Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Publication year range
1.
Gynecol Oncol ; 160(3): 688-695, 2021 03.
Article in English | MEDLINE | ID: mdl-33390325

ABSTRACT

BACKGROUND: EP0057 (formerly CRLX101) is an investigational nanoparticle-drug conjugate (NDC) of a cyclodextrin-based polymer backbone plus camptothecin, a topoisomerase-1 inhibitor. Prior studies showed efficacy in recurrent or persistent, epithelial ovarian, fallopian tube or primary peritoneal cancer (EOC). METHODS: This phase Ib/2 trial assessed safety and efficacy of EP0057 Q2W plus weekly paclitaxel in patients with EOC. The recommended phase 2 dose (RP2D) was identified using a 3+3 design. The single-arm phase 2 assessed overall response (ORR) per RECIST 1.1 in patients previously treated with bevacizumab. Secondary objectives included progression free survival (PFS) and duration of response. RESULTS: The RP2D was established as 15 mg/m2 EP0057 Q2W plus 80 mg/m2 paclitaxel administered 3 weeks on/1 week off. Nine patients enrolled on phase 1b, with no DLTs; 21 additional patients enrolled on phase 2. All completed >1 cycle. Median age was 62 (44-76) years, 57% ≥3 prior therapies. For the primary analysis, 6/19 patients with prior bevacizumab had confirmed responses (ORR=31.6% (95% CI: 15.4% to 54.0%)) including one complete response (CR). Median PFS was 5.4 months. Most common grade 3/4 adverse events attributed to treatment were decreased neutrophil count (13, 43%) and anemia (3, 10%). CONCLUSIONS: Although the observed ORR was not statistically better than the historical control rate, EP0057 remains an interesting option for treatment of recurrent EOC. EP0057 exhibits high plasma drug retention, slow clearance, and controlled slow release of CPT from the polymer when administered alone and with paclitaxel. (NCT02389985) 242 words.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Fallopian Tube Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , Peritoneal Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents, Phytogenic/pharmacology , Female , Humans , Middle Aged , Paclitaxel/pharmacology , Progression-Free Survival
2.
J Control Release ; 284: 171-178, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29879519

ABSTRACT

Circulating antibodies that specifically bind polyethylene glycol (PEG), a polymer routinely used in protein and nanoparticle therapeutics, have been associated with reduced efficacy and increased adverse reactions to some PEGylated therapeutics. In addition to acute induction of anti-PEG antibodies (APA) by PEGylated drugs, typically low but detectable levels of APA are also found in up to 70% of the general population. Despite the broad implications of APA, the dynamics of APA-mediated clearance of PEGylated drugs, and why many patients continue to respond to PEGylated drugs despite the presence of pre-existing APA, remains not well understood. Here, we developed a minimal physiologically based pharmacokinetic (mPBPK) model that incorporates various properties of APA and PEGylated drugs. Our mPBPK model reproduced clinical PK data of APA-mediated accelerated blood clearance of pegloticase, as well as APA-dependent elimination of PEGyated liposomes in mice. Our model predicts that the prolonged circulation of PEGylated drugs will be compromised only at APA concentrations greater than ~500 ng/mL, providing a quantitative explanation to why the effects of APA on PEGylated treatments appear to be limited in most patients. This mPBPK model is readily adaptable to other PEGylated drugs and particles to predict the precise levels of APA that could render them ineffective, providing a powerful tool to support the development and interpretation of preclinical and clinical studies of various PEGylated therapeutics.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Immunoglobulin G/immunology , Polyethylene Glycols/pharmacokinetics , Urate Oxidase/pharmacokinetics , Animals , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Urate Oxidase/immunology
3.
J Pharm Biomed Anal ; 45(2): 288-94, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17804188

ABSTRACT

Microdialysis is a novel and minimally invasive sampling technique, based on the diffusion of analytes from the interstitial compartment through a semi-permeable membrane, and enables direct assessment of tissue disposition and penetration of drugs. Variable antitumor responses may be associated with differences in tumor vascularity, capillary permeability or tumor interstitial pressure resulting in variable delivery of anticancer agents. In preparation of pharmacokinetic studies, aimed at measuring docetaxel concentrations in healthy and malignant tissues in vivo, in pre-clinical as well as clinical studies, in vitro recovery experiments were performed. In contrast to published data, the recovery experiments suggest that docetaxel has a very low recovery as a result of non-specific binding to currently available microdialysis catheters. Here we discuss our findings with docetaxel in a historical perspective and we report on our experience using polysorbate 80 to eliminate the non-specific binding and its effects on the recovery of docetaxel.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Microdialysis/methods , Taxoids/administration & dosage , Taxoids/pharmacokinetics , Chromatography, Liquid , Clinical Protocols , Docetaxel , Humans , Mass Spectrometry
4.
J Natl Cancer Inst ; 90(7): 505-11, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9539245

ABSTRACT

BACKGROUND: Topotecan is a topoisomerase I inhibitor with activity against xenografts of childhood solid tumors and established clinical activity against neuroblastoma and rhabdomyosarcoma. We have studied the relationship between systemic exposure to and the antitumor activity of topotecan lactone (the active form of the drug) in the xenograft models. Furthermore, we determined whether the responses seen in these models occur at systemic exposure levels that are tolerable in children. METHODS: Neuroblastoma xenografts derived from the tumors of six different patients were established subcutaneously in immune-deprived mice. Topotecan was administered by intravenous bolus injection 5 days a week for 2 consecutive weeks, repeated every 21 days for three cycles. The minimum daily doses that induced complete responses (CRs) and partial responses (PRs) were determined. Topotecan lactone pharmacokinetic studies were performed in both tumor-bearing and nontumor-bearing mice. RESULTS: The minimum doses associated with CRs and PRs in four of the six neuroblastoma xenografts were 0.61 and 0.36 mg/kg body weight, respectively. The topotecan lactone single-day systemic exposures associated with these doses were 88 and 52 ng x hr/mL, respectively. There was an approximately sixfold difference in topotecan lactone systemic exposure (290 ng x hr/mL versus 52 ng x hr/mL) associated with achieving CRs in the least-sensitive and most-sensitive tumors, respectively. CONCLUSIONS: Neuroblastoma xenografts are highly sensitive to topotecan therapy, and responses in mice are achieved at systemic exposures similar to those that are clinically effective and tolerable in children. These results support the concept of deriving preclinical data relating systemic exposure to antitumor activity in xenograft models. Such data may be valuable in making informed decisions regarding the clinical development of new agents.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neuroblastoma/drug therapy , Rhabdomyosarcoma/drug therapy , Topoisomerase I Inhibitors , Topotecan/pharmacology , Adrenal Gland Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Bone Marrow Neoplasms/drug therapy , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Inbred CBA , Retroperitoneal Space , Topotecan/pharmacokinetics , Transplantation, Heterologous
5.
J Clin Oncol ; 16(3): 945-52, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9508177

ABSTRACT

PURPOSE: To determine the maximum-tolerated dose (MTD) and dose-limiting toxicity of topotecan when combined with cyclophosphamide in pediatric patients with recurrent or refractory malignant solid tumors. PATIENTS AND METHODS: A total of 33 patients received cyclophosphamide (250 mg/m2/dose) followed by topotecan in escalating doses (0.6 to 0.75 mg/m2/dose), each given as a 30-minute infusion daily for 5 days. A total of 154 fully assessable treatment courses were given to these patients. RESULTS: Neutropenia was the dose-limiting toxicity of the therapy at both topotecan dose levels. The addition of filgrastim allowed escalation of the topotecan dose to the 0.75-mg/m2 level with acceptable neutropenia. Other significant toxicities were anemia and thrombocytopenia. Nonhematopoietic toxicity of grades > or = 3 was not observed. Responses were reported in patients with Wilms' tumor (one complete response [CR], one partial response [PR]), neuroblastoma (one CR, one PR), rhabdomyosarcoma (one PR), and osteosarcoma (one PR). Pharmacokinetic studies indicate that cyclophosphamide administered on the schedule used in this study did not alter topotecan disposition on day 5. As with previous studies, a pharmacodynamic relation between systemic exposure and myelosuppression was noted. CONCLUSION: The combination of topotecan and cyclophosphamide shows activity in a wide variety of pediatric solid tumors and can be given with acceptable hematopoietic toxicity with the use of filgrastim support. We recommend that pediatric phase II trials use cyclophosphamide 250 mg/m2 followed by topotecan 0.75 mg/m2 daily for 5 days with filgrastim for amelioration of neutropenia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Salvage Therapy , Topotecan/administration & dosage , Adolescent , Child , Child, Preschool , Cyclophosphamide/pharmacokinetics , Drug Administration Schedule , Female , Humans , Infant , Male , Topotecan/pharmacokinetics
6.
J Clin Oncol ; 18(18): 3288-94, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10986062

ABSTRACT

PURPOSE: The sequence in which chemotherapeutic agents are administered can alter their pharmacokinetics, therapeutic effect, and toxicity. We evaluated the pharmacokinetics and pharmacodynamics of docetaxel and topotecan when coadministered on two different sequences of administration. PATIENTS AND METHODS: On cycle 1, docetaxel was administered as a 1-hour infusion at 60 mg/m(2) without filgrastim and at 60, 70, and 80 mg/m(2) with filgrastim on day 1, and topotecan was administered at 0.75 mg/m(2) as a 0.5-hour infusion on days 1 to 4. On cycle 2, topotecan was administered on days 1 to 4, and docetaxel was administered on day 4. Cycles were repeated every 21 days. Blood samples for high-performance liquid chromatography measurement of docetaxel (CL(DOC)) and topotecan (CL(TPT)) total clearance were obtained on day 1 of cycle 1 and day 4 of cycle 2. CL(DOC) and CL(TPT) were calculated using compartmental methods. RESULTS: Mean +/- SD CL(DOC) in cycles 1 and 2 were 75.9 +/- 79.6 L/h/m(2) and 29.2 +/- 17.3 L/h/m(2), respectively (P: <.046). Mean +/- SD CL(TPT) in cycles 1 and 2 were 8.5 +/- 4.4 L/h/m(2) and 9.3 +/- 3.4 L/h/m(2), respectively (P: >. 05). Mean +/- SD neutrophil nadir in cycles 1 and 2 were 4,857 +/- 6, 738/microL and 2,808 +/- 4,518/microL, respectively (P: =.02). CONCLUSION: Administration of topotecan on days 1 to 4 and docetaxel on day 4 resulted in an approximately 50% decrease in docetaxel clearance and was associated with increased neutropenia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Neoplasms/drug therapy , Neoplasms/metabolism , Paclitaxel/analogs & derivatives , Taxoids , Adult , Aged , Docetaxel , Drug Administration Schedule , Drug Interactions , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Leukocyte Count/drug effects , Male , Middle Aged , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Platelet Count/drug effects , Recombinant Proteins , Topotecan/administration & dosage , Topotecan/pharmacokinetics
7.
J Clin Oncol ; 22(16): 3357-65, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15310781

ABSTRACT

PURPOSE: To assess the antitumor efficacy of pharmacokinetically guided topotecan dosing in previously untreated patients with medulloblastoma and supratentorial primitive neuroectodermal tumors, and to evaluate plasma and CSF disposition of topotecan in these patients. PATIENTS AND METHODS: After maximal surgical resection, 44 children with previously untreated high-risk medulloblastoma were enrolled, of which 36 were assessable for response. The topotecan window consisted of two cycles, administered initially as a 30-minute infusion daily for 5 days, lasting 6 weeks. Pharmacokinetic studies were conducted on day 1 to attain a topotecan lactone area under the plasma concentration-time curve (AUC) of 120 to 160 ng/mL.h. After 10 patients were enrolled, the infusion was modified to 4 hours, with dosage individualization. RESULTS: Of 36 assessable patients, four patients (11.1%) had a complete response and six (16.6%) showed a partial response, and disease was stable in 17 patients (47.2%). Toxicity was mostly hematologic, with only one patient experiencing treatment delay. The target plasma AUC was achieved in 24 of 32 studies (75%) in the 30-minute infusion group, and in 58 of 93 studies (62%) in the 4-hour infusion group. The desired CSF topotecan exposure was achieved in seven of eight pharmacokinetic studies when the topotecan plasma AUC was within target range. CONCLUSION: Topotecan is an effective agent against pediatric medulloblastoma in patients who have received no therapy other than surgery. Pharmacokinetically guided dosing achieved the target plasma AUC in the majority of patients. This drug warrants testing as part of standard postradiation chemotherapeutic regimens. Furthermore, these results emphasize the importance of translational research in drug development, which in this case identified an effective drug.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neuroectodermal Tumors, Primitive/drug therapy , Topotecan/pharmacokinetics , Topotecan/therapeutic use , Adolescent , Adult , Antineoplastic Agents/administration & dosage , Area Under Curve , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Cerebellar Neoplasms/pathology , Cerebellar Neoplasms/surgery , Child , Child, Preschool , Female , Humans , Infusions, Intravenous , Male , Medulloblastoma/pathology , Medulloblastoma/surgery , Neuroectodermal Tumors, Primitive/pathology , Neuroectodermal Tumors, Primitive/surgery , Risk Factors , Topotecan/administration & dosage , Treatment Outcome
8.
J Clin Oncol ; 17(6): 1815-24, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10561220

ABSTRACT

PURPOSE: In a preclinical model of neuroblastoma, administration of irinotecan daily 5 days per week for 2 consecutive weeks ([qd x 5] x 2) resulted in greater antitumor activity than did a single 5-day course with the same total dose. We evaluated this protracted schedule in children. PATIENTS AND METHODS: Twenty-three children with refractory solid tumors were enrolled onto a phase I study. Cohorts received irinotecan by 1-hour intravenous infusion at 20, 24, or 29 mg/m(2) (qd x 5) x 2 every 21 days. RESULTS: The 23 children (median age, 14.1 years; median prior regimens, two) received 84 courses. Predominant diagnoses were neuroblastoma (n = 5), osteosarcoma (n = 5), and rhabdomyosarcoma (n = 4). The dose-limiting toxicity was grade 3/4 diarrhea and/or abdominal cramps in six of 12 patients treated at 24 mg/m(2), despite aggressive use of loperamide. The maximum-tolerated dose (MTD) on this schedule was 20 mg/m(2)/d. Five patients had partial responses and 16 had disease stabilization. On day 1, the median systemic exposure to SN-38 (the active metabolite of irinotecan) at the MTD was 106 ng-h/mL (range, 41 to 421 ng-h/mL). CONCLUSION: This protracted schedule is well tolerated in children. The absence of significant myelosuppression and encouraging clinical responses suggest compellingly that irinotecan be further evaluated in children using the (qd x 5) x 2 schedule, beginning at a dose of 20 mg/m(2). These results imply that data obtained from xenograft models can be effectively integrated into the design of clinical trials.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/analogs & derivatives , Neoplasms/drug therapy , Neuroblastoma/drug therapy , Subrenal Capsule Assay , Adolescent , Adult , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/administration & dosage , Camptothecin/adverse effects , Camptothecin/pharmacokinetics , Child , Child, Preschool , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Evaluation , Female , Humans , Irinotecan , Male , Mice , Treatment Outcome
9.
Clin Cancer Res ; 3(3): 423-31, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815701

ABSTRACT

The efficacy of the topoisomerase I inhibitor 7-ethyl-10-(4-[1-piperidino]-1-piperidino)-carbonyloxy-camptotheci n (irinotecan, CPT-11) has been examined against a panel of six independently derived neuroblastoma xenografts. Intensive courses of therapy, where irinotecan was administered i.v. daily 5 days per week for two consecutive weeks [(dx5)2; defined as 1 cycle], were compared to more protracted low-dose schedules where cycles were repeated every 21 days for a total of three courses ¿abbreviated [(dx5)2]3¿. When administered (dx5)2 for a single cycle, the maximum tolerated daily dose was 40 mg/kg. Irinotecan induced a high frequency of complete regressions (CRs) in four of the six lines examined; however, most tumors achieving CR regrew during the period of observation (12 weeks). Furthermore, there was no advantage in high-dose regimens as compared to low dose (10 mg/kg) on the same schedule. Protracted schedules of administration, where three courses of therapy were given at 21-day intervals ¿[(dx5)2]3¿ i.v. were examined at 10 and 5 mg/kg/dose. Even at the lower dose level, irinotecan caused 100% CR in all tumor lines that were maintained at 12 weeks. To determine the minimum dose levels required to induce objective regressions of neuroblastoma xenografts, decreasing doses were examined using the [(dx5)2]3 i.v. schedule. At 2.5 mg/kg/dose, >90% of NB-1643, NB-1691, NB-1382.2, and NB-EB xenografts demonstrated CR, whereas at 1.25 mg/kg/dose, all six tumor lines evaluated demonstrated objective regressions (>/=50% volume reduction), with a high frequency of CRs in four tumor lines. The 10-hydroxy-7-ethyl CPT lactone single-day systemic exposure measured with the minimum dose (2.5 mg/kg) associated with complete response was 198, 257, and 228 ng.h/ml for mice bearing NB-1643, NB-1691, and NB-EB tumors, respectively. These results indicate that childhood neuroblastoma xenografts are highly sensitive to irinotecan given by parenteral administration, and that efficacy is schedule dependent.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Camptothecin/therapeutic use , Cell Division/drug effects , Child, Preschool , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Infant , Injections, Intravenous , Irinotecan , Mice , Mice, Inbred CBA , Neoplasm Recurrence, Local , Neuroblastoma/pathology , Thymectomy , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Clin Cancer Res ; 4(3): 783-9, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9533548

ABSTRACT

Topotecan undergoes both renal and hepatic elimination, with topotecan urinary recovery ranging from 60 to 70%. We evaluated the potential of phenytoin to alter the disposition of topotecan and its N-desmethyl metabolite. A 5-year-old child with high-risk medulloblastoma received the first course of topotecan with phenytoin and the second course without phenytoin. For both courses, topotecan doses were adjusted to achieve a target topotecan lactone plasma area under the curve (AUC). Serial plasma samples were obtained, and lactone and total plasma concentrations of topotecan, as well as total plasma and cerebrospinal fluid concentrations of N-desmethyl topotecan, were measured by high-performance liquid chromatography. Phenytoin coadministration increased lactone and total topotecan clearance from 43.4 +/- 1.9 L/h/m2 to 62.9 +/- 6.4 L/h/m2, and 20.8 +/- 2.8 L/h/m2 to 30.6 +/- 4.1 L/h/m2, respectively (P < 0.05). Concomitant phenytoin increased the plasma AUC of total N-desmethyl topotecan from 7.5 +/- 0.68 ng/ml x h to 16.3 +/- 0.53 ng/ml x h (P < 0.05) at plasma AUC of total topotecan of 226.0 +/- 5.5 ng/ml x h and 240.9 +/- 39.8 ng/ml x h, respectively. N-Desmethyl topotecan penetrated into the cerebrospinal fluid (0.12 +/- 0.01). The patient experienced no grade 3 or 4 toxicity. These are the first data documenting altered topotecan and N-desmethyl topotecan disposition when coadministered with phenytoin and suggests that topotecan may undergo further hepatic metabolism. Although there is an increase in exposure to the active N-desmethyl topotecan metabolite, it is less than the decrease in exposure to topotecan lactone. Therefore, patients concomitantly administered phenytoin may require an increase in topotecan dose to achieve a similar pharmacological effect as a patient not receiving phenytoin.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Phenytoin/therapeutic use , Topotecan/analogs & derivatives , Topotecan/pharmacokinetics , Topotecan/therapeutic use , Anticonvulsants/therapeutic use , Antineoplastic Agents/blood , Cerebellar Neoplasms/blood , Cerebellar Neoplasms/surgery , Child, Preschool , Drug Interactions , Humans , Lactones , Male , Medulloblastoma/blood , Medulloblastoma/surgery , Metabolic Clearance Rate , Models, Biological , Topotecan/blood , Topotecan/cerebrospinal fluid
11.
Clin Cancer Res ; 4(2): 455-62, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9516936

ABSTRACT

The antitumor activity of irinotecan in vitro primarily results from its hydrolysis by carboxylesterase to the active metabolite SN-38. The present study was conducted to evaluate the effect of human neuroblastoma xenografts on irinotecan and SN-38 disposition after i.v. and oral irinotecan administration. Non-tumor-bearing mice and mice bearing three different human neuroblastoma xenograft lines (NB1691, NB1643, and NBEB) were given irinotecan (10 mg/kg) by short i.v. injection into the tail vein or by oral gavage. Serial plasma samples were obtained, processed to isolate irinotecan and SN-38 lactone, and assayed with a sensitive and specific high-performance liquid chromatography assay. Noncompartmental and compartmental pharmacokinetic analyses were performed. A four-compartment model was used for analysis of irinotecan and SN-38 concentration-time data after i.v. administration. The presence of tumor increased irinotecan systemic exposure (1.2-3.8-fold; P < 0.05) after i.v. and oral administration in mice bearing neuroblastoma xenografts compared to non-tumor-bearing mice. Moreover, SN-38 systemic exposures were higher (1.3-3.8-fold; P < 0.05) in mice bearing human neuroblastoma xenografts as compared to non-tumor-bearing mice, with the greatest effect observed after oral administration of irinotecan. A schematic model is presented to provide a mechanistic basis for our observations. These results emphasize the need to perform preclinical pharmacokinetic studies to evaluate the influence of tumor on drug disposition.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Neuroblastoma/metabolism , Prodrugs/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/blood , Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/blood , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Female , Humans , Injections, Intravenous , Irinotecan , Male , Mice , Mice, Inbred CBA , Neoplasm Transplantation , Neuroblastoma/blood , Neuroblastoma/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Clin Cancer Res ; 6(3): 813-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741701

ABSTRACT

Irinotecan (IRN), a topoisomerase I interactive agent, has significant antitumor activity in early Phase I studies in children with recurrent solid tumors. However, the disposition of IRN and its metabolites, SN-38 and APC, in children has not been reported. Children with solid tumors refractory to conventional therapy received IRN by a 1-h i.v. infusion at either 20, 24, or 29 mg/m2 daily for 5 consecutive days for 2 weeks. Serial blood samples were collected after doses 1 and 10 of the first course. IRN, SN-38, and APC lactone concentrations were determined by an isocratic high-performance liquid chromatography assay. A linear four-compartment model was fit simultaneously to the IRN, SN-38, and APC plasma concentration versus time data. Systemic clearance rate for IRN was 58.7 +/- 18.8 liters/h/m2 (mean +/- SD). The mean +/- SD ng/ml x h single-day lactone SN-38 area under the concentration-time curve (AUC(0-->6) was 90.9 +/- 96.4, 103.7 +/- 62.4, and 95.3 +/- 63.9 at IRN doses of 20, 24, and 29 mg/m2, respectively. The relative extent of IRN conversion to SN-38 and metabolism to APC measured after dose 1 were 0.49 +/- 0.33 and 0.29 +/- 0.17 (mean +/- SD). No statistically significant intrapatient difference was noted for SN-38 area under the concentration-time curve. Large interpatient variability in IRN and metabolite disposition was observed. The relative extent of conversion and the SN-38 systemic exposure achieved with this protracted schedule of administration were much greater than reported in adults or children receiving larger intermittent doses.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Neoplasms/drug therapy , Adolescent , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Area Under Curve , Camptothecin/blood , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Child , Child, Preschool , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Irinotecan , Neoplasm Recurrence, Local , Neoplasms/pathology , Neoplasms, Complex and Mixed/drug therapy , Neoplasms, Complex and Mixed/pathology , Neoplasms, Connective and Soft Tissue/drug therapy , Neoplasms, Connective and Soft Tissue/pathology , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/pathology , Neutropenia/chemically induced , Time Factors
13.
Clin Cancer Res ; 4(10): 2537-44, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9796988

ABSTRACT

The purpose of this study was to define the length of topotecan (TPT) i.v. infusion necessary to attain a cytotoxic exposure for medulloblastoma cells throughout the neuraxis. In vitro studies of human medulloblastoma cell lines (Daoy, SJ-Med3) were used to estimate the length and extent of TPT systemic exposure associated with inhibition of tumor cell growth or the exposure duration threshold (EDT). We evaluated TPT systemic and cerebrospinal fluid (CSF) disposition in six male rhesus monkeys (8-12 kg) that received TPT 2.0 mg/m2 i.v. as a 30-min or 4-h infusion. Plasma and CSF samples were assayed for TPT lactone by high-performance liquid chromatography, and the CSF exposures were compared with the estimated EDT. Results of the in vitro studies defined an EDT as a TPT lactone concentration of > 1 ng/ml for 8 h (IC99) daily for 5 days. The mean +/- SD for systemic clearance (CL(SYS)), penetration into fourth ventricle (%CSF(4th)), and penetration into lumbar space (%CSF(LUM)) were similar for the 30-min and the 4-h infusions. At a TPT lactone systemic exposure (AUC(PL)) of 56.7 +/- 19.9 ng/ml x h, time above 1 ng/ml in the fourth ventricle was 1.4-fold greater for a 4-h infusion compared with a 30-min infusion. At a TPT lactone AUC(PL) of 140 ng/ml x h, the 4-h infusion achieved the desired TPT exposure throughout the neuraxis (lateral and fourth ventricles and lumbar space), whereas the 30-min infusion failed to achieve it in the lumbar space. In conclusion, prolonging TPT i.v. infusion from 30-min to 4-h at a targeted AUC(PL) achieves the EDT throughout the neuraxis and represents an alternative method of TPT administration that will be tested prospectively in patients with high-risk medulloblastoma.


Subject(s)
Antineoplastic Agents/administration & dosage , Medulloblastoma/drug therapy , Topotecan/administration & dosage , Animals , Cerebral Ventricles/metabolism , Child , Humans , Infusions, Intravenous , Lumbosacral Region , Macaca mulatta , Male , Topotecan/pharmacokinetics , Tumor Cells, Cultured
14.
Clin Cancer Res ; 4(3): 743-53, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9533544

ABSTRACT

Irinotecan, administered i.v. on days 1-5 and 8-12 [(dx5)2 i.v.] has demonstrated significant activity against advanced human tumor xenografts. To explore the feasibility of prolonged oral administration of irinotecan, we compared the efficacy of oral and i.v. irinotecan on the (dx5)2 schedule. We also evaluated oral therapy for 12 consecutive weeks [(dx5)12] at 25 and 50 mg/kg and two consecutive 5-day courses repeated every 21 days for up to four cycles ([(dx5)2]4) at 50 and 75 mg/kg/dose in a series of human colon carcinoma xenograft lines. In addition, we evaluated the effect of a sensitive (HC1) and resistant (ELC2) human colon adenocarcinoma xenograft on irinotecan and SN-38 lactone disposition after administration of irinotecan 10 mg/kg i.v. and 10 and 25 mg/kg p.o. Irinotecan i.v. at 40 mg/kg and oral at 50 and 75 mg/kg on the (dx5)2 schedule had similar activity against the panel of adult colon adenocarcinoma xenografts. Irinotecan given p.o. also demonstrated significant activity against a topotecan-resistant derivative, VRC5/TOPO. Oral administration of 75 mg/kg [(dx5)2]4 and 50 mg/kg (dx5)12 achieved complete response in five of seven xenograft lines evaluated. After i.v. administration, mice bearing HC1 xenografts had 43% greater SN-38 lactone systemic exposure compared to those with ELC2 xenografts and non-tumor-bearing mice. After oral (10 mg/kg) administration, there was a 5-fold higher molar formation of SN-38 lactone compared to i.v. (10 mg/kg) administration in tumor and non-tumor-bearing mice. SN-38 systemic exposure associated with the lowest oral dose (25 mg/kg) achieving complete response for HC1 was 942.6 ng/ml x h. These results emphasize the importance of pharmacokinetic studies as part of tumor response studies in xenograft models.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Colonic Neoplasms/drug therapy , Adenocarcinoma/blood , Adenocarcinoma/pathology , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/therapeutic use , Cell Division/drug effects , Colonic Neoplasms/blood , Colonic Neoplasms/pathology , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Injections, Intravenous , Irinotecan , Metabolic Clearance Rate , Mice , Mice, Inbred CBA , Models, Biological , Neoplasm Transplantation , Thymectomy , Transplantation, Heterologous
15.
Clin Cancer Res ; 7(8): 2301-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489805

ABSTRACT

Pharmacodynamic measures of neutropenia, such as absolute neutrophil count at nadir and neutrophil survival fraction, may not reflect the overall time course of neutropenia. We developed a pharmacokinetic-pharmacodynamic model to describe and quantify the time course of neutropenia after administration of topotecan to children and to compare this with nonhuman primates (NHPs) as a potential preclinical model of neutropenia. Topotecan was administered as a 30-min infusion daily for 5 days, repeated every 21 days. As part of a Phase I Pediatric Oncology Group study, topotecan was administered at 1.4 and 1.7 mg/m(2)/day without filgrastim (POG), and at 1.7, 2, and 2.4 mg/m(2)/day with filgrastim (POG+G). In NHPs, topotecan was administered at 5, 10, and 20 mg/m(2)/day without filgrastim. A pharmacokinetic-pharmacodynamic model was fit to profiles of topotecan lactone plasma concentrations and neutrophil survival fraction from cycle 1 and used to calculate topotecan lactone area under the plasma concentration-versus-time curve from 0 to 120 h (AUC(LAC)) and the area between the baseline and treatment-related neutrophil survival fraction (ABC) from 0 to 700 h. The mean +/- SD neutrophil survival fraction at nadir for the POG, POG+G, and NHP groups was 0.12 +/- 0.09, 0.11 +/- 0.17, and 0.09 +/- 0.08, respectively (P > 0.05). The mean +/- SD for the ratio of ABC to AUC(LAC) for the POG and NHP groups was 1.02 +/- 0.38 and 0.16 +/- 0.09, respectively (P < 0.05). The model estimate of ABC and the ratio of ABC to AUC(LAC) in children and NHPs may better reflect sensitivity to chemotherapy-induced neutropenia.


Subject(s)
Neutropenia/pathology , Topotecan/pharmacokinetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Child , Child, Preschool , Clinical Trials, Phase I as Topic , Disease Models, Animal , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Male , Metabolic Clearance Rate , Neoplasms/drug therapy , Neoplasms/metabolism , Neutropenia/chemically induced , Neutropenia/metabolism , Neutrophils/drug effects , Recombinant Proteins , Time Factors , Topoisomerase I Inhibitors , Topotecan/administration & dosage , Topotecan/adverse effects
16.
Genes Brain Behav ; 14(3): 271-80, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25727211

ABSTRACT

Initial sensitivity to psychostimulants can predict subsequent use and abuse in humans. Acute locomotor activation in response to psychostimulants is commonly used as an animal model of initial drug sensitivity and has been shown to have a substantial genetic component. Identifying the specific genetic differences that lead to phenotypic differences in initial drug sensitivity can advance our understanding of the processes that lead to addiction. Phenotyping inbred mouse strain panels are frequently used as a first step for studying the genetic architecture of complex traits. We assessed locomotor activation following a single, acute 20 mg/kg dose of cocaine (COC) in males from 45 inbred mouse strains and observed significant phenotypic variation across strains indicating a substantial genetic component. We also measured levels of COC, the active metabolite, norcocaine and the major inactive metabolite, benzoylecgonine, in plasma and brain in the same set of inbred strains. Pharmacokinetic (PK) and behavioral data were significantly correlated, but at a level that indicates that PK alone does not account for the behavioral differences observed across strains. Phenotypic data from this reference population of inbred strains can be utilized in studies aimed at examining the role of psychostimulant-induced locomotor activation on drug reward and reinforcement and to test theories about addiction processes. Moreover, these data serve as a starting point for identifying genes that alter sensitivity to the locomotor stimulatory effects of COC.


Subject(s)
Cocaine-Related Disorders/genetics , Locomotion/drug effects , Locomotion/genetics , Mice, Inbred Strains/genetics , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cocaine/pharmacokinetics , Cocaine-Related Disorders/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Animal , Motor Activity/drug effects , Motor Activity/genetics
17.
Clin Pharmacokinet ; 32(4): 324-43, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9113440

ABSTRACT

Cancer chemotherapy doses are empirical in that the majority are administered at a fixed dose (mg/m2 or mg/kg). One reason for this is the intrinsic sensitivity of the tumour or host cells to one particular chemotherapy agent is unknown. Therefore, the likelihood of response or toxicity is unpredictable a priori. This contrasts with antimicrobial chemotherapy where sensitivity (minimum inhibitory concentration) can be determined for a specific bacterium. The pharmacokinetics of cancer chemotherapy agents is also highly variable between patients. In addition, the small therapeutic index of these drugs, combined with the lack of good surrogate markers of toxicity or response, adds to the empiricism of the administration of cancer chemotherapy. In the past few years, numerous studies have established good relationships between systemic exposure to cancer chemotherapy and both response and toxicity. These relationships have been used to individualise chemotherapy dose administration a priori and a posteriori. Some examples of drugs which are individualised based on their pharmacokinetics are methotrexate, busulfan and carboplatin. Other examples of antineoplastic agents which may eventually be individualised based on their pharmacokinetics are mercaptopurine, fluorouracil, etoposide and teniposide, topotecan and suramin. New strategies are being investigated to improve the therapeutic index of cancer chemotherapy agents such as biomodulation, pharmacogenetics, circadian administration and the modification of drug scheduling. Pharmacokinetic studies have also played a major role in these areas. Thus, despite the empiricism associate with cancer chemotherapy administration, some progress has been made and shown to have an impact on outcome. However, more studies are needed to improve cancer chemotherapy administration.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasms/drug therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Circadian Rhythm/physiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Genotype , Humans , Structure-Activity Relationship , Treatment Outcome
18.
Cancer Chemother Pharmacol ; 47(1): 89-93, 2001.
Article in English | MEDLINE | ID: mdl-11221968

ABSTRACT

The pharmacokinetics of topotecan have been extensively studied in patients with normal renal function and there is one study of patients with mild to moderate renal insufficiency. However, the effect of hemodialysis on topotecan disposition has not been reported. The objective of this study was to characterize the disposition of topotecan in a patient with severe renal insufficiency receiving hemodialysis. Topotecan lactone disposition was characterized in a patient on and off hemodialysis. The topotecan lactone clearance determined after administration of topotecan alone and with hemodialysis was 5.3 l/h per m(2) vs 20.1 l/h per m2 respectively. At 30 min after the completion of hemodialysis, the topotecan plasma concentration obtained was greater than that measured at the end of hemodialysis (i.e. 8.0 ng/ml vs 4.9 ng/ml), suggesting a rebound effect. The topotecan terminal half-life off dialysis was 13.6 h, compared with an apparent half-life determined during hemodialysis of 3.0 h. These results demonstrate that topotecan plasma clearance while on hemodialysis increased approximately fourfold. Hemodialysis may be an effective systemic clearance process for topotecan and should be considered in selected clinical situations (e.g. inadvertent overdose, severe renal dysfunction).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Ovarian Neoplasms/metabolism , Renal Dialysis , Renal Insufficiency/metabolism , Topotecan/pharmacokinetics , Female , Humans , Middle Aged , Ovarian Neoplasms/drug therapy , Renal Insufficiency/etiology
19.
Cancer Chemother Pharmacol ; 40(3): 259-65, 1997.
Article in English | MEDLINE | ID: mdl-9219511

ABSTRACT

The present study was conducted to quantitate the disposition of irinotecan lactone and its active metabolite SN-38 lactone in mice following oral and intravenous administration, and to evaluate the systemic exposure of irinotecan lactone and SN-38 lactone associated with antitumor doses of irinotecan lactone in mice bearing human tumor xenografts. Nontumor-bearing mice were given a single oral or intravenous irinotecan dose (5, 10, 40, or 75 mg/kg), and serial plasma samples were subsequently obtained. Irinotecan and SN-38 lactone plasma concentrations were measured using an isocratic HPLC assay with fluorescence detection. The disposition of intravenous irinotecan lactone was modeled using a two-compartment pharmacokinetic model, and the disposition of oral irinotecan and SN-38 lactone was modeled with noncompartmental methods. Irinotecan lactone showed biphasic plasma disposition following intravenous dosing with a terminal half-life ranging between 1.1 to 3 h. Irinotecan lactone disposition was linear at lower doses (5 and 10 mg/kg), but at 40 mg/kg irinotecan lactone clearance decreased and a nonlinear increase in irinotecan lactone AUC was observed. The steady-state volume of distribution ranged from 19.1 to 48.1 l/m2. After oral dosing, peak irinotecan and SN-38 lactone concentrations occurred within 1 h, and the irinotecan lactone bioavailability was 0.12 at 10 mg/kg and 0.21 at 40 mg/kg. The percent unbound SN-38 lactone in murine plasma at 1000 ng/ml was 3.4 +/- 0.67%, whereas at 100 ng/ml the percent unbound was 1.18 +/- 0.14%. Irinotecan and SN-38 lactone AUCs in micebearing human neuroblastoma xenografts were greater than in nontumor-bearing animals. Systemic exposure to unbound SN-38 lactone in nontumor-bearing animals after a single oral irinotecan dose of 40, 10, and 5 mg/kg was 28.3, 8.6, and 2.9 ng h/ml, respectively. Data from the present study provide important information for the design of phase I studies of oral irinotecan.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacokinetics , Camptothecin/analogs & derivatives , Administration, Oral , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Area Under Curve , Blood Proteins/metabolism , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Female , Injections, Intravenous , Irinotecan , Mice , Mice, Inbred CBA , Neoplasm Transplantation , Neuroblastoma , Protein Binding
20.
Cancer Chemother Pharmacol ; 43(4): 269-76, 1999.
Article in English | MEDLINE | ID: mdl-10071976

ABSTRACT

PURPOSE: We have reported a 6-fold difference in the topotecan (TPT) lactone systemic exposure achieving a complete response in the human neuroblastoma xenografts NB-1691 and NB-1643. However, the relationship between tumor extracellular fluid (ECF) exposure to TPT and the antitumor activity in xenograft and in vitro models has not been established. METHODS: TPT was given i.v. to mice bearing NB-1691 and NB-1643 tumors. Prior to dosing, microdialysis probes were placed in tumors of mice bearing NB-1691 and NB-1643 tumors. Plasma and tumor ECF concentrations of TPT lactone were assayed by high performance liquid chromatography. The inhibitory concentration (IC50) was determined for NB-1691 and NB-1643 cell lines in vitro. RESULTS: The TPT AUC(ECF) values determined for NB-1691 (n = 10) and NB-1643 (n = 11) were 7.3 +/- 0.84 and 25.6 +/- 0.76 ng h ml(-1), respectively (P < 0.05). TPT tumor ECF penetration in NB-1691 and NB-1643 was 0.04 +/- 0.04 and 0.15 +/- 0.11 (P < 0.05), respectively. The IC50 values recorded after 6 h of TPT exposure daily for 5 consecutive days for NB-1691 and NB-1643 were 2.7 +/- 1.1 and 0.53 +/- 0.19 ng/ml, respectively (P < 0.05). CONCLUSIONS: NB-1643 was more sensitive in vitro than NB-1691, and at similar plasma TPT exposures, NB-1643 had a greater degree of TPT tumor ECF exposure and penetration as compared with NB-1691. Potential factors affecting tumor TPT ECF disposition include tumor vascularity, capillary permeability, and interstitial pressure. The clinical importance of this study is underscored by the need to select anticancer agents with a high capacity for tumor penetration and to optimize drug administration to increase tumor penetration.


Subject(s)
Extracellular Space/drug effects , Neuroblastoma/drug therapy , Topotecan/therapeutic use , Animals , Chromatography, High Pressure Liquid , Female , Humans , Immunosuppression Therapy/methods , Mice , Mice, Inbred CBA , Microdialysis , Models, Biological , Neuroblastoma/pathology , Neuroblastoma/physiopathology , Thymectomy , Tissue Distribution , Topotecan/pharmacokinetics , Topotecan/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL