Network


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

Hotspot


Dive into the research topics where Jennifer Visich is active.

Publication


Featured researches published by Jennifer Visich.


British Journal of Ophthalmology | 2014

Systemic pharmacokinetics following intravitreal injections of ranibizumab, bevacizumab or aflibercept in patients with neovascular AMD

Robert L. Avery; Nathan Steinle; Dilsher S. Dhoot; Dante J. Pieramici; Robert F. See; Stephen Couvillion; Ma’an A. Nasir; Melvin Rabena; Kha Le; Mauricio Maia; Jennifer Visich

Background Data comparing systemic exposure and systemic vascular endothelial growth factor (VEGF) suppression of ranibizumab, bevacizumab and aflibercept following intravitreal injection are lacking. Methods Fifty-six patients with wet age-related macular degeneration received intravitreal ranibizumab (0.5 mg), bevacizumab (1.25 mg), or aflibercept (2.0 mg). Serum pharmacokinetics and plasma free VEGF were evaluated after the first and third injections. Results Following the first dose, systemic exposure to aflibercept was 5-, 37-, and 9-fold higher than ranibizumab, whereas, bevacizumab was 9-, 310-, and 35-fold higher than ranibizumab, based on geometric mean ratio of peak and trough concentrations and area under the curve, respectively. The third dose showed accumulation of bevacizumab and aflibercept but not ranibizumab. Aflibercept substantially suppressed plasma free VEGF, with mean levels below lower limit of quantitation (10 pg/mL) as early as 3 h postdose until ≥7 days postdose. Mean free (unbound) VEGF levels with ranibizumab were largely unchanged, with mean trough level of 14.4 pg/mL compared with baseline of 17 pg/mL. Conclusions There are notable differences in systemic pharmacokinetics and pharmacodynamics among anti-VEGF treatments after intravitreal administration. All three agents rapidly moved into the bloodstream, but ranibizumab very quickly cleared, whereas bevacizumab and aflibercept demonstrated greater systemic exposure and produced a marked reduction in plasma free VEGF. Trial registration number NCT02118831.


Gut | 2013

A randomised phase I study of etrolizumab (rhuMAb β7) in moderate to severe ulcerative colitis

Paul Rutgeerts; Richard N. Fedorak; Daan W. Hommes; Andreas Sturm; Daniel C. Baumgart; Brian Bressler; Stefan Schreiber; John C. Mansfield; Marna Williams; Meina Tang; Jennifer Visich; Xiaohui Wei; Mary E. Keir; Diana Luca; Dimitri Danilenko; Jackson G. Egen; Sharon O'Byrne

Objective Etrolizumab (rhuMAb β7, anti-β7, PRO145223) is a humanised monoclonal antibody targeting the β7 subunit of the heterodimeric integrins α4β7 and αEβ7, which are implicated in leucocyte migration and retention in ulcerative colitis (UC). This randomised phase I study evaluated the safety and pharmacology of etrolizumab in patients with moderate to severe UC. Design In the single ascending dose (SAD) stage, etrolizumab (0.3, 1.0, 3.0, 10 mg/kg intravenous, 3.0 mg/kg subcutaneous (SC) or placebo) was administered 4:1 (n=25) in each cohort. In the multiple dose (MD) stage, new patients received monthly etrolizumab (0.5 mg/kg SC (n=4), 1.5 mg/kg SC (n=5), 3.0 mg/kg SC (n=4), 4.0 mg/kg intravenous (n=5)) or placebo (n=5). The pharmacokinetics was studied and Mayo Clinic Score evaluated at baseline, day 29 (SAD), and days 43 and 71 (MD). Results In the SAD stage, there were no dose limiting toxicities, infusion or injection site reactions. Two impaired wound healing serious adverse events occurred in two patients receiving etrolizumab. In the MD stage, there were no dose limiting toxicities, and no infusion or injection site reactions. Headache was the most common adverse event, occurring more often in etrolizumab patients. Antietrolizumab antibodies were detected in two subjects. The duration of β7 receptor full occupancy was dose related. A clinical response was observed in 12/18 patients, and clinical remission in 3/18 patients treated with etrolizumab in the MD stage, compared with 4/5 and 1/5 placebo patients, respectively. Conclusion Etrolizumab is well tolerated in moderate to severe UC. Further investigation is warranted.


mAbs | 2012

Minipig as a potential translatable model for monoclonal antibody pharmacokinetics after intravenous and subcutaneous administration

Yanan Zheng; Devin Tesar; Lisa Benincosa; Herbert Birnböck; C. Andrew Boswell; Daniela Bumbaca; Kyra J. Cowan; Dimitry M. Danilenko; Ann L. Daugherty; Paul J. Fielder; Hans Peter Grimm; Amita Joshi; Nicole Justies; Gerry Kolaitis; Nicholas Lewin-Koh; Jing Li; Sami McVay; Jennifer O'Mahony; Michael B. Otteneder; Michael Pantze; Wendy S. Putnam; Zhihua J. Qiu; Jane Ruppel; Thomas Singer; Oliver Boris Stauch; Frank-Peter Theil; Jennifer Visich; Jihong Yang; Yong Ying; Leslie A. Khawli

Subcutaneous (SC) delivery is a common route of administration for therapeutic monoclonal antibodies (mAbs) with pharmacokinetic (PK)/pharmacodynamic (PD) properties requiring long-term or frequent drug administration. An ideal in vivo preclinical model for predicting human PK following SC administration may be one in which the skin and overall physiological characteristics are similar to that of humans. In this study, the PK properties of a series of therapeutic mAbs following intravenous (IV) and SC administration in Göttingen minipigs were compared with data obtained previously from humans. The present studies demonstrated: (1) minipig is predictive of human linear clearance; (2) the SC bioavailabilities in minipigs are weakly correlated with those in human; (3) minipig mAb SC absorption rates are generally higher than those in human and (4) the SC bioavailability appears to correlate with systemic clearance in minipigs. Given the important role of the neonatal Fc-receptor (FcRn) in the PK of mAbs, the in vitro binding affinities of these IgGs against porcine, human and cynomolgus monkey FcRn were tested. The result showed comparable FcRn binding affinities across species. Further, mAbs with higher isoelectric point tended to have faster systemic clearance and lower SC bioavailability in both minipig and human. Taken together, these data lend increased support for the use of the minipig as an alternative predictive model for human IV and SC PK of mAbs.


The Journal of Clinical Pharmacology | 2012

Population Pharmacokinetics of Rituximab in Patients With Chronic Lymphocytic Leukemia

Jing Li; Jianguo Zhi; Michael K. Wenger; Nancy Valente; Anna Dmoszynska; Tadeusz Robak; Ranvir Mangat; Amita Joshi; Jennifer Visich

This retrospective analysis characterizes rituximab population pharmacokinetics in combination with fludarabine and cyclophosphamide and its effect on fludarabine and cyclophosphamide disposition in chronic lymphocytic leukemia (CLL) patients. Rituximab concentration data were well described by a 2‐compartment model comprising a time‐varying clearance component related to the target‐mediated clearance pathway and a constant clearance component reflecting catabolic elimination pathway. Marked differences were observed compared to pharmacokinetic parameters for non‐Hodgkin lymphoma (NHL) obtained previously: in CLL, time‐varying clearance at time zero (CL2) was faster, volumes of distribution (V1 and V2) were larger, and rate of change (Kdes) from the target‐mediated clearance pathway to catabolic elimination was lower than NHL. Fludarabine and cyclophosphamide disposition showed no apparent change when co‐administered with rituximab. A positive correlation between pharmacokinetic parameters and clinical response was observed, supporting the use of the higher rituximab dose of 500 mg/m2 in CLL patients (vs 375 mg/m2 in NHL) to achieve an effective clinical response.


Ophthalmology | 2014

Pharmacokinetics of Ranibizumab after Intravitreal Administration in Patients with Retinal Vein Occlusion or Diabetic Macular Edema

Yi Zhang; Zhenling Yao; Nitin Kaila; Peter J. Kuebler; Jennifer Visich; Mauricio Maia; Lisa Tuomi; Jason S. Ehrlich; Roman G. Rubio; Peter A. Campochiaro

OBJECTIVE To describe the systemic pharmacokinetics of ranibizumab after intravitreal administration in patients with retinal vein occlusion (RVO) or diabetic macular edema (DME). DESIGN A population approach of nonlinear mixed-effect pharmacokinetics modeling based on serum concentrations of ranibizumab measured at various times after intravitreal administration. PARTICIPANTS Patients with RVO (n = 441) and DME (n = 435) from 4 large, randomized, phase 3 clinical trials of monthly ranibizumab intravitreal administration. METHODS A 1-compartment pharmacokinetics model with first-order absorption and elimination rate constants previously developed in patients with age-related macular degeneration (AMD) was fitted separately to RVO and DME data. Population pharmacokinetic parameters and interindividual variability were estimated for each model. Baseline covariates were evaluated for potential effects on systemic pharmacokinetics. Model performance was validated using general diagnostic plots and a visual predictive check. MAIN OUTCOME MEASURES Ranibizumab disposition was determined in RVO and DME patients and compared with that previously seen in AMD patients. RESULTS The AMD pharmacokinetics model correctly predicted the measured serum ranibizumab concentration data for RVO and DME patients. Most observed data points were within the simulated 90% confidence interval, indicating that systemic ranibizumab concentrations were comparable among AMD, RVO, and DME patients. No disease-related covariates were identified by the population pharmacokinetics analysis. CONCLUSIONS The systemic pharmacokinetics of ranibizumab were similar among patients with AMD, RVO, or DME. Disease-related differences and patient demographics, measured in this study, did not lead to variability in ocular elimination or in systemic exposure of ranibizumab after intravitreal administration. In all disease processes tested, ranibizumab exits the eye slowly and then is eliminated rapidly from the circulation, thus minimizing systemic exposure.


Aaps Journal | 2008

Use of Quantitative Pharmacology in the Development of HAE1, a High-Affinity Anti-IgE Monoclonal Antibody

Wendy S. Putnam; Jing Li; Jonas Haggstrom; Chee Ng; Saloumeh Kadkhodayan-Fischer; Melissa Cheu; Yamo Deniz; Henry B. Lowman; Paul J. Fielder; Jennifer Visich; Amita Joshi; Nelson “Shasha” Jumbe


Cancer Chemotherapy and Pharmacology | 2014

Population pharmacokinetic and covariate analysis of pertuzumab, a HER2‑targeted monoclonal antibody, and evaluation of a fixed, non‑weight‑based dose in patients with a variety of solid tumors

Amit Garg; Angelica Quartino; Jing Li; Jin Jin; D. Russell Wada; Hanbin Li; Javier Cortes; Virginia McNally; Graham Ross; Jennifer Visich; Bert L. Lum


Cancer Chemotherapy and Pharmacology | 2013

Exposure–response analysis of pertuzumab in HER2-positive metastatic breast cancer: absence of effect on QTc prolongation and other ECG parameters

Amit Garg; Jing Li; Emma Clark; Adam Knott; Timothy J. Carrothers; Jean‑François Marier; Javier Cortes; Michael Brewster; Jennifer Visich; Bert L. Lum


Cancer Chemotherapy and Pharmacology | 2013

Characterization of the long-term pharmacokinetics of bevacizumab following last dose in patients with resected stage II and III carcinoma of the colon.

Jing Li; Manish Gupta; Denise Jin; Yan Xin; Jennifer Visich; David Edward Allison


Cancer Chemotherapy and Pharmacology | 2016

Population pharmacokinetic and exposure-response analysis for trastuzumab administered using a subcutaneous "manual syringe" injection or intravenously in women with HER2-positive early breast cancer.

Angelica Quartino; Carina Hillenbach; Jing Li; Hanbin Li; Russell Wada; Jennifer Visich; Chunze Li; Dominik Heinzmann; Jin Y. Jin; Bert L. Lum

Collaboration


Dive into the Jennifer Visich's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge