Network


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

Hotspot


Dive into the research topics where Christine Fandozzi is active.

Publication


Featured researches published by Christine Fandozzi.


Antimicrobial Agents and Chemotherapy | 2012

MK-5172, a Selective Inhibitor of Hepatitis C Virus NS3/4a Protease with Broad Activity across Genotypes and Resistant Variants

Vincenzo Summa; Steven W. Ludmerer; John A. McCauley; Christine Fandozzi; Christine Burlein; Giuliano Claudio; Paul J. Coleman; Jillian DiMuzio; Marco Ferrara; Marcello Di Filippo; Adam T. Gates; Donald J. Graham; Steven Harper; Daria J. Hazuda; Carolyn McHale; Edith Monteagudo; Vincenzo Pucci; Michael Rowley; Michael T. Rudd; Aileen Soriano; Mark W. Stahlhut; Joseph P. Vacca; David B. Olsen; Nigel Liverton; Steven S. Carroll

ABSTRACT HCV NS3/4a protease inhibitors are proven therapeutic agents against chronic hepatitis C virus infection, with boceprevir and telaprevir having recently received regulatory approval as add-on therapy to pegylated interferon/ribavirin for patients harboring genotype 1 infections. Overcoming antiviral resistance, broad genotype coverage, and a convenient dosing regimen are important attributes for future agents to be used in combinations without interferon. In this communication, we report the preclinical profile of MK-5172, a novel P2-P4 quinoxaline macrocyclic NS3/4a protease inhibitor currently in clinical development. The compound demonstrates subnanomolar activity against a broad enzyme panel encompassing major hepatitis C virus (HCV) genotypes as well as variants resistant to earlier protease inhibitors. In replicon selections, MK-5172 exerted high selective pressure, which yielded few resistant colonies. In both rat and dog, MK-5172 demonstrates good plasma and liver exposures, with 24-h liver levels suggestive of once-daily dosing. When administered to HCV-infected chimpanzees harboring chronic gt1a or gt1b infections, MK-5172 suppressed viral load between 4 to 5 logs at a dose of 1 mg/kg of body weight twice daily (b.i.d.) for 7 days. Based on its preclinical profile, MK-5172 is anticipated to be broadly active against multiple HCV genotypes and clinically important resistance variants and highly suited for incorporation into newer all-oral regimens.


ChemMedChem | 2013

Discovery of MK-8742: an HCV NS5A inhibitor with broad genotype activity.

Craig A. Coburn; Peter T. Meinke; Wei Chang; Christine Fandozzi; Donald J. Graham; Bin Hu; Qian Huang; Stacia Kargman; Joseph A. Kozlowski; Rong Liu; John A. McCauley; Amin Nomeir; Richard Soll; Joseph P. Vacca; Dahai Wang; Hao Wu; Bin Zhong; David B. Olsen; Steven W. Ludmerer

The NS5A protein plays a critical role in the replication of HCV and has been the focus of numerous research efforts over the past few years. NS5A inhibitors have shown impressive in vitro potency profiles in HCV replicon assays, making them attractive components for inclusion in all oral combination regimens. Early work in the NS5A arena led to the discovery of our first clinical candidate, MK‐4882 [2‐((S)‐pyrrolidin‐2‐yl)‐5‐(2‐(4‐(5‐((S)‐pyrrolidin‐2‐yl)‐1H‐imidazol‐2‐yl)phenyl)benzofuran‐5‐yl)‐1H‐imidazole]. While preclinical proof‐of‐concept studies in HCV‐infected chimpanzees harboring chronic genotype 1 infections resulted in significant decreases in viral load after both single‐ and multiple‐dose treatments, viral breakthrough proved to be a concern, thus necessitating the development of compounds with increased potency against a number of genotypes and NS5A resistance mutations. Modification of the MK‐4882 core scaffold by introduction of a cyclic constraint afforded a series of tetracyclic inhibitors, which showed improved virologic profiles. Herein we describe the research efforts that led to the discovery of MK‐8742, a tetracyclic indole‐based NS5A inhibitor, which is currently in phase 2b clinical trials as part of an all‐oral, interferon‐free regimen for the treatment of HCV infection.


Journal of Medicinal Chemistry | 2010

Discovery of Vaniprevir (MK-7009), a Macrocyclic Hepatitis C Virus NS3/4a Protease Inhibitor

John A. McCauley; Charles J. Mcintyre; Michael T. Rudd; Kevin Nguyen; Joseph J. Romano; John W. Butcher; Kevin F. Gilbert; Kimberly J. Bush; M. Katharine Holloway; John Swestock; Bang-Lin Wan; Steven S. Carroll; Jillian DiMuzio; Donald J. Graham; Steven W. Ludmerer; Shi-Shan Mao; Mark Stahlhut; Christine Fandozzi; Nicole Trainor; David B. Olsen; Joseph P. Vacca; Nigel J. Liverton

A new class of HCV NS3/4a protease inhibitors which contain a P2 to P4 macrocyclic constraint was designed using a molecular-modeling derived strategy. Exploration of the P2 heterocyclic region, the P2 to P4 linker, and the P1 side chain of this class of compounds via a modular synthetic strategy allowed for the optimization of enzyme potency, cellular activity, and rat liver exposure following oral dosing. These studies led to the identification of clinical candidate 35b (vaniprevir, MK-7009), which is active against both the genotype 1 and genotype 2 NS3/4a protease enzymes and has good plasma exposure and excellent liver exposure in multiple species.


Antimicrobial Agents and Chemotherapy | 2010

MK-7009, a Potent and Selective Inhibitor of Hepatitis C Virus NS3/4A Protease

Nigel J. Liverton; Steven S. Carroll; Jillian DiMuzio; Christine Fandozzi; Donald J. Graham; Daria J. Hazuda; M. Katherine Holloway; Steven W. Ludmerer; John A. McCauley; Charles J. Mcintyre; David B. Olsen; Michael T. Rudd; Mark Stahlhut; Joseph P. Vacca

ABSTRACT The administration of hepatitis C virus (HCV) NS3/4A protease inhibitors to patients with chronic HCV infections has demonstrated that they have dramatic antiviral effects and that compounds acting via this mechanism are likely to form a key component of future anti-HCV therapy. We report here on the preclinical profile of MK-7009, an inhibitor of genotype 1a and 1b proteases at subnanomolar concentrations with modestly shifted potency against genotype 2a and 2b proteases at low nanomolar concentrations. Potent activity was also observed in a cell-based HCV replicon assay in the presence of added human serum (50%). In multiple species evaluated in preclinical studies, the MK-7009 concentrations in the liver were maintained at a significant multiple of the cell-based replicon 50% effective concentration over 12 to 24 h following the administration of moderate oral doses (5 to 10 mg per kg of body weight). MK-7009 also had excellent selectivity against both a range of human proteases and a broad panel of pharmacologically relevant ion channels, receptors, and enzymes. On the basis of this favorable profile, MK-7009 was selected for clinical development and is currently being evaluated in controlled clinical trials with both healthy volunteers and HCV-infected patients.


ACS Medicinal Chemistry Letters | 2012

Discovery of MK-5172, a Macrocyclic Hepatitis C Virus NS3/4a Protease Inhibitor.

Steven Harper; John A. McCauley; Michael T. Rudd; Marco Ferrara; Marcello DiFilippo; Benedetta Crescenzi; Uwe Koch; Alessia Petrocchi; M. Katharine Holloway; John W. Butcher; Joseph J. Romano; Kimberly J. Bush; Kevin F. Gilbert; Charles J. Mcintyre; Kevin Nguyen; Emanuela Nizi; Steven S. Carroll; Steven W. Ludmerer; Christine Burlein; Jillian DiMuzio; Donald J. Graham; Carolyn McHale; Mark Stahlhut; David B. Olsen; Edith Monteagudo; Simona Cianetti; Claudio Giuliano; Vincenzo Pucci; Nicole Trainor; Christine Fandozzi

A new class of HCV NS3/4a protease inhibitors containing a P2 to P4 macrocyclic constraint was designed using a molecular modeling-derived strategy. Building on the profile of previous clinical compounds and exploring the P2 and linker regions of the series allowed for optimization of broad genotype and mutant enzyme potency, cellular activity, and rat liver exposure following oral dosing. These studies led to the identification of clinical candidate 15 (MK-5172), which is active against genotype 1-3 NS3/4a and clinically relevant mutant enzymes and has good plasma exposure and excellent liver exposure in multiple species.


Journal of the American Chemical Society | 2008

Molecular Modeling Based Approach to Potent P2−P4 Macrocyclic Inhibitors of Hepatitis C NS3/4A Protease

Nigel J. Liverton; M. Katharine Holloway; John A. McCauley; Michael T. Rudd; John W. Butcher; Steven S. Carroll; Jillian DiMuzio; Christine Fandozzi; Kevin F. Gilbert; Shi-Shan Mao; Charles J. Mcintyre; Kevin Nguyen; Joseph J. Romano; Mark Stahlhut; Bang-Lin Wan; and David B. Olsen; Joseph P. Vacca

Molecular modeling of inhibitor bound full length HCV NS3/4A protease structures proved to be a valuable tool in the design of a new series of potent NS3 protease inhibitors. Optimization of initial compounds provided 25a. The in vitro activity and selectivity as well as the rat pharmacokinetic profile of 25a compare favorably with the data for other NS3/4A protease inhibitors currently in clinical development for the treatment of HCV.


Gastroenterology | 2014

The Combination of MK-5172, Peginterferon, and Ribavirin Is Effective in Treatment-Naive Patients With Hepatitis C Virus Genotype 1 Infection Without Cirrhosis

Michael P. Manns; John M. Vierling; Bruce R. Bacon; Savino Bruno; Oren Shibolet; Yaacov Baruch; Patrick Marcellin; Luzelena Caro; Anita Y. M. Howe; Christine Fandozzi; Jacqueline Gress; Christopher L. Gilbert; Peter M. Shaw; Michael P. Cooreman; Michael N. Robertson; Peggy Hwang; Frank J. Dutko; Janice Wahl; Niloufar Mobashery

BACKGROUND & AIMS MK-5172 is an inhibitor of the hepatitis C virus (HCV) nonstructural protein 3/4A protease; MK-5172 is taken once daily and has a higher potency and barrier to resistance than licensed protease inhibitors. We investigated the efficacy and tolerability of MK-5172 with peginterferon and ribavirin (PR) in treatment-naive patients with chronic HCV genotype 1 infection without cirrhosis. METHODS We performed a multicenter, double-blind, randomized, active-controlled, dose-ranging, response-guided therapy study. A total of 332 patients received MK-5172 (100, 200, 400, or 800 mg) once daily for 12 weeks in combination with PR. Patients in the MK-5172 groups received PR for an additional 12 or 36 weeks, based on response at week 4. Patients in the control group (n = 66) received a combination of boceprevir and PR, dosed in accordance with boceprevirs US product circular. RESULTS At 24 weeks after the end of therapy, sustained virologic responses were achieved in 89%, 93%, 91%, and 86% of the patients in the groups given the combination of PR and MK-5172 (100, 200, 400, or 800 mg), respectively, vs 61% of controls. In the MK-5172 group receiving 100 mg, 91% of patients had undetectable levels of HCV RNA at week 4 and qualified for the short duration of therapy. The combination of MK-5172 and PR generally was well tolerated. Transient increases in transaminase levels were noted in the MK-5172 groups given 400 and 800 mg, at higher frequencies than in the MK-5172 groups given 100 or 200 mg, or control groups. CONCLUSIONS Once-daily MK-5172 (100 mg) with PR for 24 or 48 weeks was highly effective and well tolerated among treatment-naive patients with HCV genotype 1 infection without cirrhosis. Studies are underway to evaluate interferon-free MK-5172-based regimens. ClinicalTrials.gov number: NCT01353911.


ACS Medicinal Chemistry Letters | 2011

Discovery of MK-1220: A Macrocyclic Inhibitor of Hepatitis C Virus NS3/4A Protease with Improved Preclinical Plasma Exposure

Michael T. Rudd; John A. McCauley; John W. Butcher; Joseph J. Romano; Charles J. Mcintyre; Kevin Nguyen; Kevin F. Gilbert; Kimberly J. Bush; M. Katharine Holloway; John Swestock; Bang-Lin Wan; Steven S. Carroll; Jillian DiMuzio; Donald J. Graham; Steven W. Ludmerer; Mark Stahlhut; Christine Fandozzi; Nicole Trainor; David B. Olsen; Joseph P. Vacca; Nigel J. Liverton

The discovery of MK-1220 is reported along with the development of a series of HCV NS3/4A protease inhibitors containing a P2 to P4 macrocyclic constraint with improved preclinical pharmacokinetics. Optimization of the P2 heterocycle substitution pattern as well as the P3 amino acid led to compounds with greatly improved plasma exposure following oral dosing in both rats and dogs while maintaining excellent enzyme potency and cellular activity. These studies led to the identification of MK-1220.


Pharmacology Research & Perspectives | 2015

Preclinical pharmacology and pharmacokinetics of CERC-301, a GluN2B-selective N-methyl-D-aspartate receptor antagonist.

Rachel Garner; Shobha Gopalakrishnan; John A. McCauley; Rodney A. Bednar; Stanley L. Gaul; Scott D. Mosser; Laszlo Kiss; Joseph J. Lynch; Shil Patel; Christine Fandozzi; Armando Lagrutta; Richard Briscoe; Nigel Liverton; Blake Paterson; James Vornov; Reza Mazhari

The preclinical pharmacodynamic and pharmacokinetic properties of 4‐methylbenzyl (3S, 4R)‐3‐fluoro‐4‐[(Pyrimidin‐2‐ylamino) methyl] piperidine‐1‐carboxylate (CERC‐301), an orally bioavailable selective N‐methyl‐D‐aspartate (NMDA) receptor subunit 2B (GluN2B) antagonist, were characterized to develop a translational approach based on receptor occupancy (RO) to guide CERC‐301 dose selection in clinical trials of major depressive disorder. CERC‐301 demonstrated high‐binding affinity (Ki, 8.1 nmol L−1) specific to GluN2B with an IC50 of 3.6 nmol L−1 and no off‐target activity. CERC‐301 efficacy was demonstrated in the forced swim test with an efficacy dose (ED50) of 0.3–0.7 mg kg−1 (RO, 30–50%); increase in locomotor activity was observed at ED50 of 2 mg kg−1, corresponding to an RO of 75%. The predicted 50% RO concentration (Occ50) in humans was 400 nmol L−1, similar to that predicted for rat, dog, and monkey (300, 200, and 400 nmol L−1, respectively). Safety pharmacology and neurotoxicity studies raised no specific safety concerns. A first‐in‐human study in healthy males demonstrated a dose‐proportional pharmacokinetic profile, with Tmax of ~1 h and t1/2 of 12–17 h. Based on the preclinical and pharmacodynamic data, doses of ≥8 mg in humans are hypothesized to have an acceptable safety profile and result in clinically relevant peak plasma exposure.


Bioorganic & Medicinal Chemistry Letters | 2015

Novel oxazolidinone calcitonin gene-related peptide (CGRP) receptor antagonists for the acute treatment of migraine

Brendan M. Crowley; Craig A. Stump; Diem N. Nguyen; Craig M. Potteiger; Melody Mcwherter; Daniel V. Paone; Amy G. Quigley; Joseph G. Bruno; Dan Cui; J. Christopher Culberson; Andrew Danziger; Christine Fandozzi; Danny Gauvreau; Amanda L. Kemmerer; Karsten Menzel; Eric L. Moore; Scott D. Mosser; Vijay Bhasker G. Reddy; Rebecca B. White; Christopher A. Salvatore; Stefanie A. Kane; Ian M. Bell; Harold G. Selnick; Mark E. Fraley; Christopher S. Burgey

In our efforts to develop CGRP receptor antagonists as backups to MK-3207, 2, we employed a scaffold hopping approach to identify a series of novel oxazolidinone-based compounds. The development of a structurally diverse, potent (20, cAMP+HS IC50=0.67 nM), and selective compound (hERG IC50=19 μM) with favorable rodent pharmacokinetics (F=100%, t1/2=7h) is described. Key to this development was identification of a 3-substituted spirotetrahydropyran ring that afforded a substantial gain in potency (10 to 35-fold).

Collaboration


Dive into the Christine Fandozzi's collaboration.

Top Co-Authors

Avatar

John A. McCauley

Monell Chemical Senses Center

View shared research outputs
Top Co-Authors

Avatar

Donald J. Graham

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steven S. Carroll

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Mark Stahlhut

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

John W. Butcher

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge