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










Publication year range
1.
Pharmaceutics ; 10(2)2018 May 19.
Article in English | MEDLINE | ID: mdl-29783757

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and lethal central nervous system tumor. Recently, atovaquone has shown inhibition of signal transducer and activator transcription 3, a promising target for GBM therapy. However, it is currently unable to achieve therapeutic drug concentrations in the brain with the currently reported and marketed formulations. The present study sought to explore the efficacy of atovaquone against GBM as well as develop a formulation of atovaquone that would improve oral bioavailability, resulting in higher amounts of drug delivered to the brain. Atovaquone was formulated as an amorphous solid dispersion using an optimized formulation containing a polymer and a spontaneously emulsifying component (SEC) with greatly improved wetting, disintegration, dispersibility, and dissolution properties. Atovaquone demonstrated cytotoxicity against GBM cell lines as well as provided a confirmed target for atovaquone brain concentrations in in vitro cell viability studies. This new formulation approach was then assessed in a proof-of-concept in vivo exposure study. Based on these results, the enhanced amorphous solid dispersion is promising for providing therapeutically effective brain levels of atovaquone for the treatment of GBM.

2.
Mol Pharm ; 15(4): 1392-1402, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29485888

ABSTRACT

Effective targeting of nasal spray deposition could improve local, systemic, and CNS drug delivery; however, this has proven to be difficult due to the anatomical features of the nasal cavity, including the nasal valve and turbinate structures. Furthermore, nasal cavity geometries and dimensions vary between individuals based on differences in their age, gender, and ethnicity. The effect of patient-specific administration parameters was evaluated for their ability to overcome the barriers of targeted nasal drug delivery. The nasal spray deposition was evaluated in 10 3D-printed nasal cavity replicas developed based on the CT-scans of five pediatric and five adult subjects. Cromolyn sodium nasal solution, USP, modified with varying concentrations of hypromellose was utilized as a model nasal spray to evaluate the deposition pattern from formulations producing a variety of plume angles. A central composite design of experiments was implemented using the formulation with the narrowest plume angle to determine the patient-specific angle for targeting the turbinate region in each individual. The use of the patient-specific angle with this formulation significantly increased the turbinate deposition efficiency compared to that found for all subjects using an administration angle of 30°, around 90% compared to about 73%. Generally, we found turbinate deposition increased with decreases in the administration angle. Deposition to the upper regions of the replica was poor with any formulation or administration angle tested. Effective turbinate targeting of nasal sprays can be accomplished with the use of patient-specific administration parameters in individuals. Further research is required to see if these parameters can be device-controlled for patients and if other regions can be effectively targeted with other nasal devices.


Subject(s)
Aerosols/administration & dosage , Aerosols/chemistry , Nasal Cavity/drug effects , Administration, Intranasal/methods , Adolescent , Adult , Chemistry, Pharmaceutical/methods , Child , Drug Delivery Systems/methods , Female , Humans , Hypromellose Derivatives/chemistry , Male , Middle Aged , Nasal Sprays , Precision Medicine/methods , Printing, Three-Dimensional , Turbinates/drug effects
3.
J Geriatr Oncol ; 5(1): 65-70, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24484720

ABSTRACT

OBJECTIVE: Elderly patients with cancer are under-represented in clinical trials, and there is especially scant data on their participation in early-phase trials. In an effort to provide more data, we reviewed our Phase I experience. METHODS: We conducted a retrospective analysis of 461 patients enrolled in Phase I clinical trials at the Cancer Therapy Research Center (CTRC) from 2009 to 2011 to determine the rate of completion of at least 12 weeks of treatment, incidence of adverse events, prevalence of co-morbidities, functional status, and survival. Elderly (E) was defined as ≥70 years; non-elderly (NE) was defined as ≤69 years. RESULTS: The elderly represented 15% (69/461) of enrolled patients. The most common malignancies were colon (20%), hematologic (18%), lung (15%), and breast (8%). The median age of E was 72 years (range 70-85, SD 3.15), and 49% of the E was female. Co-morbidities (E vs. NE) include diabetes (28% vs. 23%), hypertension (65% vs. 44%), and chronic kidney disease (91% vs. 48%). Thirty-two percent of E vs. 37% of NE completed at least 12 weeks of treatment. Reasons for not completing in E vs. NE respectively were progression of disease (43% vs. 61%), toxicity (28% vs. 9%), and self-withdrawal (11% vs. 7%). Reasons for not completing the protocol was significantly associated with being elderly (p = 0.005). There were non-significant differences in toxicity in E vs. NE CONCLUSION: Elderly patients have a higher likelihood of not completing trials for reasons including toxicity. This highlights the need for better Phase I trial-designs incorporating ideal geriatric assessment tools.


Subject(s)
Clinical Trials, Phase I as Topic/statistics & numerical data , Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Case-Control Studies , Diabetes Complications/complications , Female , Humans , Hypertension/complications , Male , Middle Aged , Neoplasms/complications , Patient Compliance/statistics & numerical data , Patient Selection , Renal Insufficiency, Chronic/complications , Retrospective Studies , Texas , Young Adult
4.
J Community Med Health Educ ; 3(5): 1000230, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-25152846

ABSTRACT

BACKGROUND: Cancer does not occur in all ethnic and racial groups at similar rates. In addition, responses to treatment also vary in certain ethnic and racial groups. For Hispanics, the overall cancer incidence is generally lower yet for some specific tumor types, the incidence rates are higher compared to other populations. OBJECTIVES: Although disparities are recognized for treatment outcomes and prevention methodologies for Hispanics and other minority populations, specific recruiting and reporting of minorities remains a challenge. In order to circumvent this problem, the Cancer Therapy and Research Center (CTRC) has developed a new minority recruitment plan for all cancer related clinical trials at this Institute. The overall goal of this initiative is to increase the accrual of minorities in cancer clinical trials by implementing several key interventions. METHOD: The Cancer Therapy & Research Center (CTRC) at the University of Texas Health Science Center at San Antonio established the Clinical Trials Accrual Task Force to develop and monitor interventions designed to increase accrual to cancer clinical trials, specifically the accrual of minorities with a focus on the Hispanic population that makes up 68% of the CTRC's catchment area. RESULTS: A Minority Accrual Plan (MAP) was implemented in March 2013 as part of the process for initiating and conducting cancer-related clinical trials at the CTRC. The Minority Accrual Plan focuses on Hispanic enrollment due to the characteristics of the South Texas population served by the CTRC but could be easily adapted to other populations. CONCLUSIONS: The CTRC has designed a process to prospectively address the challenge of deliberately enrolling minority subjects and accurately accounting for the results by implementing a Minority Accrual Plan for every cancer-related clinical trial at CTRC.

5.
Cancer Chemother Pharmacol ; 67(4): 875-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20582417

ABSTRACT

Clofarabine for injection is a second-generation nucleoside analog approved in the United States (Clolar(®)) and Europe (Evoltra(®)) for the treatment of pediatric relapsed or refractory acute lymphoblastic leukemia. This report describes the population pharmacokinetics of clofarabine and its metabolite 6-ketoclofarabine in adult and pediatric patients with hematologic malignancies or solid tumors. Clofarabine pharmacokinetics were best described by a 2-compartment model with linear elimination and first-order absorption after oral administration. Clofarabine was rapidly absorbed following oral administration with a mean absorption time of less than 2 h and bioavailability of 57.5%. The important covariates affecting clofarabine pharmacokinetics were age, weight, and estimated creatinine clearance (eCrCL). No difference in pharmacokinetics was observed between sexes, races, or disease type. The elimination half-life was dependent on all the covariates but was generally less than 7 h in all cases. A difference in clofarabine pharmacokinetics was observed between adults and children. For a pediatric patient 3 years old weighing 16 kg with an eCrCL of 138 mL/min/1.73 m(2), the population estimates for total systemic clearance and volume of distribution at steady-state were 18.3 L/h (1.14 L/h/kg) and 92.9 L (5.81 L/kg), respectively. α- and ß-half-life were 0.9 and 4.4 h, respectively. For an elderly patient 82 years old weighing 96 kg with an eCrCL of 46 mL/min/1.73 m(2), the population estimates for CL and Vdss were 21.5 L/h (0.22 L/h/kg) and 257.4 L (268 L/kg), respectively. α- and ß-half-life were 0.5 and 10.6 h, respectively. Because of the difference in pharmacokinetics, adults have higher exposure than children given a similar dose standardized to body surface area. The exact mechanism of this difference is not understood. As eCrCL decreased, exposure increased due to reduced total systemic clearance. In the case of moderate (eCrCL 30 to 60 mL/min/1.73 m(2)) and severe (eCrCL <30 mL/min/1.73 m(2)) renal impairment, dose reduction may be needed to maintain similar exposure in an equivalent patient of the same age, weight, and normal renal function after both oral and intravenous administration. 6-Ketoclofarabine was a minor metabolite with peak plasma concentrations occurring about 1 h after the start of the infusion and having a metabolite ratio averaging less than 5% and not more than 8% for any particular individual. 6-Ketoclofarabine was rapidly cleared from plasma with an average apparent half-life of 4.9 h (range 3.9 to 6.2 h). No accumulation of 6-ketoclofarabine was observed with predose samples all below the limit of quantification on Days 8 and 15. Further monitoring of 6-ketoclofarabine is not required in future studies.


Subject(s)
Adenine Nucleotides/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Arabinonucleosides/pharmacokinetics , Models, Biological , Neoplasms/drug therapy , Adenine Nucleotides/administration & dosage , Adenine Nucleotides/therapeutic use , Administration, Oral , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Arabinonucleosides/administration & dosage , Arabinonucleosides/therapeutic use , Biological Availability , Body Weight , Child , Child, Preschool , Clofarabine , Creatinine/blood , Creatinine/urine , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Male , Middle Aged , Neoplasms/pathology , Young Adult
6.
Nat Rev Drug Discov ; 5(10): 855-63, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17016426

ABSTRACT

The treatment of acute leukaemias, which are the most common paediatric cancers, has improved considerably in recent decades, with complete response rates approaching approximately 90% in some cases. However, there remains a major need for treatments for patients who do not achieve or maintain complete remission, for whom the prognosis is very poor. In this article, we describe the challenges involved in the discovery and development of clofarabine, a second-generation nucleoside analogue that received accelerated approval from the US FDA at the end of 2004 for the treatment of paediatric patients 1-21 years old with relapsed or refractory acute lymphoblastic leukaemia after at least two prior regimens. It is the first such drug to be approved for paediatric leukaemia in more than a decade, and the first to receive approval for paediatric use before adult use.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Drug Design , Neoplasms/drug therapy , Adenine Nucleotides/pharmacokinetics , Adenine Nucleotides/pharmacology , Arabinonucleosides/pharmacokinetics , Arabinonucleosides/pharmacology , Clinical Trials as Topic , Clofarabine , Drug Approval , Humans , Leukemia, Myeloid, Acute/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
7.
Clin Cancer Res ; 12(17): 5207-15, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951240

ABSTRACT

PURPOSE: To determine the safety, tolerability, and pharmacokinetics and to seek preliminary evidence of anticancer activity of tasidotin (ILX651), a novel dolastatin analogue, when administered as a 30-minute i.v. infusion weekly for 3 weeks every 4 weeks. EXPERIMENTAL DESIGN: Thirty patients with advanced solid malignancies were treated with 82 courses at six dose levels ranging from 7.8 to 62.2 mg/m2 weekly, initially according to an accelerated dose-escalation scheme, which evolved into a Fibonacci scheme as a relevant degree of toxicity was observed. Plasma and urine were sampled to characterize the pharmacokinetic behavior of tasidotin. RESULTS: A high incidence of neutropenia complicated by fever (one patient), or precluding treatment on day 15 (three patients), was the principal toxicity of tasidotin, at doses above 46.8 mg/m2. At all dose levels, nonhematologic toxicities were generally mild to moderate and manageable. Grade 3 toxicities included diarrhea and vomiting (one patient each). Drug-induced neurosensory symptoms were mild and there was no evidence of cardiovascular toxicity, which has been previously associated with other dolastatins. Tasidotin pharmacokinetics were mildly nonlinear, whereas metabolite kinetics were linear. A patient with non-small cell lung carcinoma experienced a minor response, and a patient with hepatocellular carcinoma had stable disease lasting 11 months. CONCLUSIONS: The recommended dose for phase II studies of tasidotin administered on this schedule is 46.8 mg/m2. The mild myelosuppression and manageable nonhematologic toxicities at the recommended dose, the evidence of antitumor activity, and the unique mechanistic aspects of tasidotin warrant further disease-directed evaluations on this and alternative schedules.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Depsipeptides/chemistry , Disease Progression , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Follow-Up Studies , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Molecular Structure , Oligopeptides/adverse effects , Time Factors , Treatment Outcome
8.
Pediatr Blood Cancer ; 47(2): 163-8, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16317728

ABSTRACT

PURPOSE: We performed a Phase I trial of irofulven (MGI 114) to determine the maximum tolerated dose (MTD), the dose-limiting toxicity (DLT), and the plasma pharmacokinetics of irofulven in children < or=21 years of age with refractory/recurrent malignancies. EXPERIMENTAL DESIGN: Thirty-five patients were entered into the study, of whom 34 were eligible. Patients received Irofulven daily x 5 days every 28 days over 10 min, following pre-treatment with ondansetron (0.45 mg/kg) and dexamethasone (12 mg/m(2)). The initial dose of irofulven was 8 mg/m(2) daily, with subsequent escalations to 10, 13, and 17 mg/m(2). Plasma pharmacokinetic samples were obtained in a subset patients. RESULTS: Thirty-two patients were assessable for toxicity, and 30 were assessable for response. In heavily pre-treated patients, dose-limiting thrombocytopenia was observed in two patients at the 8 mg/m(2)/day, and in one patient at the 6 mg/m(2)/day dose level. In less heavily pre-treated patients, proteinuria and elevated creatinine were dose limiting in two patients at the 17 mg/m(2)/day dose level. At 13 mg/m(2)/day, constipation, hyperkalemia with elevated creatinine, and thrombocytopenia was dose limiting in three patients. There were no complete or partial responses. One patient with poorly differentiated carcinoma had stable disease and received 11 courses of therapy. Patients demonstrated a lower systemic exposure and greater clearance than adults treated at similar dose levels. CONCLUSION: The MTD of irofulven administered daily x 5 every 28 days with concomitant ondansetron and dexamethasone is 6 mg/m(2)/day in heavily pre-treated patients and 10 mg/m(2)/day in less heavily pre-treated patients.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Agents, Alkylating/pharmacokinetics , Neoplasms/drug therapy , Sesquiterpenes/adverse effects , Sesquiterpenes/pharmacokinetics , Adolescent , Adult , Antineoplastic Agents, Alkylating/administration & dosage , Child , Child, Preschool , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Male , Maximum Tolerated Dose , Sesquiterpenes/administration & dosage
9.
Clin Cancer Res ; 11(21): 7807-16, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278403

ABSTRACT

PURPOSE: To determine the maximum tolerated dose, dose-limiting toxicity, and pharmacokinetics of the dolastatin-15 analogue, tasidotin (ILX651), when administered i.v. daily for 5 days every 3 weeks. EXPERIMENTAL DESIGN: Thirty-six patients with advanced solid tumors received a total of 114 courses through eight dose levels ranging from 2.3 to 36.3 mg/m(2). Pharmacokinetic samples were collected in cycle 1. RESULTS: Neutropenia was the principal dose-limiting toxicity at 36.3 mg/m(2)/d along with grade 3 ileus and elevated aspartate amino transaminase/alanine amino transaminase (n = 1). At the maximum tolerated dose, 27.3 mg/m(2), 4 of 14 patients experienced dose-limiting grade 4 neutropenia. The other principal toxicities consisted of mild-to-moderate elevated transaminases, alopecia, fatigue, and nausea. One patient with melanoma metastatic to liver and bone treated at 15.4 mg/m(2)/d experienced a complete response and received 20 courses of tasidotin. Two other patients with melanoma had mixed responses of cutaneous metastases at 27.3 mg/m(2)/d associated with either stable or progressive visceral disease. In addition, nine patients had stable disease. There was no accumulation of tasidotin following repeated daily dosing. Tasidotin decayed from plasma in a biphasic fashion with a half-life of <45 minutes in most cases. CONCLUSION: The maximum tolerated dose and recommended phase II dose for tasidotin when administered on this schedule was 27.3 mg/m(2)/d. The favorable toxicity profile of tasidotin compared with other antitubulin agents (particularly the lack of severe cumulative neuropathy, peripheral edema, and fatigue), the observed antitumor activity of tasidotin, and its novel mechanism of action support further disease-directed evaluations of this agent on this 5-day schedule every 3 weeks.


Subject(s)
Antineoplastic Agents/administration & dosage , Depsipeptides/administration & dosage , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/chemistry , Area Under Curve , Depsipeptides/chemistry , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Melanoma/drug therapy , Middle Aged , Neoplasm Metastasis , Oligopeptides/chemistry , Time Factors , Tomography, X-Ray Computed
10.
Clin Cancer Res ; 11(21): 7825-33, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16278405

ABSTRACT

PURPOSE: To determine the maximum tolerated dose (MTD), dose-limiting toxicity (DLT), and pharmacokinetics of tasidotin (ILX651), a dolastatin-15 analogue, when administered on days 1, 3, and 5 every 3 weeks in patients with advanced solid tumors. PATIENTS AND METHODS: Thirty-two patients were treated with 92 courses of tasidotin through seven dose levels determined by a modified Fibonacci scheme ranging from 3.9 to 45.7 mg/m(2). Pharmacokinetic samples were collected during the first course. RESULTS: Neutropenia was the principal DLT at the 45.7 mg/m(2)/d dose level. In addition, one patient also experienced grade 3 neutropenia complicated with grade 3 esophageal candidiasis and grade 3 dehydration. Only 1 of 11 patients treated at the MTD, 34.4 mg/m(2), experienced dose-limiting neutropenia. Other common, drug-related toxicities included mild to moderate fatigue, anemia, nausea, anorexia, emesis, alopecia, and diarrhea. The best observed antitumor response consisted of stable disease and was noted in 10 patients (31%); the median duration on study for those patients with stable disease was 99.5 days compared with 37.5 days for those patients with progressive disease. Tasidotin plasma concentrations declined biphasically with an effective half-life of < or =55 minutes, and approximately 11% was excreted unchanged in the urine. CONCLUSION: The recommended dose for phase II studies and the MTD when tasidotin is administered on days 1, 3, and 5 every 3 weeks is 34.4 mg/m(2). The favorable toxicity profile of tasidotin compared with other antitubulin agents, including other dolastatin analogues, and its novel mechanism of action support further disease-directed evaluation of this agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Adult , Aged , Aged, 80 and over , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Models, Chemical , Time Factors
11.
J Pediatr Hematol Oncol ; 27(3): 166-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15750451

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is the most common myeloproliferative/myelodysplastic disorder seen in children. The treatment of choice, allogeneic stem cell transplantation, provides the only known cure for the disease, but relapse after transplant is common. The authors describe a 5-year-old boy diagnosed at age 34 months with JMML that evolved to acute myeloid leukemia. Initial treatment consisted of fludarabine and cis-retinoic acid therapy, followed by a matched sibling bone marrow transplant. After a relapse, he received a second transplant from the same donor, using peripheral blood stem cells, followed by repeated donor leukocyte infusions. After the second relapse, he received the farnesyltransferase inhibitor R115777 (tipifarnib, Zarnestra), but the leukemia persisted. When bone marrow blasts numbered 60% of the mononuclear cells, he received single-agent clofarabine induction (52 mg/m/d) for 5 days. After three courses, he attained a remission marrow with 5% blasts and disappearance of the 5q- and 9q- cytogenetic abnormalities.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myelomonocytic, Chronic/drug therapy , Adenine Nucleotides , Bone Marrow Transplantation , Child , Clofarabine , Humans , Male , Neoplasm Recurrence, Local/drug therapy , Remission Induction , Treatment Outcome
12.
J Clin Pharmacol ; 44(11): 1309-22, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15496649

ABSTRACT

The population pharmacokinetics of plasma clofarabine and intracellular clofarabine triphosphate were characterized in pediatric patients with acute leukemias. Traditional model-building techniques with NONMEM were used. Covariates were entered into the base model using a forward selection significance level of .05 and a backwards deletion criterion of .005. Model performance, stability, and influence analysis were assessed using the nonparametric bootstrap and n-1 jackknife. Simulations were used to understand the relationship between important covariates and exposure. A 2-compartment model with weight (scaled to a 40-kg reference patient) modeled as a power function on all pharmacokinetic parameters (0.75 on clearance-related terms and 1.0 on volume-related terms) was fit to plasma clofarabine concentrations (n = 32). White blood cell (WBC) count, modeled as a power function (scaled to a WBC count of 10 x 10(3)/microL), was a significant predictor of central volume with power term 0.128 +/- 0.0314. A reference patient had a systemic clearance of 32.8 L/h (27% between-subject variability [BSV]), a central volume of 115 L (56% BSV), an intercompartmental clearance of 20.5 L/h (27% BSV), and a peripheral volume of 94.5 L (39% BSV). Intracellular clofarabine triphosphate concentrations were modeled using a random intercept model without any covariates. The average predicted concentration was 11.6 +/- 2.62 microM (80% BSV), and although clofarabine triphosphate half-life could not be definitively estimated, its value was taken to be longer than 24 hours. The results confirm that clofarabine should continue being dosed on a per-squaremeter or per-body-weight basis.


Subject(s)
Arabinonucleosides/pharmacokinetics , Leukemia, Myeloid, Acute/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adenine Nucleotides , Adolescent , Adult , Arabinonucleosides/administration & dosage , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clofarabine , Computer Simulation , Half-Life , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukocyte Count , Metabolic Clearance Rate , Models, Biological , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Reproducibility of Results
13.
Cancer Res ; 64(9): 3276-80, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15126370

ABSTRACT

Several members of the kinesin family of microtubule motor proteins play essential roles in mitotic spindle function and are potential targets for the discovery of novel antimitotic cancer therapies. KSP, also known as HsEg5, is a kinesin that plays an essential role in formation of a bipolar mitotic spindle and is required for cell cycle progression through mitosis. We identified a potent inhibitor of KSP, CK0106023, which causes mitotic arrest and growth inhibition in several human tumor cell lines. Here we show that CK0106023 is an allosteric inhibitor of KSP motor domain ATPase with a Ki of 12 nM. Among five kinesins tested, CK0106023 was specific for KSP. In tumor-bearing mice, CK0106023 exhibited antitumor activity comparable to or exceeding that of paclitaxel and caused the formation of monopolar mitotic figures identical to those produced in cultured cells. KSP was most abundant in proliferating human tissues and was absent from cultured postmitotic neurons. These findings are the first to demonstrate the feasibility of targeting mitotic kinesins for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Kinesins/antagonists & inhibitors , Pyrimidines/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Female , Humans , Kinesins/metabolism , Mice , Mice, Nude , Xenograft Model Antitumor Assays
14.
J Clin Oncol ; 22(8): 1413-9, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15084615

ABSTRACT

PURPOSE: To determine the dose limiting toxicity (DLT), maximum-tolerated dose (MTD), and pharmacokinetic profile of tirapazamine (Sanofi Synthelabo Research, Malvern, PA) combined with cyclophosphamide in children with recurrent solid tumors. PATIENTS AND METHODS: Patients received a 2-hour infusion of tirapazamine, followed by 1,500 mg/m(2) cyclophosphamide, and mesna once every 3 weeks. Dose escalation of tirapazamine began at 250 mg/m(2) and was increased by 30% in subsequent cohorts. If DLT was hematologic, less-heavily pretreated patients were to be enrolled until their DLTs were encountered, and MTDs defined. Pharmacokinetic profiles were also characterized. RESULTS: Twenty-three patients were enrolled onto the study. Pharmacokinetic data were calculated for 22 patients. Prolonged neutropenia was the DLT at 420 mg/m(2) in heavily pretreated patients. Grade 3, reversible ototoxicity was the DLT in less-heavily pretreated patients at 420 mg/m(2). Two (one with neuroblastoma and one with rhabdomyosarcoma) had partial responses. One child with neuroblastoma had prolonged stable disease (10 cycles) at a dose of 250 mg/m(2). This patient had disease detectable in the bone marrow only and all evidence of bone marrow involvement resolved for 17 cycles of therapy. Four other patients had stable disease. An apparent dose-proportional increase in tirapazamine maximal concentration and area under the curve(last) was observed. Tirapazamine clearance, volume of distribution at steady-state, and terminal half-life did not appear to be dose-dependent. CONCLUSION: The recommended dose of tirapazamine given with 1,500 mg/m(2) of cyclophosphamide once every 3 weeks is 325 mg/m(2). Neutropenia and ototoxicity were dose-limiting. Based on early evidence of antitumor activity, additional studies appear warranted.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/therapeutic use , Neoplasms/drug therapy , Triazines/therapeutic use , Adolescent , Adult , Child , Child, Preschool , Cyclophosphamide/adverse effects , Cyclophosphamide/metabolism , Female , Humans , Male , Neoplasm Recurrence, Local , Neutropenia/chemically induced , Tirapazamine , Triazines/adverse effects , Triazines/metabolism
15.
J Pediatr Hematol Oncol ; 25(7): 539-42, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12847320

ABSTRACT

PURPOSE: To perform a phase 1 trial to determine the maximum tolerated dose and the dose-limiting toxicities of paclitaxel in children with refractory or recurrent solid tumors. Paclitaxel was administered twice weekly, increasing from four to six doses every 21 to 28 days. METHODS: Paclitaxel was administered as a 3-hour intravenous infusion twice weekly. The initial dose was fixed at 50 mg/m2/dose twice weekly for 2 weeks (four doses), every 21 days. The number of twice-weekly doses per course was increased to six in the next cohort. In subsequent cohorts, the number of twice-weekly doses per course was fixed at six, every 28 days, and dosage was increased in 25% increments. RESULTS: Sixteen assessable patients were enrolled at three levels. Neutropenia was the dose-limiting toxicity at 65 mg/m2/dose, twice weekly x 6 doses, every 28 days. Nonhematologic toxicities were minor. No antitumor responses were observed. CONCLUSIONS: Protracted twice-weekly dosing of paclitaxel is limited by neutropenia. The maximum tolerated dose of paclitaxel administered twice weekly x 6 doses, every 28 days, was 50 mg/m2/dose.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Neoplasms/drug therapy , Paclitaxel/adverse effects , Anemia/chemically induced , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Child , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Neoplasms/blood , Neutropenia/chemically induced , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Patient Selection , Platelet Count
16.
J Pharm Sci ; 92(1): 161-72, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12486692

ABSTRACT

The purpose of this study was to systematically evaluate the pharmacokinetic profiles of carbendazim, a novel anticancer drug. Carbendazim reached the highest concentrations in stomach and small intestine by 1 h after oral administration (500 mg/kg) to tumor-bearing nude mice. Four hours later, carbendazim in the large intestine reached maximum concentrations, probably because of pH-induced precipitation of the drug in the large intestine. The highest concentrations of carbendazim in well-perfused tissues, solid tumor, and blood ranged from 63 to 164 microg/g by 4 h. The percentage of carbendazim distributed to solid tumor by 4 h was higher than most well-perfused tissues. Carbendazim concentrations in blood were similar to, or somewhat lower than, those in tumor and other tissues. By 24 h post-dosing, carbendazim concentrations in tissues and blood declined to almost basal levels. The total percentage of administered carbendazim eliminated in urine was 25.7%, and in feces 16.6% within 24 h. Carbendazim exhibited fast permeation across Caco-2 and HT-29 carcinoma cell lines with corresponding permeability coefficients 7.74-8.06 x 10(-5) and 6.8-8.42 x 10(-5) (cm/s). The overall plasma protein binding of carbendazim (0.2-125 microg/mL) assessed by ultrafiltration ranged from 60 to 74%. Comparative pharmacokinetics was conducted in rats by high-pressure liquid chromatography to evaluate the relative bioavailability of carbendazim versus its nanoparticle formulation. Carbendazim and its nanoparticle reached T(max) at 2.01 and 1.57 h, respectively. The relative bioavailability of nanoparticle carbendazim versus regular carbendazim was 166%. High-pressure liquid chromatography analysis of the rat serum obtained at 20 h after oral dosing revealed a carbendazim metabolite, which was identified by mass spectroscopy analysis as 2-aminobenzimidazole, a hydrolyzed product of carbendazim. Incubation of carbendazim with human and rat liver microsomes produced a metabolite identified by mass spectrometry as 5(6)- or 4(7)-hydroxyl carbendazim. The comprehensive pharmacokinetic information is important to the current clinical investigation of carbendazim.


Subject(s)
Benzimidazoles/pharmacokinetics , Carbamates , Cell Membrane Permeability/physiology , Nanotechnology/methods , Animals , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Caco-2 Cells/metabolism , HT29 Cells/metabolism , Humans , Mice , Mice, Nude , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Tissue Distribution/physiology , Xenograft Model Antitumor Assays/methods
17.
Mol Cancer Ther ; 1(8): 565-73, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12479216

ABSTRACT

Cationic porphyrins are being studied as possible anticancer agents because of their ability to bind to and stabilize DNA guanine quadruplexes (G-quadruplexes). We have shown previously that the cationic porphyrin TMPyP4 is able to bind to and stabilize G-quadruplexes in human telomere sequences, resulting in inhibition of telomerase activity. To better understand the mechanism of action behind telomerase inhibition by TMPyP4, we performed a cDNA microarray analysis on cells treated with TMPyP4 and TMPyP2, a positional isomer of TMPyP4 that has low affinity for G-quadruplexes. Analysis of time course data from the microarray experiments revealed that TMPyP4 and TMPyP2 treatment altered the expression of several gene clusters. We found that c-MYC, an oncogene nearly ubiquitous in human tumors that bears the potential in its promoter to form a G-quadruplex, was among the genes specifically down-regulated by TMPyP4, but not by TMPyP2. The hTERT gene, which encodes the catalytic subunit of telomerase, is transcriptionally regulated by c-MYC, and we have found that TMPyP4 also causes a decrease in human telomerase reverse transcriptase transcripts, suggesting two possible mechanisms for the effect of TMPyP4 on telomerase activity. We also show that TMPyP4, but not TMPyP2, is able to prolong survival and decrease tumor growth rates in two xenograft tumor models. We believe that, because of the actions of TMPyP4 in decreasing both c-MYC protein levels and telomerase activity, as well as its anticancer effects in vivo, it is a worthwhile agent to pursue and develop further.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Down-Regulation , Porphyrins/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Telomerase/metabolism , Animals , Blotting, Northern , Blotting, Western , DNA/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins , HeLa Cells , Humans , Mice , Models, Chemical , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
18.
Pharm Res ; 19(8): 1091-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12240933

ABSTRACT

PURPOSE: To compare Caco-2 monolayer permeability and in vivo bioavailability of microparticle with nanoparticle 301029, a thiadiazole derivative, and to determine whether nanonization could improve oral bioavailability of the poorly soluble compound. METHODS: The mean particle size of 301029 was reduced from 7 microm to 280 nm by pearl milling. In the ex vivo assay, both microparticle and nanoparticle 301029 at the same concentration were separately added to apical side and were collected from basolateral side of Caco-2 monolayer. In the bioavailability study, the two particle sizes of 301029 were orally administered to rats, respectively, and blood samples were collected. Nanoparticle 301029 in culture medium and rat serum was detected by a liquid chomatography-mass spectrometer (LC/MS) coupled with atmospheric pressure chemical ionization (APCI). RESULTS: Permeability rate and permeated amounts of nanoparticle 301029 across the Caco-2 monolayer were about four times higher than those of microparticle 301029. In a pharmacokinetic study, nanoparticle 301029 showed Tmax about 1 h, whereas the microparticle 301029 showed Tmax at 4 h. The Cmax and AUC of nanoparticle 301029 were 3- to 4-fold greater than those of microparticle 301029, resulting in a significant increase in oral bioavailability of 301029 as compared with microparticle 301029. The ex vivo permeability and in vivo pharmacokinetic data indicate that nanoparticle formulation improves both absorption rate and absorption extent of the poorly soluble drug. CONCLUSIONS: Nanoparticle formulation enhances both Caco-2 monolayer permeability and rat oral bioavailability of the poorly soluble 301029. The result also demonstrates a close correlation between ex vivo Caco-2 permeability model and in vivo gastrointestinal absorption.


Subject(s)
Nanotechnology/methods , Thiadiazoles/pharmacokinetics , Absorption , Animals , Biological Availability , Caco-2 Cells/metabolism , Chromatography, Liquid/methods , Humans , Mass Spectrometry/methods , Rats , Rats, Sprague-Dawley , Thiadiazoles/chemistry
19.
Clin Cancer Res ; 8(4): 1189-95, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11948132

ABSTRACT

PURPOSE: DNA ligase I is an essential enzyme for completing DNA replication and DNA repair by ligating Okazaki fragments and by joining single-strand breaks formed either directly by DNA-damaging agents or indirectly by DNA repair enzymes, respectively. In this study, we examined whether the DNA ligase I level could be modulated in human tumor cell lines by treatment with gemcitabine (2', 2'-difluoro-2'-deoxycytidine), which is a nucleoside analogue of cytidine with proven antitumor activity against a broad spectrum of human cancers in clinical studies. EXPERIMENTAL DESIGN: To determine the effect of gemcitabine on DNA ligase I expression, Western blot analysis was used to measure the DNA ligase I levels in MiaPaCa, NGP, and SK-N-BE cells treated with different concentrations of gemcitabine and harvested at different time intervals. Cell cycle analysis was also performed to determine the underlying mechanism of DNA ligase I level enhancement in response to gemcitabine. In addition, other agents that share the same mechanism of action with gemcitabine were used to elucidate further details. RESULTS: When different types of tumor cell lines, including MiaPaCa, NGP, and SK-N-BE, were treated with gemcitabine, the level of DNA ligase I increased severalfold despite significant cell growth inhibition. In contrast, other DNA ligases (III and IV) either remained unchanged or decreased with treatment. Cell cycle analysis showed that arrest in S-phase corresponded to an increase of DNA ligase I levels in gemcitabine treated cells. Other agents, such as 1-beta-D-arabinofuranosylcytosine and hydroxyurea, which partly share mechanisms of action with gemcitabine by targeting DNA polymerases and ribonucleotide reductase, respectively, also caused an increase of DNA ligase I levels. However, 5-fluorouracil, which predominantly targets thymidylate synthase, did not cause an increase of DNA ligase I level. CONCLUSIONS: Our results suggest that an arrest of DNA replication caused by gemcitabine treatment through incorporation of gemcitabine triphosphate into replicating DNA and inhibition of ribonucleotide reductase would trigger an increase in DNA ligase I levels in cancer cells. The elevated presence of DNA ligase I in S-phase-arrested cells leads us to speculate that DNA ligase I might have an important role in repairing DNA damage caused by stalled replication forks.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , DNA Ligases/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cytarabine/pharmacology , DNA Ligase ATP , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fluorouracil/pharmacology , Humans , Hydroxyurea/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Gemcitabine
20.
J Pharm Biomed Anal ; 28(1): 65-72, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11861109

ABSTRACT

Carbendazim is a novel anticancer agent. The aim of this study was to prepare and characterize a remote loaded liposome preparation of carbendazim, and compare its pharmacokinetic profile to that of unencapsulated carbendazim. Carbendazim was encapsulated in liposomes composed of sphingomyelin-cholesterol (3:1, w/w) by remote loading in response to a transmembrane pH gradient (pH 0.5 in/pH 4.0 out), which resulted in encapsulation of more than 95% of the available drug in preformed vesicles. High drug/lipid ratios were prepared which correspond to a molar ratio of up to 0.8. Physical isolation of the free drug and dialysis were used to determine the in vitro release of carbendazim from liposomes. The release was independent of the initial drug/lipid ratio and choice of internal buffer composition. Liposomal carbendazim (200 mg kg(-1)) was intravenously administered to athymic nude mice and the serum levels of free carbendazim were determined by HPLC analysis after a methanol-induced protein precipitation. Administration of liposomal carbendazim to mice resulted in significant alterations in the pharmacokinetics. Serum levels of free carbendazim were approximately 10-fold greater than those achieved for the same dose of unencapsulated drug. Liposomal carbendazim showed both high C(max), AUC and low clearance rate. Liposomal carbendazim and unencapsulated carbendazim displayed a similar terminal half-life (43-48 min). The relatively large volume of distribution of carbendazim suggests that the compound may partially enter cells or be bound to some extravascular structures. The results indicate that the liposomal formulation of carbendazim significantly increases its blood concentrations.


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Carbamates , Drug Evaluation, Preclinical/methods , Liposomes/administration & dosage , Liposomes/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/blood , Chemistry, Pharmaceutical , Fungicides, Industrial/administration & dosage , Fungicides, Industrial/blood , Fungicides, Industrial/pharmacokinetics , Injections, Intravenous , Liposomes/blood , Mice , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...