Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Drug Metab Dispos ; 46(2): 109-121, 2018 02.
Article in English | MEDLINE | ID: mdl-29117990

ABSTRACT

Midostaurin (PKC412) is being investigated for the treatment of acute myeloid leukemia (AML) and advanced systemic mastocytosis (advSM). It is extensively metabolized by CYP3A4 to form two major active metabolites, CGP52421 and CGP62221. In vitro and clinical drug-drug interaction (DDI) studies indicated that midostaurin and its metabolites are substrates, reversible and time-dependent inhibitors, and inducers of CYP3A4. A simultaneous pharmacokinetic model of parent and active metabolites was initially developed by incorporating data from in vitro, preclinical, and clinical pharmacokinetic studies in healthy volunteers and in patients with AML or advSM. The model reasonably predicted changes in midostaurin exposure after single-dose administration with ketoconazole (a 5.8-fold predicted versus 6.1-fold observed increase) and rifampicin (90% predicted versus 94% observed reduction) as well as changes in midazolam exposure (1.0 predicted versus 1.2 observed ratio) after daily dosing of midostaurin for 4 days. The qualified model was then applied to predict the DDI effect with other CYP3A4 inhibitors or inducers and the DDI potential with midazolam under steady-state conditions. The simulated midazolam area under the curve ratio of 0.54 and an accompanying observed 1.9-fold increase in the CYP3A4 activity of biomarker 4ß-hydroxycholesterol indicated a weak-to-moderate CYP3A4 induction by midostaurin and its metabolites at steady state in patients with advSM. In conclusion, a simultaneous parent-and-active-metabolite modeling approach allowed predictions under steady-state conditions that were not possible to achieve in healthy subjects. Furthermore, endogenous biomarker data enabled evaluation of the net effect of midostaurin and its metabolites on CYP3A4 activity at steady state and increased confidence in DDI predictions.


Subject(s)
Cytochrome P-450 CYP3A/metabolism , Drug Interactions/physiology , Staurosporine/analogs & derivatives , Adult , Biomarkers/metabolism , Cytochrome P-450 CYP3A Inducers/metabolism , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/metabolism , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Female , Humans , Hydroxycholesterols/metabolism , Ketoconazole/metabolism , Ketoconazole/pharmacokinetics , Male , Midazolam/metabolism , Midazolam/pharmacokinetics , Middle Aged , Models, Biological , Rifampin/metabolism , Rifampin/pharmacokinetics , Staurosporine/metabolism , Staurosporine/pharmacokinetics , Young Adult
2.
Lancet Oncol ; 15(6): 592-600, 2014 May.
Article in English | MEDLINE | ID: mdl-24739897

ABSTRACT

BACKGROUND: The androgen receptor inhibitor enzalutamide is approved for the treatment of metastatic castration-resistant prostate cancer that has progressed on docetaxel. Our aim was to assess the activity and safety of enzalutamide monotherapy in men with hormone-naive prostate cancer. METHODS: This trial is an ongoing open-label, single-arm, phase 2 study, done across 12 European sites. Men aged over 18 years, with hormone-naive prostate cancer for whom hormone therapy was indicated, and who had non-castration levels of testosterone and prostate-specific antigen (PSA) of 2 ng/mL or greater at screening, and an Eastern Cooperative Oncology Group score of 0, received oral enzalutamide 160 mg/day. The primary outcome was the proportion of patients with an 80% or greater decline in PSA at week 25. All analyses included all patients who had received at least one dose of the study drug. This study is registered with ClinicalTrials.gov, number NCT01302041. FINDINGS: 67 men were enrolled into the study. 62 patients (92.5%, 95% CI 86.2-98.8) had a decline in PSA of 80% or greater at week 25. The most commonly reported treatment-emergent adverse events up to week 25 were gynaecomastia (n=24), fatigue (n=23), nipple pain (n=13), and hot flush (n=12), all of which were of mild to moderate severity. Nine patients had a treatment-emergent adverse event of grade 3 or higher, most of which were reported in one patient each, except for pneumonia (grade 3, two patients) and hypertension (grade 3, four patients). Five patients reported serious adverse events, none of which were deemed to be treatment related. INTERPRETATION: Our findings suggest that enzalutamide monotherapy in men with hormone-naive prostate cancer of varying severity provides a level of disease suppression, and was generally well tolerated. These findings provide a rationale for further investigation of clinical response and outcomes with enzalutamide in non-castrate men with prostate cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/drug therapy , Adenocarcinoma/blood , Aged , Aged, 80 and over , Androgen Antagonists/therapeutic use , Benzamides , Humans , Male , Middle Aged , Nitriles , Phenylthiohydantoin/therapeutic use , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Treatment Outcome
3.
Invest New Drugs ; 32(5): 860-70, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24981575

ABSTRACT

BACKGROUND: Aldo-keto reductase 1C3 [AKR1C3;17ß-hydroxysteroid dehydrogenase type 5 (17ßHSD5)], plays a crucial role in persistent production of androgens despite castration, by catalysing conversion of the adrenal androgens dehydroepiandrosterone and androstenedione (AD) into androstenediol and testosterone (T). Hence, AKR1C3 is a promising therapeutic target in castration-resistant prostate cancer, as combination of an AKR1C3 inhibitor and a gonadotropin-releasing hormone analogue may lead to complete androgen blockade. This study describes the preclinical characterisation of the novel AKR1C3 inhibitor ASP9521. METHODS: The inhibitory effect of ASP9521 on AKR1C3-mediated conversion from AD into T was evaluated both in vitro and in vivo, using CWR22R xenografted mice. The effect of ASP9521 on PSA production and cell proliferation was tested using LNCaP cells stably expressing human AKR1C3 (LNCaP-AKR1C3). Pharmacokinetics of ASP9521 were studied in rats, dogs and cynomolgus monkeys. RESULTS: ASP9521 inhibited conversion of AD into T by recombinant human or cynomolgus monkey AKR1C3 in a concentration-dependent manner (IC50,human: 11 nmol/L; IC50,monkey: 49 nmol/L). ASP9521 showed >100-fold selectivity for AKR1C3 over the isoform AKR1C2. In LNCaP-AKR1C3 cells, ASP9521 suppressed AD-dependent PSA production and cell proliferation. In CWR22R xenografts, single oral administration of ASP9521 (3 mg/kg) inhibited AD-induced intratumoural T production and this inhibitory effect was maintained for 24 h. After oral administration, ASP9521 was rapidly eliminated from plasma, while its intratumoural concentration remained high. The bioavailability of ASP9521 after oral administration (1 mg/kg) was 35 %, 78 % and 58 % in rats, dogs and monkeys, respectively. CONCLUSIONS: ASP9521 is a potent, selective, orally bioavailable AKR1C3 inhibitor.


Subject(s)
Enzyme Inhibitors/pharmacology , Estradiol Dehydrogenases/antagonists & inhibitors , Indoles/pharmacology , Piperidines/pharmacology , Administration, Oral , Androstenedione/metabolism , Animals , Biological Availability , Cell Line, Tumor , Dogs , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Humans , Indoles/blood , Indoles/pharmacokinetics , Macaca fascicularis , Male , Mice, Inbred BALB C , Piperidines/blood , Piperidines/pharmacokinetics , Prostatic Neoplasms/metabolism , Rats , Rats, Sprague-Dawley , Testosterone/metabolism
4.
Invest New Drugs ; 32(5): 995-1004, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24771350

ABSTRACT

BACKGROUND: ASP9521 is a first-in-class orally available inhibitor of the enzyme 17 ß-hydroxysteroid dehydrogenase type 5 (17 ßHSD5; AKR1C3), catalysing the conversion of dehydroepiandrosterone and androstenedione into 5-androstenediol and testosterone. It has demonstrated anti-tumour activity in in vitro and in vivo preclinical models. MATERIAL AND METHODS: This first-in-man phase I/II study utilised a 3 + 3 dose escalation design starting at 30 mg ASP9521/day, with the aim of defining a maximum tolerated dose, as defined by the incidence of dose-limiting toxicities. Eligible patients received ASP9521 orally for 12 weeks. Safety, tolerability, pharmacokinetics (PK), pharmacodynamics and anti-tumour activity were assessed. RESULTS: Thirteen patients (median age: 68 years; range 52-76) with metastatic castration-resistant prostate cancer (mCRPC) progressing after chemotherapy were included; 12 patients discontinued treatment at or before week 13, mainly due to disease progression. The most common adverse events were grade 1/2 and included asthenia (N = 5), constipation (N = 4), diarrhoea (N = 3), back pain (N = 3) and cancer pain (N = 3). PK demonstrated a half-life (t1/2) ranging from 16 to 35 h, rapid absorption and dose proportionality. No biochemical or radiological responses were identified; neither endocrine biomarker levels nor circulating tumour cell counts were altered by ASP9521. Given the lack of observable clinical activity, the study was terminated without implementing a planned 12-week dose expansion part at selected doses or a planned food-effect study part. CONCLUSIONS: In patients with mCRPC, ASP9521 demonstrated dose-proportional increase in exposure over the doses evaluated, with an acceptable safety and tolerability profile. However, the novel androgen biosynthesis inhibitor showed no relevant evidence of clinical activity.


Subject(s)
3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Androgen Antagonists/therapeutic use , Antineoplastic Agents/therapeutic use , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Indoles/therapeutic use , Piperidines/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Aged , Aldo-Keto Reductase Family 1 Member C3 , Androgen Antagonists/adverse effects , Androgen Antagonists/pharmacokinetics , Androgen Antagonists/pharmacology , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Count , Dihydrotestosterone/blood , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Indoles/pharmacology , Kallikreins/blood , Male , Middle Aged , Piperidines/adverse effects , Piperidines/pharmacokinetics , Piperidines/pharmacology , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/pathology , Testosterone/blood , Treatment Outcome
5.
Cancer Chemother Pharmacol ; 90(1): 19-27, 2022 07.
Article in English | MEDLINE | ID: mdl-35751657

ABSTRACT

PURPOSE: Midostaurin, approved for the treatment of newly diagnosed, FLT3-mutated acute myeloid leukemia (AML), is metabolized by cytochrome P450 3A4 (CYP3A4). Midostaurin with concomitant strong CYP3A4 inhibitors use (e.g., antifungal azoles) may result in drug-drug interactions. This post hoc analysis of RATIFY phase 3 study data evaluated effects of strong CYP3A4 inhibitor use on the exposure and safety of midostaurin. METHODS: Trough concentrations were used to assess midostaurin and metabolite exposure in the presence and absence of strong CYP3A4 inhibitors. Adverse event (AE) frequency was assessed in patients who received concomitant strong CYP3A4 inhibitors vs those who did not. Time to first clinically notable AE (CNAE) was also assessed in patients with high midostaurin plasma exposure vs those of matched placebo controls. RESULTS: Use of concomitant strong CYP3A4 inhibitors was most frequent during the induction phase (60.8%). A 1.44-fold increase in midostaurin plasma exposure was observed in patients with concomitant strong CYP3A4 inhibitor use vs those without. Midostaurin-treated patients who received concomitant strong CYP3A4 inhibitors experienced grade 3/4 infection-related AEs more frequently vs those who did not. Patients with high levels of midostaurin exposure had a shorter median time to first grade 3/4 CNAE vs placebo controls (36 vs 41 days, respectively; P = .012). CONCLUSION: Although concomitantly administered strong CYP3A4 inhibitors increased midostaurin exposure 1.44-fold, no clinically relevant differences in safety were noted. Midostaurin dose adjustment is not necessary with concomitant strong CYP3A4 inhibitors in patients with FLT3-mutated AML; however, caution is advised, and patients should be closely monitored.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Leukemia, Myeloid, Acute , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Drug Interactions , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Protein Kinase Inhibitors , Staurosporine/adverse effects , Staurosporine/analogs & derivatives , fms-Like Tyrosine Kinase 3/genetics
6.
Cancer Chemother Pharmacol ; 84(4): 749-757, 2019 10.
Article in English | MEDLINE | ID: mdl-31324935

ABSTRACT

PURPOSE: Ruxolitinib is metabolized by cytochrome P450 (CYP)3A4 and CYP2C9. Dual inhibitors of these enzymes (like fluconazole) lead to increased ruxolitinib exposure relative to a single pathway inhibition of CYP3A4 or CYP2C9. The magnitude of this interaction, previously assessed via physiologically based pharmacokinetic (PBPK) models, was confirmed in an open-label, phase 1 study in healthy subjects. METHODS: The effect of multiple doses (200 mg) of fluconazole on single-dose (10 mg) PK of ruxolitinib was investigated including evaluation of the safety and tolerability. The PK parameters of ruxolitinib alone (reference) were compared to those of ruxolitinib combined with fluconazole (test). The point estimate and corresponding two-sided 90% confidence interval for the difference between means of test and reference parameters were determined. RESULTS: All enrolled subjects (N = 15) completed the study. When coadministered with fluconazole, geometric means of ruxolitinib PK parameters Cmax, AUClast, and AUCinf increased by 47%, 234%, and 232%, respectively, vs ruxolitinib alone. The median Tmax decreased slightly, apparent clearance decreased approximately threefold, and elimination half-life increased approximately 2.5-fold, upon ruxolitinib administration with fluconazole vs ruxolitinib alone. These results were consistent with the prospective predictions from a SimCYP PBPK model. Adverse events (AEs) were reported in six subjects (none were suspected to be related to ruxolitinib); no death or on-treatment serious AE was reported. CONCLUSIONS: Coadministration of ruxolitinib with fluconazole significantly increased ruxolitinib systemic exposure; however, no AEs were attributed to ruxolitinib. Concomitant use of ruxolitinib with fluconazole (dose ≤ 200 mg) may require dose reduction/modification of ruxolitinib.


Subject(s)
Dose-Response Relationship, Drug , Fluconazole/pharmacokinetics , Metabolic Clearance Rate/drug effects , Pyrazoles/pharmacokinetics , Signal Transduction/drug effects , Adult , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Administration Schedule , Drug Interactions , Enzyme Inhibitors/pharmacokinetics , Female , Half-Life , Healthy Volunteers , Humans , Janus Kinases/metabolism , Male , Middle Aged , Nitriles , Pyrimidines
7.
Cancer Chemother Pharmacol ; 82(5): 847-855, 2018 11.
Article in English | MEDLINE | ID: mdl-30171280

ABSTRACT

PURPOSE: Eltrombopag is indicated in patients with severe aplastic anemia (SAA) refractory to prior immunosuppressive therapy. The combination of eltrombopag and immunosuppressive therapy (such as cyclosporine) is currently being evaluated in patients with treatment-naive SAA. Cyclosporine is a human breast cancer resistance protein (BCRP) inhibitor, and can potentially alter plasma exposure to eltrombopag, a BCRP substrate. This phase 1, open-label, randomized, 3-period, crossover study evaluated the effect of cyclosporine on the pharmacokinetics of eltrombopag in healthy adults. METHODS: Thirty-nine subjects were randomized to either single dose of eltrombopag 50 mg, cyclosporine 200 mg + eltrombopag 50 mg or cyclosporine 600 mg + eltrombopag 50 mg treatment groups. Eltrombopag pharmacokinetic parameters (Cmax, tmax, AUClast, AUCinf, %AUCex, t1/2, and CL/F) were determined using noncompartmental methods. RESULTS: Geometric mean AUCinf, AUClast, and Cmax, were decreased by 18, 20, and 25%, respectively, for cyclosporine 200 mg + eltrombopag and by 24, 22, and 39%, respectively, for cyclosporine 600 mg + eltrombopag groups compared to the eltrombopag alone group. The median tmax was prolonged by ~ 1 h in both coadministration treatments. The geometric mean t1/2 was ≈ 21, ≈ 24, and ≈ 26 h, respectively, in cyclosporine 200 mg + eltrombopag, cyclosporine 600 mg + eltrombopag and eltrombopag alone groups. All the treatments were safe and well-tolerated. No serious adverse event or death was reported during the study. CONCLUSION: These changes in exposure were not considered clinically meaningful as the dose of eltrombopag is adjusted using within-patient dose titration based on platelet counts.


Subject(s)
Benzoates/administration & dosage , Benzoates/blood , Cyclosporine/administration & dosage , Hydrazines/administration & dosage , Hydrazines/blood , Immunosuppressive Agents/administration & dosage , Pyrazoles/administration & dosage , Pyrazoles/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adolescent , Adult , Area Under Curve , Benzoates/adverse effects , Cross-Over Studies , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Female , Healthy Volunteers , Humans , Hydrazines/adverse effects , Immunosuppressive Agents/pharmacology , Male , Metabolic Clearance Rate , Middle Aged , Neoplasm Proteins/metabolism , Pyrazoles/adverse effects , Receptors, Thrombopoietin/agonists , Substrate Specificity , Young Adult
8.
Clin Pharmacol Ther ; 104(6): 1199-1207, 2018 12.
Article in English | MEDLINE | ID: mdl-29536526

ABSTRACT

Our objective was to support initial eltrombopag doses and dose titration based on modeling and simulation of plasma exposure and platelet count response in pediatric patients aged 1-17 years with previously treated chronic immune thrombocytopenia enrolled in two clinical studies. Data from 168 pediatric patients were used to develop a life-span population pharmacokinetic and pharmacodynamic model including three pharmacokinetic and four pharmacodynamic compartments enabling simulation of platelet counts for various starting doses and dose titration schedules. This work supported initial eltrombopag doses of 50 mg once daily (q.d.) for non-Asian patients aged ≥ 6 years and 25 mg q.d. for Asian patients, regardless of age, and for all patients aged 1-5 years, regardless of ethnic origin. Doses were escalated at 2-week intervals or reduced as needed according to each patient's platelet counts to both minimize the time to achieve target platelet counts and mitigate thrombocytosis. Clinicaltrials.gov Identifier: NCT00908037, NCT01520909.


Subject(s)
Benzoates/administration & dosage , Blood Platelets/drug effects , Computer Simulation , Drug Dosage Calculations , Hematinics/administration & dosage , Hydrazines/administration & dosage , Models, Biological , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Pyrazoles/administration & dosage , Adolescent , Age Factors , Benzoates/adverse effects , Benzoates/pharmacokinetics , Child , Child, Preschool , Female , Hematinics/adverse effects , Hematinics/pharmacokinetics , Humans , Hydrazines/adverse effects , Hydrazines/pharmacokinetics , Infant , Male , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/diagnosis , Purpura, Thrombocytopenic, Idiopathic/ethnology , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Treatment Outcome
9.
Clin Cancer Res ; 12(15): 4575-82, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16899604

ABSTRACT

PURPOSE: We evaluated the expression of endocan, a soluble lung- and kidney-selective endothelial cell-specific dermatan sulfate proteoglycan, in non-small cell lung tumors compared with normal lung and studied the significance of high levels of circulating endocan in patients with non-small cell lung cancer. MATERIAL AND METHODS: Endocan and vascular endothelial growth factor mRNA expression were evaluated by semiquantitative PCR in tumoral and nontumoral lung tissue samples from a first series of 24 patients submitted to curative surgery. Relationships between survival, time to tumor progression, and serum levels of endocan were evaluated in a second series of 30 previously untreated patients addressed for staging. RESULTS: In non-small cell lung cancers, endocan mRNA was overexpressed compared with control lung. Immunohistochemistry shows that endocan was expressed only by tumor endothelium in all cases, especially in the periphery of the tumors, with no differences between adenocarcinoma and squamous cell carcinoma. Endocan and vascular endothelial growth factor mRNA expression was positively correlated in lung tumors. Serum endocan levels, as well as tumor, node, and metastasis status, were correlated with both survival and time to tumor progression. However, endocan serum level was not an independent prognostic factor due to its correlation with the presence of metastasis. CONCLUSION: Endocan is overexpressed in non-small cell lung tumors compared with healthy lung and probably represents a response of tumoral endothelium to proangiogenic growth factor stimulation. Circulating levels of endocan might reflect tumor angiogenic stimulation and present prognostic significance.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Profiling , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Proteoglycans/genetics , Aged , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/surgery , Disease Progression , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/surgery , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Prognosis , Proteoglycans/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Survival Rate , Vascular Endothelial Growth Factor A/genetics
10.
Clin Cancer Res ; 22(15): 3774-81, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26858312

ABSTRACT

PURPOSE: Preclinical evidence suggests that both docetaxel and enzalutamide target androgen receptor translocation and signaling. This phase Ib study assessed the safety, tolerability, and pharmacokinetics of docetaxel when administered with enzalutamide as first-line systemic chemotherapy in men with metastatic castration-resistant prostate cancer (mCRPC). EXPERIMENTAL METHODS: Docetaxel-naïve patients received 21-day cycles of docetaxel (75 mg/m(2)). Enzalutamide (160 mg/day) was administered daily starting on day 2 of cycle 1. Patients were allowed to stop and restart docetaxel at any time following cycle 2. Treatment continued indefinitely until unacceptable toxicity or discontinuation due to investigator or patient preference. RESULTS: A total of 22 patients received docetaxel, of whom 21 also received enzalutamide. Docetaxel was administered for a median of 5.0 cycles and enzalutamide for a median of 12.0 months. With concomitant treatment, geometric mean docetaxel exposure decreased by 11.8%, whereas peak concentrations decreased by 3.7% relative to docetaxel alone. The most common toxicities observed during the period of concomitant therapy were neutropenia (86.4%) and fatigue (77.3%). Common toxicities observed with post-docetaxel enzalutamide were constipation (23.8%), decreased appetite (19.0%), fatigue (19.0%), and musculoskeletal pain (19.0%). Treatment with enzalutamide and docetaxel resulted in prostate-specific antigen decreases in almost all patients based on exploratory analysis of available baseline and on-study prostate-specific antigen data. CONCLUSIONS: The combination of docetaxel and enzalutamide is feasible, although higher rates of neutropenia and neutropenic fever than anticipated were observed. Reductions in docetaxel exposure with enzalutamide coadministration were not considered clinically meaningful. This combination warrants further study in a larger mCRPC population. Clin Cancer Res; 22(15); 3774-81. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzamides , Combined Modality Therapy , Docetaxel , Drug Monitoring , Humans , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/mortality , Retreatment , Taxoids/administration & dosage , Treatment Outcome
11.
Biochim Biophys Acta ; 1650(1-2): 73-91, 2003 Aug 21.
Article in English | MEDLINE | ID: mdl-12922171

ABSTRACT

Here, we describe a proteomics approach to study protein expression changes in differentiating Caco-2 cells. Caco-2 is a colorectal carcinoma cell line, which upon differentiation loses its tumorigenic phenotype and displays characteristics of mature enterocytes, including brush borders with microvilli. Cells were grown in culture flasks and harvested at different stages of differentiation (days post-confluence: -3, 0, 3, 7, 10, 14, and 18). Two-dimensional gel electrophoresis was used to analyse proteome changes. Approximately 1400 protein spots were detected within the Caco-2 proteome, within the pH 4-7 range. Two-dimensional gel electrophoresis allowed for the detection of 18 proteins from which the levels of expression were found to be associated with differentiation. Of these proteins, 11 were identified by means of MALDI-TOF or NANO-ESI-MS/MS mass spectrometry and include liver fatty acid binding protein (FABL), three forms of alpha-enolase (ENOA), nucleoside diphosphate kinase A (NDKA), cofilin-1 (COF1), translationally controlled tumour protein (TCTP), mitochondrial 60-kDa heat shock protein (CH60), probable protein disulfide isomerase (ER60), creatine kinase B (KCRB), and glutathione S-transferase alpha (GTA1). Thus, proteomics revealed that the differentiation-related change in phenotype of Caco-2 involves changes in a variety of distinct biochemical pathways. Some of these proteins have not been shown before to be associated with Caco-2 differentiation (ER60; COF1; CH60; NDKA; TCTP and ENOA). Therefore, processes related to protein folding and disulfide bridge formation, cytoskeleton formation and maintenance, nucleotide metabolism, glycolysis as well as tumorigenesis-associated proteins may be involved in Caco-2 differentiation. Changes in the expression of CH60, TCTP, GTA1, NDKA, and FABL have also been reported to be associated with in vivo colon carcinogenesis. These findings illustrate that a combination of proteomics and cell culture is a useful approach to find markers for Caco-2 differentiation, which could contribute to the comprehension of the process of colon carcinogenesis.


Subject(s)
Caco-2 Cells/physiology , Cell Differentiation/physiology , Proteome/physiology , Alkaline Phosphatase/metabolism , Blotting, Western , Caco-2 Cells/cytology , Cell Division/physiology , Humans , Principal Component Analysis , Tumor Protein, Translationally-Controlled 1
12.
Clin Cancer Res ; 9(10 Pt 1): 3763-72, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14506169

ABSTRACT

PURPOSE: Long-term elevation of metastasis suppressor gene expression in micrometastases represents a novel therapeutic strategy for breast and other cancers. We searched for well-tolerated compounds that could elevate Nm23 metastasis suppressor expression in metastatic human breast cancer cell lines. EXPERIMENTAL DESIGN: MDA-MB-435 and MDA-MB-231 human breast carcinoma cells were treated with dexamethasone or medroxyprogesterone acetate (MPA) in cultures containing either charcoal-stripped serum or FCS. Aspects of nm23 expression and function were determined. RESULTS: Previous investigation of the nm23-H1 promoter suggested that glucocorticoids may contribute to the elevation of Nm23-H1 expression. Dexamethasone elevated Nm23-H1 and Nm23-H2 protein levels in two metastatic human breast carcinoma cell lines 2-3-fold over a 4-day time course when cultured in steroid-free culture medium, with high-dose inhibition, via a traditional transcriptional mechanism. Elevation of Nm23-H1 expression was not observed using FCS-containing culture medium, which contains endogenous levels of corticosteroids, limiting the potential in vivo use of dexamethasone. MPA was investigated as a glucocorticoid receptor agonist. MPA elevated breast carcinoma Nm23-H1 protein expression 3-fold over a 10 nM to 1 micro M dose range when cultured in steroid-free or FCS-containing medium, with a shorter time course. Elevation of Nm23-H1 expression in the presence of endogenous corticosteroids found in FCS involved a distinct, glucocorticoid receptor-dependent, posttranscriptional mechanism of action. MPA had no effect on proliferation in vitro but reduced the soft agar colonization of metastatic breast cancer cell lines by approximately 50%. CONCLUSIONS: MPA represents a first generation lead agent for the elevation of Nm23-H1 metastasis suppressor expression and the inhibition of metastatic colonization.


Subject(s)
Breast Neoplasms/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic , Medroxyprogesterone/pharmacology , Nucleoside-Diphosphate Kinase , Protein Biosynthesis , Agar/chemistry , Antineoplastic Agents, Hormonal/pharmacology , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Cell Division , Cell Line, Tumor , Contraceptives, Oral, Synthetic/pharmacology , Dexamethasone/metabolism , Dose-Response Relationship, Drug , Glucocorticoids/metabolism , Humans , Medroxyprogesterone Acetate/metabolism , Mutagenesis , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Time Factors , Transcription, Genetic , Transfection
13.
Clin Pharmacokinet ; 54(10): 1043-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25917876

ABSTRACT

BACKGROUND AND OBJECTIVES: Oral enzalutamide (160 mg once daily) is approved for the treatment of metastatic castration-resistant prostate cancer (mCRPC). This article describes the pharmacokinetics of enzalutamide and its active metabolite N-desmethyl enzalutamide. METHODS: Results are reported from five clinical studies. RESULTS: In a dose-escalation study (n = 140), enzalutamide half-life was 5.8 days, steady state was achieved by day 28, accumulation was 8.3-fold, exposure was approximately dose proportional from 30-360 mg/day, and intersubject variability was ≤30 %. In a mass balance study (n = 6), enzalutamide was primarily eliminated by hepatic metabolism. Renal excretion was an insignificant elimination pathway for enzalutamide and N-desmethyl enzalutamide. In a food-effect study (n = 60), food did not have a meaningful effect on area under the plasma concentration-time curve (AUC) of enzalutamide or N-desmethyl enzalutamide, and in an hepatic impairment study, AUC of the sum of enzalutamide plus N-desmethyl enzalutamide was similar in men with mild (n = 6) or moderate (n = 8) impairment (Child-Pugh Class A and B) versus men with normal hepatic function (n = 14). In a phase III trial, an exposure-response analysis of steady-state predose (trough) concentrations (C trough) versus overall survival (n = 1103) showed that active treatment C trough quartiles for 160 mg/day were uniformly beneficial relative to placebo, and no threshold of C trough was associated with a statistically significant better response. CONCLUSIONS: Enzalutamide has predictable pharmacokinetics, with low intersubject variability. Similar efficacy was observed in patients across the concentration/exposure range associated with a fixed oral dose of enzalutamide 160 mg/day.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/blood , Benzamides , Biotransformation , Dose-Response Relationship, Drug , Drug Administration Schedule , Food-Drug Interactions , Humans , Male , Middle Aged , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/blood , Phenylthiohydantoin/pharmacokinetics , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/metabolism
14.
Clin Pharmacokinet ; 54(10): 1057-69, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25929560

ABSTRACT

BACKGROUND AND OBJECTIVES: Two phase I drug interaction studies were performed with oral enzalutamide, which is approved for the treatment of metastatic castration-resistant prostate cancer (mCRPC). METHODS: A parallel-treatment design (n = 41) was used to evaluate the effects of a strong cytochrome P450 (CYP) 2C8 inhibitor (oral gemfibrozil 600 mg twice daily) or strong CYP3A4 inhibitor (oral itraconazole 200 mg once daily) on the pharmacokinetics of enzalutamide and its active metabolite N-desmethyl enzalutamide after a single dose of enzalutamide (160 mg). A single-sequence crossover design (n = 14) was used to determine the effects of enzalutamide 160 mg/day on the pharmacokinetics of a single oral dose of sensitive substrates for CYP2C8 (pioglitazone 30 mg), CYP2C9 (warfarin 10 mg), CYP2C19 (omeprazole 20 mg), or CYP3A4 (midazolam 2 mg). RESULTS: Coadministration of gemfibrozil increased the composite area under the plasma concentration-time curve from time zero to infinity (AUC∞) of enzalutamide plus active metabolite by 2.2-fold, and coadministration of itraconazole increased the composite AUC∞ by 1.3-fold. Enzalutamide did not affect exposure to oral pioglitazone. Enzalutamide reduced the AUC∞ of oral S-warfarin, omeprazole, and midazolam by 56, 70, and 86 %, respectively; therefore, enzalutamide is a moderate inducer of CYP2C9 and CYP2C19 and a strong inducer of CYP3A4. CONCLUSIONS: If a patient requires coadministration of a strong CYP2C8 inhibitor with enzalutamide, then the enzalutamide dose should be reduced to 80 mg/day. It is recommended to avoid concomitant use of enzalutamide with narrow therapeutic index drugs metabolized by CYP2C9, CYP2C19, or CYP3A4, as enzalutamide may decrease their exposure.


Subject(s)
Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Adult , Aged , Aged, 80 and over , Benzamides , Cross-Over Studies , Cytochrome P-450 CYP2C8 Inhibitors/administration & dosage , Cytochrome P-450 CYP2C8 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Humans , Male , Midazolam/pharmacokinetics , Middle Aged , Nitriles , Phenylthiohydantoin/administration & dosage , Phenylthiohydantoin/pharmacokinetics , Prostatic Neoplasms, Castration-Resistant/enzymology
15.
Eur Urol ; 68(5): 787-94, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25687533

ABSTRACT

BACKGROUND: Enzalutamide is an androgen receptor inhibitor with a demonstrated overall survival benefit in metastatic castration-resistant prostate cancer. A phase 2 study of enzalutamide monotherapy in patients with hormone-naïve prostate cancer (HNPC) showed a high response rate for the prespecified primary endpoint (ie, prostate-specific antigen [PSA] response at week 25), regardless of metastases at baseline, and favorable tolerability. OBJECTIVE: To determine the long-term efficacy and safety of enzalutamide monotherapy at 1 and 2 yr. DESIGN, SETTING, AND PARTICIPANTS: Open-label, single-arm study in patients with HNPC and noncastrate testosterone (≥230 ng/dl). INTERVENTION: Oral enzalutamide 160mg/d until disease progression or unacceptable toxicity. OUTCOME MEASUREMENTS AND ANALYSIS: PSA response (≥80% decline from baseline) assessed at 1 yr (49 wk) and 2 yr (97 wk). RESULTS AND LIMITATIONS: The median (range) age was 73 (48-86) yr and 26 patients (39%) presented with metastases at study entry. Of 67 patients enrolled, 45 (67%) remained on enzalutamide at week 97. For patients remaining on therapy, the PSA response rate at week 97 was 100% (95% confidence interval 92-100%). Of 26 patients with metastases at baseline, 13 (50%) had a complete and four (15.4%) had a partial response as best overall tumor response up to 97 wk on treatment. There was overall maintenance of total-body bone mineral density (BMD) and moderate changes in lean and fat body mass at 49 and 97 wk. The most common adverse events were gynecomastia, nipple pain, fatigue, and hot flushes. The study limitations include lack of a control group and of endocrine, glycemic, and lipid data at 97 wk. CONCLUSIONS: Long-term enzalutamide monotherapy in men with noncastrate HNPC is associated with large sustained reductions in PSA, signals indicating a favorable tumor response, and favorable safety/tolerability profile, with relatively small negative effects on total-body BMD. PATIENT SUMMARY: In this long-term follow-up of the efficacy and safety of enzalutamide monotherapy in patients with hormone-naïve prostate cancer, enzalutamide maintained long-term reductions in prostate-specific antigen, with a minimal impact on total-body bone mineral density. TRIAL REGISTRATION: NCT01302041.


Subject(s)
Androgen Receptor Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Neoplasm Recurrence, Local/blood , Phenylthiohydantoin/analogs & derivatives , Prostatectomy , Prostatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Benzamides , Chemotherapy, Adjuvant , Follow-Up Studies , Humans , Kallikreins/blood , Longitudinal Studies , Male , Middle Aged , Nitriles , Phenylthiohydantoin/therapeutic use , Prostate-Specific Antigen/blood , Prostatic Neoplasms/pathology , Treatment Outcome
16.
Clin Exp Metastasis ; 20(1): 3-10, 2003.
Article in English | MEDLINE | ID: mdl-12650601

ABSTRACT

The first metastasis suppressor gene identified was nm23. Transfection of nm23 into metastatic cell lines resulted in the inhibition of metastasis, but not primary tumor size in vivo. Using in vitro assays, nm23 overexpression resulted in reduced anchorage-independent colonization in response to TGF-beta, reduced invasion and motility in response to multiple factors, and increased differentiation. We hypothesize that the mechanism of action of Nm23 in metastasis suppression involves diminished signal transduction downstream of a particular receptor. Candidate biochemical mechanisms are identified and discussed herein.


Subject(s)
Genes, Tumor Suppressor , Monomeric GTP-Binding Proteins/genetics , Neoplasm Metastasis/genetics , Nucleoside-Diphosphate Kinase , Signal Transduction/genetics , Transcription Factors/genetics , Animals , Cell Differentiation , Histidine Kinase , Humans , Models, Biological , NM23 Nucleoside Diphosphate Kinases , Protein Kinases/metabolism , Structure-Activity Relationship , Transfection
17.
Clin Exp Metastasis ; 19(1): 35-42, 2002.
Article in English | MEDLINE | ID: mdl-11918081

ABSTRACT

We hypothesize that elevation of nm23-HI metastasis suppressor gene expression in micrometastatic tumor cells may reduce their subsequent colonization and invasion, and induce differentiation, with a clinical benefit. This report presents the first analysis of the nm23-HI promoter to identify sites which can increase its transcription. Deletion mapping of a 2.1 kb nm23-H1 promoter fragment tethered to a reporter gene identified three regions involved in its differential expression levels among a panel of human breast carcinoma cell lines: a 195 bp NheI-XbaI fragment responsible for basal expression levels, a 248 bp AvrII-Nhel fragment which contributed to the elevated nm23-H1 expression observed in the high expressing cell lines, and a 544 bp AvrII fragment containing an inhibitory element. Examination of the 248 bp AvrII-NheI fragment revealed the unexpected presence of three transcription factor binding sites (MAF/Ets, CTF/NF1 half site and ACAAAG enhancer) previously identified in the MMTV-LTR, and in WAP and milk gene promoters, proposed to mediate mammary-specific gene expression. Mutation of the three sites, individually or together, resulted in two-fold reductions in reporter gene expression. As controls, the same panel of mutations caused a different pattern of reporter gene expression in a non-mammary cell line, and mutation of another nearby site was without effect on nm23-HI. Our data identify a complex regulatory pattern for nm23-H1 transcription, and suggest that a mammary-specific cassette of transcription factors contribute to its elevated expression


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Mammary Tumor Virus, Mouse/genetics , Monomeric GTP-Binding Proteins/biosynthesis , Neoplasm Metastasis/genetics , Neoplasm Proteins/biosynthesis , Nucleoside-Diphosphate Kinase , Promoter Regions, Genetic/genetics , Terminal Repeat Sequences/genetics , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Consensus Sequence , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/genetics , Female , Genes, Reporter , Humans , Monomeric GTP-Binding Proteins/genetics , Mutagenesis , NFI Transcription Factors , NM23 Nucleoside Diphosphate Kinases , Neoplasm Proteins/genetics , Organ Specificity , Polymorphism, Restriction Fragment Length , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Recombinant Fusion Proteins/biosynthesis , Sequence Deletion , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured
18.
Cancer Lett ; 190(1): 1-12, 2003 Feb 10.
Article in English | MEDLINE | ID: mdl-12536071

ABSTRACT

Intensive investigation of protein tyrosine, serine and threonine phosphorylation has lead to advances in signal transduction research and cancer treatment. This feature summarizes research on mammalian proteins exhibiting histidine phosphorylation. Histidine kinases are well known in prokaryotic and lower eukaryotic systems where they form the 'two-component' signal transduction system. The relative invisibility of histidine phosphorylation in mammalian cells may result from technical obstacles such as its acid lability, which precludes detection in electrophoretic systems, amino acid sequencing, etc. Emerging data have identified mammalian histidine kinases for the kinase suppressor of ras, a scaffold molecule for the Map kinase pathway, as well as histone H4, aldolase C and the beta-subunit of heterotrimeric G proteins. Additional mammalian proteins of interest to signal transduction and cancer research exhibit histidine phosphorylation, including P-selectin, annexin I and the 20S proteasome. Other candidate histidine phosphorylated proteins are identified. These data suggest the existence of another series of phosphorylation patterns in signal transduction.


Subject(s)
Histidine/metabolism , Neoplasms/metabolism , Nucleoside-Diphosphate Kinase , Protein Kinases/physiology , Signal Transduction , Animals , Annexin A1/metabolism , Cysteine Endopeptidases/metabolism , GTP-Binding Proteins/metabolism , Histidine Kinase , Humans , Models, Biological , Models, Chemical , Monomeric GTP-Binding Proteins/metabolism , Multienzyme Complexes/metabolism , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , P-Selectin/metabolism , Phosphorylation , Proteasome Endopeptidase Complex , Transcription Factors/metabolism
19.
Clin Breast Cancer ; 4(1): 51-62, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12744759

ABSTRACT

Metastatic disease remains a significant contributor to morbidity and mortality in patients with breast cancer. An improved molecular and biochemical understanding of the metastatic process is expected to fuel the development of new therapeutic approaches. The suppression of tumor metastasis, despite tumor cell expression of oncogenes and metastasis-promoting events, has become a diverse and fruitful field of investigation. Although many genetic events promote metastasis, several genes show relatively reduced expression levels in metastatic tumor cells in mouse model systems and in aggressive human tumors. Re-expression of a metastasis-suppressor gene in a metastatic tumor cell line results in a significant reduction in metastatic behavior in vivo with no effect on tumorigenicity. The known metastasis-suppressor gene products nm23, KAI1, mitogen-activated protein kinase kinase 4, breast cancer metastasis suppressor-1, KiSS1, RHOGDI2, CRSP3, and vitamin D3-upregulated protein/thioredoxin interacting protein exhibit unexpected biochemical functions that have shed new light on signaling events that are important in metastasis. Most metastasis suppressors function at the translationally important stage of outgrowth of micrometastatic tumor cells at a distant site. We hypothesize that elevation of metastasis suppressor gene expression in micrometastatic tumor cells in the adjuvant high-risk population of patients with breast cancer will halt metastatic colonization and have a clinical benefit. DNA methylation inhibitors have shown limited promise in increasing metastasis-suppressor gene expression, and ligands of the nuclear hormone receptor family are currently under investigation in vitro and in vivo. Clinical testing of agents that increase metastasis-suppressor gene expression is expected to require tailored trial designs.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor , Animals , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Division/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis/genetics
20.
J Natl Cancer Inst ; 97(9): 632-42, 2005 May 04.
Article in English | MEDLINE | ID: mdl-15870434

ABSTRACT

BACKGROUND: Reestablishment of metastasis suppressor gene expression may constitute a therapeutic strategy for high-risk breast cancer patients. We previously showed that medroxyprogesterone acetate (MPA), a progestin that has been tested as treatment for advanced breast cancer, elevates expression of the Nm23-H1 metastasis suppressor gene in hormone receptor-negative metastatic human breast carcinoma cell lines in vitro via a glucocorticoid receptor-based mechanism. Here, we tested whether MPA treatment inhibits metastatic colonization of a hormone receptor-negative breast cancer cell line in vivo. METHODS: We tested the soft-agar colony-forming efficiency of untransfected MDA-MB-231T human breast carcinoma cells and MDA-MB-231T cells transfected with antisense Nm23-H1 in the presence and absence of MPA. Pharmacokinetic studies were used to establish dose and injection schedules that led to MPA serum levels in mice similar to those achievable in humans. For in vivo studies, nude mice were injected intravenously with MDA-MB-231T cells. After 4 weeks, mice were randomized to control or MPA arms. Endpoints included incidence, number, and size of gross pulmonary metastases; Nm23-H1 protein expression in gross metastases; and side effects. All statistical tests were two-sided. RESULTS: MPA reduced colony formation of MDA-MB-231T cells by 40%-50% but had no effect on colony formation of Nm23-H1 antisense transfectants. Metastases developed in 100% (95% confidence interval [CI] = 78% to 100% and 77% to 100%, respectively) of control mice injected with MDA-MB-231T cells. In two independent experiments, only 73% (95% CI = 45% to 92%) and 64% (95% CI = 35% to 87%) of mice injected with 2 mg of MPA developed metastases. Mice injected with 2 mg of MPA showed reductions in the mean numbers, per mouse, of all metastases and of large (>3 mm) metastases (P = .04 and .013, respectively). Nm23-H1 was expressed at high levels in 43% of pulmonary metastases in MPA-treated mice but only 13% of metastases in untreated mice. Mice receiving at least 1-mg doses of MPA gained more weight than control-treated mice but exhibited no bone density alterations or abnormal mammary fat pad histology. CONCLUSION: Our preclinical results show that MPA appears to elevate Nm23-H1 metastasis suppressor gene expression, thereby reducing metastatic colonization. The data suggest a new use for an old agent in a molecularly defined subset of breast cancer patients.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor/drug effects , Medroxyprogesterone Acetate/pharmacology , Nucleoside-Diphosphate Kinase/drug effects , Nucleoside-Diphosphate Kinase/metabolism , Animals , Blotting, Western , Breast Neoplasms/blood , Cell Line, Tumor , DNA, Antisense , Female , Humans , Immunohistochemistry , Mice , NM23 Nucleoside Diphosphate Kinases , Nucleoside-Diphosphate Kinase/blood , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL