Network


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

Hotspot


Dive into the research topics where Tosca Kinchelow is active.

Publication


Featured researches published by Tosca Kinchelow.


Journal of Acquired Immune Deficiency Syndromes | 2005

Safety of Enfuvirtide in Combination With an Optimized Background of Antiretrovirals in Treatment-Experienced HIV-1-Infected Adults Over 48 Weeks

Benoit Trottier; Sharon Walmsley; Jacques Reynes; Peter J. Piliero; Mary O'Hearn; Mark Nelson; Julio S. G. Montaner; Adriano Lazzarin; Jacob Lalezari; Christine Katlama; Keith Henry; David A. Cooper; Bonaventura Clotet; Keikanus Arasteh; Jean-Francois Delfraissy; Hans-Jiirgen Stellbrink; Joep M. A. Lange; Daniel R. Kuritzkes; Joseph J. Eron; Calvin Cohen; Tosca Kinchelow; Anne Bertasso; Emily Labriola-Tompkins; Anna Shikhman; Belinda Atkins; Laurence Bourdeau; Christopher Natale; Fiona J. Hughes; Jain Chung; Denise Guimaraes

Background:Antiretroviral tolerability is a critical factor contributing to treatment outcome. The T-20 Versus Optimized Background Regimen Only (TORO) studies assessed the safety and efficacy of enfuvirtide in treatment-experienced HIV-1-infected patients. Methods:A total of 997 patients were randomized at a 2:1 ratio to an optimized background antiretroviral regimen plus enfuvirtide (n = 663) or an optimized background regimen alone (control group; n = 334). Control patients could switch to enfuvirtide on virologic failure. Results:In total, 26.5% of patients randomized to enfuvirtide and 36.6% to the control group discontinued study treatment before week 48; the percentage of patients withdrawn for safety reasons (including adverse events [AEs], deaths, and laboratory abnormalities) was 14.0% in the enfuvirtide group and 11.6% in the control group. Injection site reactions (ISRs) occurred in 98% of enfuvirtide patients and led to treatment discontinuation in 4.4%. Treatment-related (defined as possibly, probably, or remotely) AE rates per 100 patient-years were lower with enfuvirtide (96.2) than in the control group (149.9); diarrhea, nausea, and fatigue, the most frequently reported AEs, were significantly less frequent with enfuvirtide than in the control group. Pneumonia was significantly more frequent in patients treated with enfuvirtide (6.7 vs. 0.6 events per 100 patient-years), although the incidence was within expected ranges for this population. Lymphadenopathy was also higher in enfuvirtide-treated patients (7.1 vs. 1.2 events per 100 patient-years) for control patients. Conclusion:The addition of enfuvirtide to an optimized background regimen does not exacerbate AEs commonly associated with antiretrovirals. ISRs limited treatment in <5% of patients.


Clinical Pharmacology & Therapeutics | 2002

Pharmacokinetics of plasma enfuvirtide after subcutaneous administration to patientswith human immunodeficiency virus: Inverse Gaussian density absorption and 2-compartment disposition*

Xiaoping Zhang; Keith Nieforth; Jean‐Marie Lang; Régine Rouzier-Panis; Jacques Reynes; Albert Dorr; Stanley J. Kolis; Mark R. Stiles; Tosca Kinchelow; Indravadan H. Patel

Enfuvirtide (T‐20) is the first of a novel class of human immunodeficiency virus (HIV) drugs that block gp41‐mediated viral fusion to host cells. The objectives of this study were to develop a structural pharmacokinetic model that would adequately characterize the absorption and disposition of enfuvirtide pharmacokinetics after both intravenous and subcutaneous administration and to evaluate the dose proportionality of enfuvirtide pharmacokinetic parameters at a subcutaneous dose higher than that currently used in phase III studies.


Clinical Infectious Diseases | 2003

Guillain-Barré Syndrome Associated with Immune Reconstitution

Peter J. Piliero; Douglas G. Fish; Sandra L. Preston; Donna Cunningham; Tosca Kinchelow; Miklos Salgo; Jiang Qian; George L. Drusano

We report a case of acute Guillain-Barré syndrome (GBS) associated with a prompt and vigorous immune reconstitution and decrease in the virus load noted during treatment with a potent regimen of highly active antiretroviral therapy. We hypothesize that GBS may have been due to an aberrant immune response or an adverse drug reaction in association with preexisting peripheral neurologic disease.


The Journal of Clinical Pharmacology | 2003

Lack of Enzyme‐Inducing Effect of Rifampicin on the Pharmacokinetics of Enfuvirtide

Mark A. Boyd; Xiaoping Zhang; Albert Dorr; Kiat Ruxrungtham; Stanley J. Kolis; Keith Nieforth; Tosca Kinchelow; Neil Buss; Indravadan H. Patel

The primary objective was to determine whether rifampicin influences the pharmacokinetics of enfuvirtide in HIV‐1‐infected patients. In a single‐center, open‐label, one‐sequence crossover, clinical pharmacology study, 12 HIV‐1‐infected adults received enfuvirtide (90 mg, twice daily) on days 1 to 3 and days 11 to 13 (morning dose only on days 3 and 13) and rifampicin (600 mg, once daily) from days 4 to 13. Plasma concentrations were measured for enfuvirtide and its metabolite (days 3 and 13) and rifampicin (day 13 only). The ratios of least squares means (LSM) and 90% confidence intervals for enfuvirtide and enfuvirtide metabolite pharmacokinetic parameters (AUC12h, Cmax, Ctrough) were estimated in the presence and absence of rifampicin. Treatments were compared using an analysis of variance for natural log‐transformed variables, with factors patient and treatment. Efficacy and safety were also monitored. Steady‐state rifampicin had no appreciable effect on any of the pharmacokinetic parameters assessed for either enfuvirtide or its metabolite. The ratio of LSM for AUC12h, Cmax, and Ctrough for enfuvirtide was 97.5%, 103%, and 84.9%, respectively, and 108%, 112%, and 92.9%, for the enfuvirtide metabolite. Rifampicin did not affect the t1/2 of enfuvirtide or its metabolite. There were no unexpected effects of rifampicin on the short‐term antiviral effect or safety of the administered antiretroviral treatment. The pharmacokinetics of enfuvirtide are not induced by a 10‐day pretreatment with rifampicin.


Clinical Pharmacology & Therapeutics | 2004

Assessment of drug-drug interaction potential of enfuvirtide in human immunodeficiency virus type 1-infected patients

Xiaoping Zhang; Jacob Lalezari; Andrew D. Badley; Albert Dorr; Stanley J. Kolis; Tosca Kinchelow; Indravadan H. Patel

Enfuvirtide is the first drug to block human immunodeficiency virus type 1 (HIV‐1) glycoprotein 41–mediated viral fusion to host cells. This study investigated whether enfuvirtide can influence the activities of cytochrome P450 (CYP) enzymes in HIV‐1–infected patients.


Journal of Clinical Virology | 2003

Influence of subcutaneous injection site on the steady-state pharmacokinetics of enfuvirtide (T-20) in HIV-1-infected patients

Jacob Lalezari; Indravadan H. Patel; Xiaoping Zhang; Albert Dorr; Nina Hawker; Zikia Siddique; Stanley J. Kolis; Tosca Kinchelow

BACKGROUND Enfuvirtide is the first in a new class of antiretrovirals (ARVs), the fusion inhibitors, and the first ARV to be administered by subcutaneous (s.c.) injection. OBJECTIVES The primary objective of this study was to determine the steady-state pharmacokinetics and relative bioavailability of enfuvirtide following sc injection at three separate anatomical sites: abdomen (A), thigh (B) and arm (C). STUDY DESIGN A single-center, open-label, multiple-dose, three-way randomized, crossover study. Twelve HIV-1-infected adults were recruited from three ongoing Phase II enfuvirtide clinical trials and randomized into three groups. Each group continued to receive s.c. injection of enfuvirtide, at a dose of 90 mg twice daily (bid), according to one of three treatment sequences: ABC, BCA or CAB; over three consecutive periods of approximately 7 days each. Plasma concentrations of enfuvirtide and its metabolite (Ro 50-6343) were measured using a validated liquid chromatography-tandem mass spectrometry method. RESULTS The relative bioavailability of enfuvirtide, based on AUC12h and abdomen as a reference site, was 101% for thigh and 117% for arm. The AUC12h of Ro 50-6343 ranged from 14 to 16% of that for enfuvirtide. Although injection site reactions (ISRs) were common, the overall grading (based on pain or discomfort) of all reported ISRs was Grade 1 (mild). The incidence of ISRs varied according to the site of injection, as did the signs and symptoms associated with them. No patient required treatment for an ISR. CONCLUSIONS Comparability among the three injection sites, in terms of both absorption and the ISR profile, allows HIV-1-infected patients the freedom to choose and to rotate, if necessary, the site of enfuvirtide injection among the three anatomical sites.


The Journal of Clinical Pharmacology | 2004

Lack of Interaction between Enfuvirtide and Ritonavir or Ritonavir‐Boosted Saquinavir in HIV‐1‐Infected Patients

Kiat Ruxrungtham; Mark A. Boyd; S. Eralp Bellibas; Xiaoping Zhang; Albert Dorr; Stanley J. Kolis; Tosca Kinchelow; Neil Buss; Indravadan H. Patel

Enfuvirtide (Fuzeon™) is an HIV fusion inhibitor, the first drug in a new class of antiretrovirals. The HIV protease inhibitors ritonavir and saquinavir both inhibit cytochrome P450 (CYP450) isoenzymes, and low‐dose ritonavir is often used to boost pharmacokinetic exposure to full‐dose protease inhibitors. These two studies were designed to assess whether ritonavir and ritonavir‐boosted saquinavir influence the steady‐state pharmacokinetics of enfuvirtide. Both studies were single‐center, open‐label, one‐sequence crossover clinical pharmacology studies in 12 HIV‐1‐infected patients each. Patients received enfuvirtide (90 mg twice daily [bid], subcutaneous injection) for 7 days and either ritonavir (200 mg bid, ritonavir study, orally) or saquinavir/ritonavir (1000/100 mg bid, saquinavir/ritonavir study, orally) for 4 days on days 4 to 7. Serial blood samples were collected up to 24 hours after the morning dose of enfuvirtide on days 3 and 7. Plasma concentrations for enfuvirtide, enfuvirtide metabolite, saquinavir, and ritonavir were measured using validated liquid chromatography tandem mass spectrometry methods. Efficacy and safety were also monitored. Bioequivalence criteria require the 90% confidence interval (CI) for the least squares means (LSM) of Cmax and AUC12h to be between 80% and 125%. In the present studies, analysis of variance showed that when coadministered with ritonavir, the ratio of LSM for enfuvirtide was 124% for Cmax (90% confidence interval [CI]: 109%‐141%), 122% for AUC12h (90% CI: 108%‐137%), and 114% for Ctrough (90% CI: 102%‐128%). Although the bioequivalence criteria were not met, the increase in enfuvirtide exposure was small (< 25%) and not clinically relevant. When administered with ritonavir‐boosted saquinavir, the ratio of LSM for enfuvirtide was 107% for Cmax (90% CI: 94.3%‐121%) and 114% for AUC12h (90% CI: 105%‐124%), which therefore met bioequivalence criteria, and 126% for Ctrough (90% CI: 117%‐135%). The pharmacokinetics of enfuvirtide are affected to a small extent when coadministered with ritonavir at a dose of 200 mg bid but not when coadministered with a saquinavir‐ritonavir combination (1000/100 mg bid). However, previous clinical studies have shown that such increases in enfuvirtide exposure are not clinically relevant. Thus, no dosage adjustments are warranted when enfuvirtide is coadministered with low‐dose ritonavir or saquinavir boosted with a low dose of ritonavir.


Clinical Pharmacology & Therapeutics | 2003

Assessment of Metabolic Inhibition Potential of Enfuvirtide Using a 5‐Drug Cocktail in HIV‐1 Infected Patients

Xiaoping Zhang; Indravadan H. Patel; Jacob Lalezari; Andrew D. Badley; Nina Hawker; Albert Dorr; Stanley J. Kolis; Tosca Kinchelow

Clinical Pharmacology & Therapeutics (2003) 73, P81–P81; doi:


Statistics in Medicine | 2005

Safety of Enfuvirtide in Combination With an Optimized Background of Antiretrovirals in Treatment-Experienced HIV-1-Infected Adults Over 48 Weeks:

Benoit Trottier; Sharon Walmsley; Jacques Reynes; Peter J. Piliero; Mary O'Hearn; Mark Nelson; Julio S. G. Montaner; Adriano Lazzarin; Jacob Lalezari; Christine Katlama; Keith Henry; David A. Cooper; Bonaventura Clotet; Keikawus Arastéh; Jean-Fran ois Delfraissy; Hans-J rgen Stellbrink; Joep M. A. Lange; Daniel R. Kuritzkes; Joseph J. Eron; Calvin Cohen; Tosca Kinchelow; Anne Bertasso; Emily Labriola-Tompkins; Anna Shikhman; Belinda Atkins; Laurence Bourdeau; Christopher Natale; Fiona J. Hughes; Jain Chung; Denise Guimaraes


Conference on Retroviruses and Opportunistic Infections | 2003

Injection site reactions with the HIV-1 fusion inhibitor enfuvirtide

Russell A. Ball; Tosca Kinchelow

Collaboration


Dive into the Tosca Kinchelow's collaboration.

Top Co-Authors

Avatar

Albert Dorr

Center for Global Development

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacob Lalezari

University of California

View shared research outputs
Top Co-Authors

Avatar

Jacques Reynes

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benoit Trottier

Université du Québec à Montréal

View shared research outputs
Top Co-Authors

Avatar

Julio S. G. Montaner

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sharon Walmsley

University Health Network

View shared research outputs
Researchain Logo
Decentralizing Knowledge