Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Peter J. Houghton is active.

Publication


Featured researches published by Peter J. Houghton.


Journal of Clinical Oncology | 1999

Direct Translation of a Protracted Irinotecan Schedule From a Xenograft Model to a Phase I Trial in Children

Wayne L. Furman; Clinton F. Stewart; Catherine A. Poquette; Charles B. Pratt; Victor M. Santana; William C. Zamboni; Laura C. Bowman; Margaret K. Ma; Fredrick A. Hoffer; William H. Meyer; Alberto S. Pappo; Andrew W. Walter; Peter J. Houghton

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.


Journal of Clinical Oncology | 2005

Improved Response in High-Risk Neuroblastoma With Protracted Topotecan Administration Using a Pharmacokinetically Guided Dosing Approach

Victor M. Santana; Wayne L. Furman; Catherine A. Billups; Fredric A. Hoffer; Andrew M. Davidoff; Peter J. Houghton; Clinton F. Stewart

PURPOSE To estimate the response rate and toxicity associated with intravenous topotecan when it is administered on a protracted schedule according to a pharmacokinetically guided dosing approach to treat childhood high-risk neuroblastoma. PATIENTS AND METHODS In this prospective phase II trial, topotecan was administered intravenously daily for 5 days for each of 2 consecutive weeks for two cycles. On the basis of topotecan systemic clearance, doses were individualized to attain a single-day topotecan lactone area under the plasma concentration-time curve (AUC) of 80 to 120 ng/mL . h. Patients subsequently received standard treatment. RESULTS Both cycles were administered to 28 (93%) of the 30 enrolled patients (median age, 3.1 years). Target topotecan AUCs were achieved in 92 (72%) of the 127 measurements conducted after pharmacokinetically guided adjustment; the median dosage required to achieve target AUCs was 2.7 mg/m(2) (range, 0.95 to 3.8 mg/m(2)). The response rate was 60% (95% CI, 41% to 77%); there were one complete and 17 partial responses. No patient experienced disease progression during initial topotecan therapy. Primary tumor volumes decreased (median decrease, -58.2%; range, -95.1% to -4.9%) in the 26 patients with available size data. Homovanillic acid levels in 16 (89%) of 18 patients and vanillylmandelic acid levels in 14 (78%) of 18 patients were lower (P = .002 and P = .018, respectively) after topotecan therapy. Reversible grade 4 myelosuppression occurred in all patients, but no deaths occurred as a result of infection or toxicity. CONCLUSION Topotecan is active against neuroblastoma when it is administered on a protracted schedule and targeted systemic exposure is achieved.


Anti-Cancer Drugs | 1997

Efficacy of oral irinotecan against neuroblastoma xenografts

Joyce Thompson; William C. Zamboni; Pamela J. Cheshire; Lois B. Richmond; Xiaolong Luo; Janet A. Houghton; Clinton F. Stewart; Peter J. Houghton

The efficacy of the topoisomerase I inhibitor, 7-ethyl-10-(4-[1-piperidino]-1-piperidino)-carbonyloxy-camptotheci n (irinotecan, CPT-11), administered by oral gavage has been examined against a panel of six independently derived neuroblastoma xenografts. Irinotecan was administered either daily for 5 days on 12 consecutive weeks ¿(d x 5)12¿ or for 5 days on two consecutive weeks repeated every 21 days for 4 cycles ¿[(d x 5)2]4¿. Given on the (d x 5)12 schedule the maximum tolerated dose (MTD) was 50 mg/kg. For intermittent scheduling ¿[(d x 5)2]4¿, the MTD was 75 mg/kg, resulting in the same total dose being administered (3 g/kg) over the period of treatment. At the MTD for the 12 consecutive week schedule there were two of 42 toxicity related deaths, whereas intermittent scheduling at the MTD resulted in none of 42 deaths. The intermittent schedule ¿[(d x 5)2]4¿ was less toxic than therapy given (d x 5)12, as at the end of treatment mice weighed 92 +/- 4% (SD; n = 6 experiments) and 81 +/- 4% (SD; n = 6 experiments) of their body weight at the start of therapy, respectively. The latter schedule was associated with loose feces starting around week 8 of therapy, broken teeth and a high incidence of swelling of the orbital conjunctiva that developed late in the course of therapy. Given (d x 5)12, irinotecan caused complete regressions of all six neuroblastoma xenograft lines. Because mice tolerate significantly greater systemic exposure to SN-38 lactone than do patients (as determined by plasma AUC at the respective MTD), we evaluated the intermittent schedule of administration, reducing the dose/administration to determine the lowest dose levels that produced objective regressions of these neuroblastoma xenografts and determined the daily systemic exposure associated with these dose levels. In four lines examined objective responses were obtained at dose levels of 12.5 or 6.25 mg/kg. The daily plasma AUC exposures associated with minimum dose achieving response in NB1691 (12.5 mg/kg), NB1643 (6.25 mg/kg) and NBEB (12.5 mg/kg) for irinotecan lactone were 219, 152 and 653 ng-h/ml, respectively; and for SN-38 lactone were 704, 418 and 987 ng-h/ml, respectively. These results indicate that childhood neuroblastoma xenografts are highly sensitive to irinotecan given by oral administration and therapeutic activity is similar to i.v. irinotecan administered on similar schedules.


Cancer Chemotherapy and Pharmacology | 1999

Relationship between tumor extracellular fluid exposure to topotecan and tumor response in human neuroblastoma xenograft and cell lines

William C. Zamboni; Peter J. Houghton; Jeff L. Hulstein; Mark N. Kirstein; Jessica Walsh; Pam J. Cheshire; Suzan K. Hanna; Mary K. Danks; Clinton F. Stewart

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 AUCECF 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.


Journal of Clinical Oncology | 2004

Phase I and Pharmacokinetic Study of Topotecan Administered Orally Once Daily for 5 Days for 2 Consecutive Weeks to Pediatric Patients With Refractory Solid Tumors

Najat C. Daw; Victor M. Santana; Lisa C. Iacono; Wayne L. Furman; Dana R. Hawkins; Peter J. Houghton; J. Carl Panetta; Amar Gajjar; Clinton F. Stewart

PURPOSE We conducted a phase I trial of the injectable formulation of topotecan given orally once daily for 5 days for 2 consecutive weeks (qd x 5 x 2) in pediatric patients with refractory solid tumors. PATIENTS AND METHODS Cohorts of two to six patients received oral topotecan at 0.8, 1.1, 1.4, 1.8, and 2.3 mg/m(2)/d every 28 days for a maximum of six courses. Twenty patients (median age, 10.6 years) received a total of 51 courses. Eight patients received topotecan capsules during course 2 only. RESULTS Dose-limiting toxicity occurred at 2.3 mg/m(2)/d and consisted of prolonged grade 4 neutropenia (n = 2), grade 3 stomatitis as a result of radiation recall (n = 1), grade 3 hemorrhage (epistaxis) in the presence of grade 4 thrombocytopenia (n = 1), and grade 3 diarrhea in the presence of Clostridium difficile infection (n = 1). Dose-limiting, prolonged grade 4 neutropenia and thrombocytopenia occurred in one patient at 1.4 mg/m(2)/d. Infrequent toxicities were mild nausea, vomiting, elevated liver ALT or AST, and rash. The maximum-tolerated dosage was 1.8 mg/m(2)/d; the mean (+/- standard deviation) area under the plasma concentration-time curve for topotecan lactone at this dosage was 20.9 +/- 8.4 ng/mL. h. The population mean (+/- standard error) oral bioavailability of the injectable formulation was 0.27 +/- 0.03; that of capsules was 0.36 +/- 0.06 (P =.16). Disease stabilized in nine of 19 assessable patients for 1.5 to 6 months. CONCLUSION Oral topotecan (1.8 mg/m(2)/d) on a qd x 5 x 2 schedule is well tolerated and warrants additional testing in pediatric patients.


Clinical Pharmacology & Therapeutics | 1996

Interpatient variability in oral (PO) absorption of topotecan (TPT) in children with relapsed solid tumors

William C. Zamboni; William R. Crom; Laura C. Bowman; Charles B. Pratt; Peter J. Houghton; Clinton F. Stewart

Clinical Pharmacology & Therapeutics (1996) 59, 198–198; doi: 10.1038/sj.clpt.1996.293


Clinical Pharmacology & Therapeutics | 2004

Cefixime (CFX) enables higher plasma exposures of SN‐38 to be achieved in pediatric patients receiving oral irinotecan (IRN)

K. R. Crews; Clinton F. Stewart; John C. Panetta; Peter J. Houghton; Victor M. Santana; Wayne L. Furman

The mechanism of IRN‐induced diarrhea has been attributed to local deglucuronidation by enteric bacteria of the metabolite SN‐38G to the active metabolite SN‐38. In preclinical studies, administration of oral antibiotics with IRN reduced diarrhea. Based on these observations, we performed a Phase I study of the intravenous (IV) formulation of IRN, given orally daily x 5 days (d) for 2 weeks (qdx5x2) in children with refractory solid tumors and evaluated whether concomitant CFX use ameliorates diarrhea and allows further dose‐escalation of IRN. We determined the pharmacokinetics (PK) of IRN given both IV and orally in patients who received (n=11) or did not receive CFX (n=20), at a dosage of 8 mg/kg/d, beginning 5 d prior to the start of oral IRN and continuing for the entire 21‐d course. Cohorts of 3 to 7 children received dosages of IRN from 15 to 45 mg/m2/dx5x2 without CFX and from 45 to 75 mg/m2/dx5x2 with CFX. Diarrhea was dose‐limiting at 45 mg/m2/d without CFX. The population mean and interpatient variance (CV%) IRN lactone clearance was 42.4 L/h/m2 (58%) and bioavailability (F) was 8.2% (94%) across all dosages studied. IRN clearance and F were not significantly altered with CFX co‐administration (p>0.5). Plasma SN‐38 lactone AUC increased with respect to dose over the entire dosage range (p=0.001). Median (range) plasma SN‐38 lactone AUC was 44 (18–77) ng*h/mL at the highest dose level (75mg/m2/d) with CFX (n=4, enrollment at this level ongoing) compared to 12 (4–17) ng*h/mL at the MTD without CFX (40 mg/m2/d; n=7). Fecal beta‐glucuronidase activity decreased after 5 d of CFX administration in 3 of 4 patients studied. Plasma total SN38G AUC:SN‐38 AUC ratio was not altered with the administration of CFX (p>0.5). Concomitant use of CFX with oral IRN allows higher dosages of IRN to be given, which results in higher plasma exposures to the active topoisomerase‐I inhibitor SN‐38.


Clinical Cancer Research | 1997

Efficacy of systemic administration of irinotecan against neuroblastoma xenografts.

Joyce Thompson; William C. Zamboni; Pamela J. Cheshire; Lois Lutz; Xiaolong Luo; Yulan Li; Janet A. Houghton; Clinton F. Stewart; Peter J. Houghton


Clinical Cancer Research | 2003

A Pilot Study of Protracted Topotecan Dosing Using a Pharmacokinetically Guided Dosing Approach in Children with Solid Tumors

Victor M. Santana; William C. Zamboni; Mark N. Kirstein; Ming Tan; Tiebin Liu; Amar Gajjar; Peter J. Houghton; Clinton F. Stewart


Clinical Cancer Research | 1998

Phenytoin alters the disposition of topotecan and N-desmethyl topotecan in a patient with medulloblastoma.

William C. Zamboni; Amar Gajjar; Richard L. Heideman; Jos H. Beijnen; Hilde Rosing; Peter J. Houghton; Clinton F. Stewart

Collaboration


Dive into the Peter J. Houghton's collaboration.

Top Co-Authors

Avatar

Clinton F. Stewart

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

William C. Zamboni

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Victor M. Santana

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Wayne L. Furman

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Amar Gajjar

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Suzan K. Hanna

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joyce Thompson

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Lois B. Richmond

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Pamela J. Cheshire

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge