Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(11): e0142845, 2015.
Article in English | MEDLINE | ID: mdl-26571496

ABSTRACT

PURPOSE: The paradigm shift in cancer treatment from cytotoxic drugs to tumor targeted therapies poses new challenges, including optimization of dose and schedule based on a biologically effective dose, rather than the historical maximum tolerated dose. Optimal dosing is currently determined using concentrations of tyrosine kinase inhibitors in plasma as a surrogate for tumor concentrations. To examine this plasma-tumor relationship, we explored the association between lapatinib levels in tumor and plasma in mice and humans, and those effects on phosphorylation of human epidermal growth factor receptors (HER) in human tumors. EXPERIMENTAL DESIGN: Mice bearing BT474 HER2+ human breast cancer xenografts were dosed once or twice daily (BID) with lapatinib. Drug concentrations were measured in blood, tumor, liver, and kidney. In a randomized phase I clinical trial, 28 treatment-naïve female patients with early stage HER2+ breast cancer received lapatinib 1000 or 1500 mg once daily (QD) or 500 mg BID before evaluating steady-state lapatinib levels in plasma and tumor. RESULTS: In mice, lapatinib levels were 4-fold higher in tumor than blood with a 4-fold longer half-life. Tumor concentrations exceeded the in vitro IC90 (~ 900 nM or 500 ng/mL) for inhibition of HER2 phosphorylation throughout the 12-hour dosing interval. In patients, tumor levels were 6- and 10-fold higher with QD and BID dosing, respectively, compared to plasma trough levels. The relationship between tumor and plasma concentration was complex, indicating multiple determinants. HER receptor phosphorylation varied depending upon lapatinib tumor concentrations, suggestive of changes in the repertoire of HER homo- and heterodimers. CONCLUSION: Plasma lapatinib concentrations underestimated tumor drug levels, suggesting that optimal dosing should be focused on the site of action to avoid to inappropriate dose escalation. Larger clinical trials are required to determine optimal dose and schedule to achieve tumor concentrations that maximally inhibit HER receptors. CLINICAL TRIAL REGISTRATION: NCT00359190.


Subject(s)
Antineoplastic Agents/blood , ErbB Receptors/metabolism , Quinazolines/blood , Administration, Oral , Adult , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Area Under Curve , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Administration Schedule , Drug Dosage Calculations , ErbB Receptors/antagonists & inhibitors , Female , Half-Life , Humans , Immunohistochemistry , Lapatinib , Mice , Mice, SCID , Phosphorylation/drug effects , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , ROC Curve , Tandem Mass Spectrometry , Transplantation, Heterologous
2.
Clin Pharmacol Drug Dev ; 4(6): 449-53, 2015 11.
Article in English | MEDLINE | ID: mdl-27137717

ABSTRACT

The potential for an interaction between lapatinib and absorption of the P-glycoprotein (ABCB1) substrate digoxin at a therapeutic dose in breast cancer patients was characterized. Seventeen women with HER2-positive metastatic breast cancer received a single oral 0.5-mg dose of digoxin on days 1 and 9 and oral lapatinib 1500 mg once daily on days 2 through 9. Digoxin pharmacokinetic parameters were determined on day 1 (digoxin administration alone) and on day 9 (coadministration of lapatinib and digoxin), and parameters were compared to determine the effects of lapatinib on digoxin absorption. Concomitant medications that could affect ABCB1 were accounted for. Lapatinib 1500 mg/day increased digoxin absorption approximately 80%, implicating lapatinib inhibition of intestinal ABCB1-mediated efflux. In summary, coadministration of lapatinib with narrow therapeutic index drugs that are substrates of ABCB1 should be undertaken with caution and dose adjustment should be considered.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Cardiotonic Agents/administration & dosage , Digoxin/administration & dosage , Digoxin/pharmacokinetics , Gastrointestinal Absorption/drug effects , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/metabolism , Administration, Oral , Adult , Alberta , Antineoplastic Agents/adverse effects , Area Under Curve , Breast Neoplasms/blood , Cardiotonic Agents/adverse effects , Cardiotonic Agents/blood , Cardiotonic Agents/pharmacokinetics , Cross-Over Studies , Digoxin/adverse effects , Digoxin/blood , Drug Interactions , Female , Half-Life , Humans , Lapatinib , Metabolic Clearance Rate , Middle Aged , Protein Kinase Inhibitors/adverse effects , Quinazolines/adverse effects , Risk Assessment , Seoul
3.
Clin Pharmacol Drug Dev ; 4(3): 203-9, 2015.
Article in English | MEDLINE | ID: mdl-27140800

ABSTRACT

Lapatinib is approved for use in various therapeutic combinations for treating metastatic breast cancers that over-express HER2. To deliver the approved doses, up to six large tablets need to be ingested with the current 250-mg tablets. For ease of ingestion, a powder for oral suspension was developed. This study was an open-label, randomized, adaptive design, two-period crossover bioequivalence study of the powder for suspension relative to the commercial tablet at steady state following once daily dosing for 7 days in patients with advanced cancer. To minimize the number of cancer patients required for a pivotal bioequivalence study (144 in this case), a four-stage adaptive group sequential design with interim analyses after every 36 subjects was implemented to allow for early termination. Bioequivalence for the oral suspension relative to the commercial tablet was demonstrated in both the first (and only) interim analysis and the final analysis, as the 90% confidence intervals for the treatment comparison ratios for both AUC0-24 and Cmax were contained within the acceptance criteria (0.80, 1.25). Additionally, there was no statistical difference in tlag or tmax , suggesting no difference in the absorption rate between treatments. There were no unexpected safety findings during this study.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Administration, Oral , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Area Under Curve , Cross-Over Studies , Drug Administration Schedule , Drug Compounding , Female , Humans , Lapatinib , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Neoplasms/blood , Neoplasms/enzymology , Pharmaceutical Solutions , Powders , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Quinazolines/adverse effects , Quinazolines/blood , Tablets , Therapeutic Equivalency , United States
4.
Invest New Drugs ; 32(3): 481-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24346280

ABSTRACT

AIM: To quantify the effect of food on the systemic exposure of lapatinib at steady state when administered 1 h before and after meals, and to observe the safety and tolerability of lapatinib under these conditions in patients with advanced solid tumours. METHODS: This was a three-treatment, randomised, three-sequence cross-over study. Lapatinib was administered 1 h after a low- [B] or a high-fat [C] meal and systemic exposure was compared with that obtained following administration 1 h before a low-fat meal [A]. RESULTS: In total, 25 patients were included, of whom 12 were evaluable for the pharmacokinetic analysis. Both low-fat and high-fat meals affected lapatinib exposure. Lapatinib AUC0-24 increased following lapatinib administration 1 h after a low-fat meal by 1.80-fold (90 % CI: 1.37-2.37) and after a high-fat meal by 2.61-fold (90 % CI: 1.98-3.43). Lapatinib Cmax increased following lapatinib administration 1 h after a low-fat meal by 1.90-fold (90 % CI: 1.49-2.43) and after a high-fat meal by 2.66-fold (90 % CI: 2.08-3.41). The most commonly occurring treatment-related toxicity was diarrhoea (8/25, 32 % CTCAE grade 1 and 2/25, 8 % grade 2) and one treatment-related grade ≥ 3 event occurred (fatigue grade 3, 4 %). CONCLUSIONS: Both low-fat and high-fat food consumed 1 h before lapatinib administration increased lapatinib systemic exposure compared with lapatinib administration 1 h before a low-fat meal. In order to administer lapatinib in a fasted state, it is advised to administer the drug 1 h before a meal.


Subject(s)
Antineoplastic Agents/administration & dosage , Dietary Fats/administration & dosage , Food-Drug Interactions , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Cross-Over Studies , Dietary Fats/pharmacokinetics , Humans , Lapatinib , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/adverse effects , Quinazolines/blood , Quinazolines/pharmacokinetics , Receptor, ErbB-2/metabolism
5.
Clin Pharmacol Drug Dev ; 2(4): 336-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-27121938

ABSTRACT

The aqueous solubility of lapatinib declines significantly at pH >4, suggesting that its bioavailability might be lowered by acid-reducing drugs. A study was therefore conducted to assess the effects of esomeprazole on lapatinib pharmacokinetics (PK). Women with metastatic human epidermal growth factor receptor 2 positive (HER2(+) ) breast cancer were enrolled. Patients received 1,250 mg lapatinib once daily (QD) in the morning on Days 1-7 (Period 1) and Days 8-14 (Period 2) with 40 mg esomeprazole QD at bedtime 3 hours after dinner on Days 8-14. Lapatinib PK sampling occurred during the 24-hour steady-state dosing intervals on Day 7 (lapatinib alone) and Day 14 (lapatinib with esomeprazole). Esomeprazole treatment resulted in decreased lapatinib bioavailability (mean 26%, range 6-49%) that was inversely associated with patient age as a significant covariate.

6.
Antivir Ther ; 9(4): 529-36, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15456084

ABSTRACT

Reverset (2',3'-didehydro-2',3'-dideoxy-5-fluorocytidine, RVT) is a potent inhibitor of HIV-1 replication in cell culture, with a 90% effective concentration at or below 1 microM. In vitro, RVT retains its activity against isolates harbouring mutations in the reverse transcriptase (RT) gene that otherwise confer resistance to lamivudine and/or zidovudine. The pharmacokinetics and safety of single oral doses of RVT (10-200 mg) were evaluated in an initial Phase I clinical trial. The viral load changes were determined on 18 HIV-1-infected antiretroviral therapy-naive subjects that were randomized into three cohorts, each cohort consisting of three study periods. The subjects received up to two oral doses of active drug and one placebo dose with a 1-week washout period separating the three study periods. Quantification of viral RNA was performed on the pre-dose, 12, 24 and 48 h post-dose plasma samples. A single oral dose of RVT to antiretroviral-naive subjects significantly reduced plasma viral load by 0.45 +/- 0.10 log10 copies/ml (P=0.0003). A mean drop of 0.37 +/- 0.12 log10 copies/ml (P=0.001) was obtained at the lowest dose of 10 mg. Sequence analysis of the HIV-1 RT gene performed before and after RVT dosing detected no genotypic changes in this short-term study. The viral RT gene of one subject had at predose the following genotype: L41 + N103 + C181 + W210 + D215, indicating prior exposure to zidovudine and non-nucleoside analogues, and anticipating high-level resistance against these agents. A single 10 mg RVT dose resulted in a viral load drop of 0.61 +/- 0.05 log10 providing evidence that a viral strain with the indicated genotype is susceptible to RVT.


Subject(s)
Cytidine Triphosphate/analogs & derivatives , Cytidine Triphosphate/therapeutic use , HIV Infections/drug therapy , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/isolation & purification , Reverse Transcriptase Inhibitors/therapeutic use , Administration, Oral , Adolescent , Adult , Cohort Studies , Cytidine Triphosphate/administration & dosage , Cytidine Triphosphate/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Genotype , HIV Infections/metabolism , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Middle Aged , Reverse Transcriptase Inhibitors/pharmacokinetics , Sequence Analysis, RNA , Viral Load , Zalcitabine/analogs & derivatives
7.
Curr Med Chem ; 11(6): 775-93, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15032731

ABSTRACT

Antimicrobial resistance in hospital and community settings is growing at an alarming rate and has been attributed to such organisms as methicillin-resistant staphylococcus aureus, staphylococci with decreased susceptibility to vancomycin, vancomycin-resistant enterococci, multi-drug resistant pseudomonas spp., klebsiella spp., enterobacter spp, and acinetobacter spp., as well as Streptococcus pneumoniae with decreased susceptibility to penicillin and other antibacterials. To address the need for new therapies to combat resistant organisms, drug companies are refocusing their discovery efforts on developing novel agents with new mechanisms of action. The hope is that rapidly emerging technologies including combinatorial chemistry, high throughput screening, proteomics and microbial genomics will have a positive impact on antimicrobial drug discovery. These technologies should aid in the identification of novel drug targets and compounds with unique mechanisms of action other than those currently provided by the traditional antibiotics. Nucleosides are one class of compounds worthy of further investigation as antibacterials since some derivatives have shown moderate to good activity against specific bacterial strains. For example, 5'-peptidyl nucleoside derivatives can inhibit peptide deformylase, an enzyme essential for bacterial survival that is not vital to human cells. This review also includes a list of miscellaneous nucleosides that have been synthesized as potential antibacterials. More detailed investigations on structure, as it relates to the antimicrobial activity of the various classes of nucleosides, need to be conducted in order to maximize the potential of developing a potent nucleoside for the treatment of bacterial infections. This review begins with an introduction to terms followed by discussions regarding the general background and relevance for developing novel antimicrobial agents. Challenges facing the antimicrobial drug discovery process are discussed along with relevant drug targets. An overview of nucleoside chemistry as it relates to antimicrobial activity is presented, followed by a discussion of the evidence which supports the potential of this class of compounds to yield the novel antimicrobial therapies needed in the new millennium.


Subject(s)
Anti-Infective Agents , Drug Design , Drug Resistance, Microbial , Nucleosides , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Combinatorial Chemistry Techniques , Communicable Diseases/drug therapy , Drug Resistance, Microbial/genetics , Humans , Nucleosides/chemistry , Nucleosides/pharmacology
8.
Antimicrob Agents Chemother ; 48(2): 651-4, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742230

ABSTRACT

2'-Deoxy-2'-fluorocytidine (FdC) is a potent inhibitor of the hepatitis C virus RNA replicon in culture, and FdC-5'-triphosphate is an effective inhibitor of the NS5B polymerase. Dynamic profiling of cell growth in an antiviral assay showed that FdC caused cytostasis due to an S-phase arrest. These observations demonstrate that FdC treatment is affecting both a viral target and a cellular target.


Subject(s)
Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hepacivirus/drug effects , Hepacivirus/genetics , RNA, Viral/biosynthesis , RNA, Viral/genetics , Replicon/drug effects , Replicon/genetics , Animals , Cattle , Cell Division/drug effects , Cell Line , Diarrhea Viruses, Bovine Viral/genetics , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , S Phase/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors
9.
Mol Cancer Ther ; 2(1): 83-93, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12533676

ABSTRACT

Interactions between the protein kinase C activator bryostatin 1 and the cyclin-dependent kinase (CDK) inhibitor flavopiridol (FP) have been examined in human myeloid leukemia cells (U937 and HL-60). Previous studies have demonstrated synergistic induction of apoptosis in leukemic cells exposed to the potent differentiation-inducer phorbol 12-myristate 13-acetate (PMA) in conjunction with FP [L. Cartee et al., Cancer Res., 61: 2583-2591, 2001]. Although bryostatin 1 (10 nM) is a very weak inducer of differentiation compared with PMA in these cells, coadministration of a minimally toxic concentration of FP (100 nM) did not promote bryostatin 1-related maturation but instead caused a marked increase in mitochondrial damage (e.g., cytochrome c release; loss of Deltapsi(m)), caspase activation, poly(ADP-ribose) polymerase cleavage, and apoptosis. Bryostatin 1/FP-induced apoptosis was significantly diminished in cells ectopically expressing dominant-negative Fas-associated death domain or by coadministration of tumor necrosis factor (TNF)-alpha soluble receptors, implicating the extrinsic pathway in bryostatin 1/FP actions. Enhanced apoptosis in bryostatin 1/FP-treated cells was accompanied by down-regulation of Mcl-1 and a sustained increase in TNF-alpha release. The selective protein kinase C inhibitor GFX blocked TNF-alpha and cytochrome c release in bryostatin 1/FP-treated cells and attenuated apoptosis. Finally, coadministration of bryostatin 1 (or PMA) with FP induced a marked increase in apoptosis in U937 cells ectopically expressing an NH(2)-terminal phosphorylation loop-deleted Bcl-2 protein, which are otherwise highly resistant to FP-mediated lethality. Taken together, these findings suggest that synergistic induction of apoptosis by bryostatin 1 and FP does not stem from disruption of the leukemic cell maturation process but instead results from enhanced release of TNF-alpha and activation of the extrinsic apoptotic cascade, culminating in cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Death/physiology , Flavonoids/pharmacology , Lactones/pharmacology , Piperidines/pharmacology , Protein Kinase C/metabolism , Receptors, Tumor Necrosis Factor/physiology , Apoptosis/drug effects , Bryostatins , Cell Adhesion/drug effects , Cell Death/drug effects , Drug Synergism , HL-60 Cells , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Kinetics , Macrolides , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mitochondria/drug effects , Mitochondria/physiology , U937 Cells
10.
Mol Cancer Ther ; 1(4): 253-66, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12467221

ABSTRACT

Interactions between the cyclin-dependent kinase inhibitor flavopiridol (FP) and the histone deacetylase inhibitor sodium butyrate (SB) have been examined in human leukemia cells (U937) in relation to differentiation and apoptosis. Whereas 1 mM of SB or 100 nM of FP minimally induced apoptosis (4% and 10%, respectively) at 24 h, simultaneous exposure of U937 cells to these agents dramatically increased cell death (e.g., approximately 60%), reflected by both morphological and Annexin/propidium iodide-staining features, procaspase 3 activation, and poly(ADP-ribose) polymerase cleavage. Similar interactions were observed in human promyelocytic (HL-60), B-lymphoblastic (Raji), and T-lymphoblastic (Jurkat) leukemia cells. Coadministration of FP opposed SB-mediated accumulation of cells in G0G1 and differentiation, reflected by reduced CD11b expression, but instead dramatically increased procaspase-3, procaspase-8, Bid, and poly(ADP-ribose) polymerase cleavage, as well as mitochondrial damage (e.g., loss of mitochondrial membrane potential and cytochrome c release). FP also blocked SB-related p21WAF1-CIP1 induction through a caspase-independent mechanism and triggered the caspase-mediated cleavage of p27KIP1 and retinoblastoma protein. The latter event was accompanied by a marked reduction in retinoblastoma protein/E2F1 complex formation. However, FP did not modify the extent of SB-associated acetylation of histones H3 and H4. Treatment of cells with FP/SB also resulted in the caspase-mediated cleavage of Bcl-2 and caspase-independent down-regulation of Mcl-1. Levels of cyclins A, D1, and E, and X-linked inhibitor of apoptosis also declined in SB/FP-treated cells. Finally, FP/SB coexposure potently induced apoptosis in two primary acute myelogenous leukemia samples. Together, these findings demonstrate that FP, when combined with SB, induces multiple perturbations in cell cycle and apoptosis regulatory proteins, which oppose leukemic cell differentiation but instead promote mitochondrial damage and apoptosis.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/biosynthesis , Cyclins/metabolism , Cyclins/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Leukemia/pathology , Piperidines/pharmacology , Blotting, Western , Caspases/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cytochrome c Group/metabolism , Enzyme Inhibitors/metabolism , Humans , Leukemia/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Tumor Cells, Cultured/pathology
11.
Int J Oncol ; 21(2): 351-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12118331

ABSTRACT

A clinically relevant dose (2.0 Gy) of ionizing radiation (IR) was employed to determine if subsequent exposure to the protein kinase C (PKC) and Chk 1 inhibitor UCN-01 for 24 h could abrogate IR-induced G2/M arrest and promote apoptosis in U937 leukemic cells ectopically expressing Bcl-2 (U937/Bcl-2). To this end, empty-vector control (U937/pCEP4) and U937/Bcl-2 cells were exposed to two UCN-01 concentrations following IR: i) a 50 nM concentration, which by itself was minimally toxic to both cell lines, and ii) a 150 nM concentration, which modestly induced apoptosis (e.g., ~19%) in control cells after 24 h. The effects of UCN-01 on IR responses were examined in relation to apoptosis induction, suspension culture growth inhibition, loss of clonogenic survival, and cell cycle perturbations. IR (2 Gy) alone minimally induced apoptosis in both U937 transfectant cell lines (e.g., <5% at 24 h in each case). Although UCN-01 failed to potentiate IR-mediated apoptosis at either early (e.g., 24 h) or late (e.g., 72 h) intervals, exposure to 50 or 150 nM UCN-01 resulted in a significant, albeit modest, reduction in proliferation and colony formation in irradiated U937/pCEP4 and U937/Bcl-2 cells. Despite failing to enhance apoptosis, UCN-01 treatment abrogated IR-induced G2/M arrest in both cell lines, an event associated with enhanced activation of cyclin-dependent kinase 1 (cdk1), promotion of G0/G1 arrest, and dephosphorylation of the retinoblastoma protein (pRb). Together, these findings indicate that exposure of U937 cells ectopically-expressing Bcl-2 to the combination of UCN-01 + IR leads to a further reduction in cell proliferation, and that this phenomenon appears to involve a non-apoptotic mechanism.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Proto-Oncogene Proteins c-bcl-2/metabolism , U937 Cells/drug effects , U937 Cells/radiation effects , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Combined Modality Therapy , Humans , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Protein Kinase C/antagonists & inhibitors , Radiation, Ionizing , Staurosporine/analogs & derivatives , Transfection , Tumor Stem Cell Assay , U937 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...