin D1 degradation via the GSK3beta/CRM1-dependent nuclear export/26S proteasomal degradation pathway in MCF-7 cells. CONCLUSION: We have demonstrated that rapid TSA-induced cyclin D1 degradation in MCF-7 cells requires GSK3beta-mediated Thr-286 phosphorylation and the ubiquitin-dependent 26S proteasome pathway. Drug induced cyclin D1 repression contributes to the inhibition of breast cancer cell proliferation and can sensitize cells to CDK and Akt inhibitors. In addition, anti-cyclin D1 therapy may be highly specific for treating human breast cancer. The development of potent and effective cyclin D1 ablative agents is therefore of clinical relevance. Our findings suggest that HDAC inhibitors may have therapeutic potential as small-molecule cyclin D1 ablative agents.


Subject(s)
Cell Nucleus/metabolism , Cyclin D1/metabolism , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Ubiquitin/metabolism , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/metabolism , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Karyopherins/metabolism , Leupeptins/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , RNA Interference , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Exportin 1 Protein
8.
Cancer Res ; 65(21): 10104-12, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16267037

ABSTRACT

There is an unmet need to develop imaging methods for the early and objective assessment of breast tumors to therapy. 3'-Deoxy-3'-[18F]fluorothymidine ([18F]FLT)-positron emission tomography represents a new approach to imaging thymidine kinase activity, and hence, cellular proliferation. We compared graphical, spectral, and semiquantitative analytic methodologies for quantifying [18F]FLT kinetics in tumor and normal tissue of patients with locally advanced and metastatic breast cancer. The resultant kinetic parameters were correlated with the Ki-67 labeling index from tumor biopsies. [18F]FLT accumulation was detected in primary tumor, nodal disease, and lung metastasis. In large tumors, there was substantial heterogeneity in regional radiotracer uptake, reflecting heterogeneity in cellular proliferation; radiotracer uptake in primary tumors also differed from that of metastases. [18F]FLT was metabolized in patients to a single metabolite [18F]FLT-glucuronide. Unmetabolized [18F]FLT accounted for 71.54 +/- 1.50% of plasma radioactivity by 90 minutes. The rate constant for the metabolite-corrected net irreversible uptake of [18F]FLT (Ki) ranged from 0.6 to 10.4 x 10(-4) and from 0 to 0.6 x 10(-4) mL plasma cleared/s/mL tissue in tumor (29 regions, 15 patients) and normal tissues, respectively. Tumor Ki and fractional retention of radiotracer determined by spectral analysis correlated with Ki-67 labeling index (r = 0.92, P < 0.0001 and r = 0.92, P < 0.0001, respectively). These correlations were superior to those determined by semiquantitative methods. We conclude that [18F]FLT-positron emission tomography is a promising clinical tool for imaging cellular proliferation in breast cancer, and is most predictive when analyzed by graphical and spectral methods.


Subject(s)
Breast Neoplasms/diagnostic imaging , Dideoxynucleosides , Radiopharmaceuticals , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Dideoxynucleosides/blood , Dideoxynucleosides/pharmacokinetics , Fluorine Radioisotopes , Humans , Ki-67 Antigen/metabolism , Positron-Emission Tomography , Radiopharmaceuticals/blood , Radiopharmaceuticals/pharmacokinetics
9.
Lancet Oncol ; 6(6): 383-91, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15925816

ABSTRACT

BACKGROUND: Some oestrogen-receptor (ER) positive breast cancers express epidermal growth factor receptor (EGFR), but whether inhibition of EGFR can suppress proliferation of breast cancer cells and ER function is not known. METHODS: In a double-blind, placebo-controlled randomised trial of 56 postmenopausal patients with ER-positive and EGFR-positive primary breast cancer, 27 women were randomly assigned to the tyrosine-kinase inhibitor of EGFR gefitinib (250 mg given orally once a day) and the aromatase inhibitor anastrozole (1 mg given orally once a day), and 29 women to gefitinib (250 mg given orally once a day) and placebo of identical appearance to anastrozole given orally once a day, all given for 4-6 weeks before surgery. Primary outcome was inhibition of tumour-cell proliferation, as measured by Ki67 antigen labelling index. Secondary outcomes were reduction in EGFR phosphorylation at Tyr 845, reduction in ER phosphorylation at Ser 118, tumour size, and toxic effects. Analyses were by intention to treat. FINDINGS: Patients assigned gefitinib and anastrozole had a greater reduction from pretreatment values in proliferation-related Ki67 labelling index than did those assigned gefitinib alone (mean % reduction 98.0 [95% CI 96.1-98.9] vs 92.4 [85.1-96.1]; difference between groups 5.6% [5.1-6.0], p=0.0054). Tumour size was reduced by 30-99% (partial response) in 14 of 28 patients assigned gefitinib and [corrected]in 12 of 22 assigned gefitinib, as assessed by ultrasonography. Reduction in phosphorylation of ER at Ser 118 was similar for both groups. Treatment was well tolerated and much the same for both groups. INTERPRETATION: Single-agent gefitinib and gefitinib combined with anastrozole are well-tolerated and effective treatments for reducing the size of breast tumours and levels of ER phosphorylation when given as neoadjuvant therapy.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Nitriles/administration & dosage , Quinazolines/administration & dosage , Triazoles/administration & dosage , Anastrozole , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/pathology , Double-Blind Method , England , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Ki-67 Antigen , Middle Aged , Neoadjuvant Therapy , Neoplasms, Hormone-Dependent/diagnostic imaging , Neoplasms, Hormone-Dependent/pathology , Postmenopause , Protein Kinase Inhibitors/administration & dosage , Receptors, Estrogen/antagonists & inhibitors , Treatment Outcome , Ultrasonography
11.
J Biol Chem ; 280(5): 3185-96, 2005 Feb 04.
Article in English | MEDLINE | ID: mdl-15557281

ABSTRACT

We used the estrogen-responsive MCF-7 breast cancer cell line as a relevant model to study the anti-proliferative effects of ICI182,780 and identified the negative cell cycle regulator p21Waf1 as a specific target of ICI182,780. Furthermore, silencing of the p21Waf1 expression by small interfering RNA overcame the G0/G1 cell cycle arrest induced by ICI182,780, suggesting that the induction of p21Waf1 expression has a direct role in mediating the ICI182,780-induced G0/G1 arrest. We further demonstrated that the induction of p21Waf1 by ICI182,780 is mediated at transcriptional and gene promoter levels through the proximal Sp1 sites located near the transcription start site. Co-immunoprecipitation, DNA "pull-down," and chromatin immunoprecipitation experiments together showed that in cycling cells, estrogen receptor alpha and histone deacetylase 1 (HDAC1) are recruited to the proximal Sp1 sites of the promoter to repress p21Waf1 expression. In the presence of ICI182,780, estrogen receptor alpha and HDACs are dissociated from Sp1, resulting in increased histone acetylation and de-repression of the p21Waf1 promoter and induction of p21Waf1 expression. The fact that p21Waf1 expression is normally repressed by HDAC activity in cycling cells is further demonstrated by the finding that p21Waf1 transcription can be induced by the silencing of HDACs with small interfering RNA or treatment with HDAC inhibitors.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms , Cell Cycle Proteins/genetics , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Histone Deacetylases/metabolism , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Inhibitors/pharmacology , Fulvestrant , G1 Phase/drug effects , Gene Silencing , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic/physiology , RNA, Small Interfering , Sp1 Transcription Factor/metabolism , Transcriptional Activation/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
12.
Clin Cancer Res ; 10(23): 8094-104, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15585645

ABSTRACT

PURPOSE: Estrogen receptor alpha (ERalpha)-positive breast cancer cell lines are up to 10 times more sensitive than ERalpha-negative cell lines to the antiproliferative activity of the histone deacetylase inhibitor trichostatin A (TSA). The purpose of the study was to investigate the mechanisms underlying this differential response. EXPERIMENTAL DESIGN AND RESULTS: In the ERalpha-positive MCF-7 cell line, TSA repressed ERalpha and cyclin D1 transcription and induced ubiquitin dependent proteasomal degradation of cyclin D1, leading primarily to G(1)-S-phase cell cycle arrest. By contrast, cyclin D1 degradation was enhanced but its transcription unaffected by TSA in the ERalpha-negative MDA-MB-231 cell line, which arrested in G(2)-M phase. Cyclin D1 degradation involved Skp2/p45, a regulatory component of the Skp1/Cullin/F-box complex; silencing SKP2 gene expression by RNA interference stabilized cyclin D1 and abrogated the cyclin D1 down-regulation response to TSA. CONCLUSIONS: Tamoxifen has been shown to inhibit ERalpha-mediated cyclin D1 transcription, and acquired resistance to tamoxifen is associated with a shift to ERalpha-independent cyclin D1 up-regulation. Taken together, our data show that TSA effectively induces cyclin D1 down-regulation through both ERalpha-dependent and ERalpha-independent mechanisms, providing an important new strategy for combating resistance to antiestrogens.


Subject(s)
Breast Neoplasms/metabolism , Cyclin D1/metabolism , Estrogen Receptor alpha/metabolism , Hydroxamic Acids/pharmacology , Transcription, Genetic/drug effects , Uterine Neoplasms/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cysteine Proteinase Inhibitors/pharmacology , Drug Resistance, Neoplasm , Endopeptidases/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors , Humans , Leupeptins/pharmacology , RNA Interference , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , S-Phase Kinase-Associated Proteins/genetics , Tamoxifen/pharmacology , Tumor Cells, Cultured , Uterine Neoplasms/pathology
13.
J Reprod Med ; 49(7): 527-30, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15305823

ABSTRACT

OBJECTIVE: To assess whether a complete hydatidiform mole (CHM) carries an increased risk of later requiring chemotherapy in pregnancies continued to term. STUDY DESIGN: The Charing Cross gestational trophoblastic neoplasia (GTN) database was screened between 1973 and 2002 to identify registered singleton CHMs with a known gestational age at the time of evacuation. Of the 8,313 cases 2,800 were centrally histopathologically reviewed by us and confirmed as CHM. The proportion of patients requiring chemotherapyfor both total registered and centrally reviewed patients was analyzed by trimester of evacuation (< 13, 13-24, > 24 weeks). Statistical significance was assessed by the chi2 test. RESULTS: For the total population, including non-centrally reviewed patients, evacuation occurring in the first, second or third trimester was associated with a treatment rate of 13.9% (601 of 4,333), 10.8% (412 of 3,803) and 5.1% (9 of 177), respectively. In patientsfor whom a central pathologic review had been performed to confirm the diagnosis, the treatment rates were 27.7% (525 of 1,897), 27% (241 of 893) and 20% (2 of 10). The higher apparent treatment rates reflect an error in the denominator as we do not review all nontreated cases. In the total population, evacuation in the third trimester correlated with a reduction in risk of subsequent treatment (P<.001). Most of these late deliveries were induced (before adequate ultrasound), whereas the earlier pregnancies were mostly terminated via suction dilatation and curettage. CONCLUSION: There is no evidence that delayed evacuation/delivery of singleton CHM increases the risk of subsequently requiring chemotherapy.


Subject(s)
Cell Transformation, Neoplastic , Hydatidiform Mole/surgery , Uterine Neoplasms/surgery , Antineoplastic Agents/therapeutic use , Female , Gestational Age , Humans , Hydatidiform Mole/drug therapy , Hydatidiform Mole/physiopathology , Obstetric Surgical Procedures/methods , Pregnancy , Risk Factors , Time Factors , Uterine Neoplasms/drug therapy , Uterine Neoplasms/physiopathology
17.
Bioorg Med Chem Lett ; 14(10): 2477-81, 2004 May 17.
Article in English | MEDLINE | ID: mdl-15109636

ABSTRACT

Syntheses of (2E,4E)-5-arylpenta-2,4-dienoic acid hydroxyamides are described, some of which are potent inhibitors of histone deacetylase, a double bond conferring more than a 10-fold increase in potency compared with the triple bond analogue oxamflatin. Variation of substituents on the aromatic ring has a marked effect on potency, in vitro IC(50) values down to 50 nM being obtained.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Amides/chemical synthesis , Amides/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemical synthesis , Inhibitory Concentration 50 , Stereoisomerism , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 13(21): 3661-3, 2003 Nov 03.
Article in English | MEDLINE | ID: mdl-14552752

ABSTRACT

Some pyrazino[1,2-a]indole-1,4-diones, structurally simplified analogues of the natural mycotoxin gliotoxin, have been synthesised and investigated as inhibitors of prenyltransferases; one compound, 3-acetylthio-9-methoxy-2-methyl-2,3-dihydropyrazino[1,2-a]indole-1,4-dione 10 shows slightly greater selectivity (8-fold) for geranylgeranyltransferase type I (GGTase I) than gliotoxin itself.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Gliotoxin/analogs & derivatives , Gliotoxin/pharmacology , Indoles/chemical synthesis , Indoles/pharmacology , Pyrazines/chemical synthesis , Pyrazines/pharmacology , Farnesyltranstransferase , Genes, ras/genetics , Recombinant Proteins/chemical synthesis , Recombinant Proteins/pharmacology , Structure-Activity Relationship
20.
Curr Opin Investig Drugs ; 3(9): 1396-402, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12498018

ABSTRACT

The depsipeptide FR-901228 is a histone deacetylas inhibitor under development by Fujisawa and the National Cancer Institute as a potential treatment for neoplasm. By May 2002, phase II trials had commenced in the US [452248], [461017].


Subject(s)
Depsipeptides , Drug Industry/methods , Peptides, Cyclic/therapeutic use , Animals , Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Drug Industry/legislation & jurisprudence , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , National Institutes of Health (U.S.) , Neoplasms/drug therapy , Neoplasms/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...