Network


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

Hotspot


Dive into the research topics where John M. McCall is active.

Publication


Featured researches published by John M. McCall.


Nature | 2012

A call for transparent reporting to optimize the predictive value of preclinical research

Story C. Landis; Susan G. Amara; Khusru Asadullah; Christopher P. Austin; Robi Blumenstein; Eileen W. Bradley; Ronald G. Crystal; Robert B. Darnell; Robert J. Ferrante; Howard Fillit; Robert Finkelstein; Marc Fisher; Howard E. Gendelman; Robert M. Golub; John L. Goudreau; Robert A. Gross; Amelie K. Gubitz; Sharon E. Hesterlee; David W. Howells; John R. Huguenard; Katrina Kelner; Walter J. Koroshetz; Dimitri Krainc; Stanley E. Lazic; Michael S. Levine; Malcolm R. Macleod; John M. McCall; Richard T. Moxley; Kalyani Narasimhan; L.J. Noble

The US National Institute of Neurological Disorders and Stroke convened major stakeholders in June 2012 to discuss how to improve the methodological reporting of animal studies in grant applications and publications. The main workshop recommendation is that at a minimum studies should report on sample-size estimation, whether and how animals were randomized, whether investigators were blind to the treatment, and the handling of data. We recognize that achieving a meaningful improvement in the quality of reporting will require a concerted effort by investigators, reviewers, funding agencies and journal editors. Requiring better reporting of animal studies will raise awareness of the importance of rigorous study design to accelerate scientific progress.


Bioorganic & Medicinal Chemistry | 2013

VBP15: Preclinical characterization of a novel anti-inflammatory delta 9,11 steroid

Erica K.M. Reeves; Eric P. Hoffman; Kanneboyina Nagaraju; Jesse M. Damsker; John M. McCall

Δ9,11 modifications of glucocorticoids (21-aminosteroids) have been developed as drugs for protection against cell damage (lipid peroxidation; lazaroids) and inhibition of neovascularization (anecortave). Part of the rationale for developing these compounds has been the loss of glucocorticoid receptor binding due to the Δ9,11 modification, thus avoiding many immunosuppressive activities and deleterious side effect profiles associated with binding to glucocorticoid and mineralocorticoid receptors. We recently demonstrated that anecortave acetate and its 21-hydroxy analog (VBP1) do, in fact, show glucocorticoid and mineralocorticoid receptor binding activities, with potent translocation of the glucocorticoid receptor to the cell nucleus. We concluded that Δ9,11 steroids showed novel anti-inflammatory properties, retaining NF-κB inhibition, but losing deleterious glucocorticoid side effect profiles. Evidence for this was developed in pre-clinical trials of chronic muscle inflammation. Here, we describe a drug development program aimed at optimizing the Δ9,11 chemistry. Twenty Δ9,11 derivatives were tested in in vitro screens for NF-κB inhibition and GR translocation to the nucleus, and low cell toxicity. VBP15 was selected as the lead compound due to potent NF-κB inhibition and GR translocation similar to prednisone and dexamethasone, lack of transactivation properties, and good bioavailability. Phamacokinetics were similar to traditional glucocorticoid drugs with terminal half-life of 0.35 h (mice), 0.58 h (rats), 5.42 h (dogs), and bioavailability of 74.5% (mice), and 53.2% (dogs). Metabolic stability showed ≥80% remaining at 1 h of VBP6 and VBP15 in human, dog, and monkey liver microsomes. Solubility, permeability and plasma protein binding were within acceptable limits. VBP15 moderately induced CYP3A4 across the three human hepatocyte donors (24-42%), similar to other steroids. VBP15 is currently under development for treatment of Duchenne muscular dystrophy.


Journal of Pharmacology and Experimental Therapeutics | 2012

Δ-9,11 Modification of Glucocorticoids Dissociates Nuclear Factor-κB Inhibitory Efficacy from Glucocorticoid Response Element-Associated Side Effects

Andreas R. Baudy; Erica K.M. Reeves; Jesse M. Damsker; Christopher R. Heier; Lindsay M. Garvin; Blythe C. Dillingham; John M. McCall; Sree Rayavarapu; Zuyi Wang; Jack Vandermeulen; Arpana Sali; Vanessa Jahnke; Stephanie Duguez; Debra C. DuBois; Mary C. Rose; Kanneboyina Nagaraju; Eric P. Hoffman

Glucocorticoids are standard of care for many inflammatory conditions, but chronic use is associated with a broad array of side effects. This has led to a search for dissociative glucocorticoids—drugs able to retain or improve efficacy associated with transrepression [nuclear factor-κB (NF-κB) inhibition] but with the loss of side effects associated with transactivation (receptor-mediated transcriptional activation through glucocorticoid response element gene promoter elements). We investigated a glucocorticoid derivative with a Δ-9,11 modification as a dissociative steroid. The Δ-9,11 analog showed potent inhibition of tumor necrosis factor-α-induced NF-κB signaling in cell reporter assays, and this transrepression activity was blocked by 17β-hydroxy-11β-[4-dimethylamino phenyl]-17α-[1-propynyl]estra-4,9-dien-3-one (RU-486), showing the requirement for the glucocorticoid receptor (GR). The Δ-9,11 analog induced the nuclear translocation of GR but showed the loss of transactivation as assayed by GR-luciferase constructs as well as mRNA profiles of treated cells. The Δ-9,11 analog was tested for efficacy and side effects in two mouse models of muscular dystrophy: mdx (dystrophin deficiency), and SJL (dysferlin deficiency). Daily oral delivery of the Δ-9,11 analog showed a reduction of muscle inflammation and improvements in multiple muscle function assays yet no reductions in body weight or spleen size, suggesting the loss of key side effects. Our data demonstrate that a Δ-9,11 analog dissociates the GR-mediated transcriptional activities from anti-inflammatory activities. Accordingly, Δ-9,11 analogs may hold promise as a source of safer therapeutic agents for chronic inflammatory disorders.


Cellular and Molecular Neurobiology | 2015

VBP15, a Novel Anti-Inflammatory, is Effective at Reducing the Severity of Murine Experimental Autoimmune Encephalomyelitis

Blythe C. Dillingham; Susan M. Knoblach; Gina M. Many; Brennan Harmon; Amanda M. Mullen; Christopher R. Heier; Luca Bello; John M. McCall; Eric P. Hoffman; Edward M. Connor; Kanneboyina Nagaraju; Erica K.M. Reeves; Jesse M. Damsker

Multiple sclerosis is a chronic disease of the central nervous system characterized by an autoimmune inflammatory reaction that leads to axonal demyelination and tissue damage. Glucocorticoids, such as prednisolone, are effective in the treatment of multiple sclerosis in large part due to their ability to inhibit pro-inflammatory pathways (e.g., NFκB). However, despite their effectiveness, long-term treatment is limited by adverse side effects. VBP15 is a recently described compound synthesized based on the lazeroid steroidal backbone that shows activity in acute and chronic inflammatory conditions, yet displays a much-reduced side effect profile compared to traditional glucocorticoids. The purpose of this study was to determine the effectiveness of VBP15 in inhibiting inflammation and disease progression in experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of multiple sclerosis. Our data show that VBP15 is effective at reducing both disease onset and severity. In parallel studies, we observed that VBP15 was able to inhibit the production of NFκB-regulated pro-inflammatory transcripts in human macrophages. Furthermore, treatment with prednisolone—but not VBP15—increased expression of genes associated with bone loss and muscle atrophy, suggesting lack of side effects of VBP15. These findings suggest that VBP15 may represent a potentially safer alternative to traditional glucocorticoids in the treatment of multiple sclerosis and other inflammatory diseases.


Steroids | 2018

Phase 1 trial of vamorolone, a first-in-class steroid, shows improvements in side effects via biomarkers bridged to clinical outcomes.

Eric P. Hoffman; Valerie Riddle; Maxime A. Siegler; Daniel Dickerson; Miroslav Backonja; William G. Kramer; Kanneboyina Nagaraju; Heather Gordish-Dressman; Jesse M. Damsker; John M. McCall

HighlightsVamorolone (VBP15) is a steroidal drug that has shown dissociative properties.The X‐ray crystal structure of vamorolone is presented.In a Phase 1 clinical trial in adult volunteers, vamorolone was shown to be safe to 20 mg/kg/day.Vamorolone showed no changes in bone turnover biomarkers, or markers of insulin resistance.Vamorolone showed no adrenal suppression through 3.0 mg/kg/day. Background: Glucocorticoid drugs are highly effective anti‐inflammatory agents, but chronic use is associated with extensive pharmacodynamic safety concerns that have a considerable negative impact on patient quality of life. Purpose: Vamorolone (VBP15) is a first‐in‐class steroidal multi‐functional drug that shows potent inhibition of pro‐inflammatory NFkB pathways via high‐affinity binding to the glucocorticoid receptor, high affinity antagonism for the mineralocorticoid receptor, and membrane stabilization properties. Pre‐clinical data in multiple mouse models of inflammation showed retention of anti‐inflammatory efficacy, but loss of most or all side effects. Experimental approach: We report first‐in‐human Phase 1 clinical trials (86 healthy adult males), with single ascending doses (0.1–20.0 mg/kg), and multiple ascending doses (1.0–20 mg/kg/day; 14 days treatment). Key results: Vamorolone was well‐tolerated at all dose levels. Vamorolone showed pharmacokinetic and metabolism profiles similar to prednisone. Biomarker studies showed loss of side effects of traditional glucocorticoid drugs (bone fragility, metabolic disturbance, immune suppression). Suppression of the adrenal axis was 10‐fold less than prednisone. The crystallographic structure of vamorolone was solved, and compared to prednisone and dexamethasone. There was overlap in structure, but differences in conformation at the C‐ring where glucocorticoids interact with Asn564 of the glucocorticoid receptor. The predicted loss of Asn564 binding to vamorolone may underlie the loss of gene transcriptional activity. Conclusions and interpretations: Vamorolone is a dissociative steroid that retains high affinity binding and nuclear translocation of both glucocorticoid (agonist) and mineralocorticoid (antagonist) receptors, but does not show pharmacodynamic safety concerns of existing glucocorticoid drugs at up to 20 mg/kg/day.


Pharmacological Research | 2018

Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first-in-class dissociative steroidal anti-inflammatory drug

Laurie S. Conklin; Jesse M. Damsker; Eric P. Hoffman; William J. Jusko; Panteleimon D. Mavroudis; Benjamin D. Schwartz; Laurel J. Mengle-Gaw; Edward C. Smith; Jean K. Mah; M. Guglieri; Yoram Nevo; Nancy L. Kuntz; Craig M. McDonald; M. Tulinius; Monique M. Ryan; Richard Webster; Diana Castro; Richard S. Finkel; Andrea L. Smith; Lauren P. Morgenroth; A. Arrieta; Maya Shimony; Mark Jaros; Phil Shale; John M. McCall; Yetrib Hathout; Kanneboyina Nagaraju; John N. van den Anker; Leanne Ward; Alexandra Ahmet

ABSTRACT We report a first‐in‐patient study of vamorolone, a first‐in‐class dissociative steroidal anti‐inflammatory drug, in Duchenne muscular dystrophy. This 2‐week, open‐label Phase IIa multiple ascending dose study (0.25, 0.75, 2.0, and 6.0 mg/kg/day) enrolled 48 boys with Duchenne muscular dystrophy (4 to <7 years), with outcomes including clinical safety, pharmacokinetics and pharmacodynamic biomarkers. The study design included pharmacodynamic biomarkers in three contexts of use: 1. Secondary outcomes for pharmacodynamic safety (insulin resistance, adrenal suppression, bone turnover); 2. Exploratory outcomes for drug mechanism of action; 3. Exploratory outcomes for expanded pharmacodynamic safety. Vamorolone was safe and well‐tolerated through the highest dose tested (6.0 mg/kg/day) and pharmacokinetics of vamorolone were similar to prednisolone. Using pharmacodynamic biomarkers, the study demonstrated improved safety of vamorolone versus glucocorticoids as shown by reduction of insulin resistance, beneficial changes in bone turnover (loss of increased bone resorption and decreased bone formation only at the highest dose level), and a reduction in adrenal suppression. Exploratory biomarkers of pharmacodynamic efficacy showed an anti‐inflammatory mechanism of action and a beneficial effect on plasma membrane stability, as demonstrated by a dose‐responsive decrease in serum creatine kinase activity. With an array of pre‐selected biomarkers in multiple contexts of use, we demonstrate the development of the first dissociative steroid that preserves anti‐inflammatory efficacy and decreases steroid‐associated safety concerns. Ongoing extension studies offer the potential to bridge exploratory efficacy biomarkers to clinical outcomes.


Physical Medicine and Rehabilitation Clinics of North America | 2012

Novel Approaches to Corticosteroid Treatment in Duchenne Muscular Dystrophy

Eric P. Hoffman; Erica K.M. Reeves; Jesse M. Damsker; Kanneboyina Nagaraju; John M. McCall; Edward M. Connor; Kate Bushby


Orphanet Journal of Rare Diseases | 2015

The TREAT-NMD advisory committee for therapeutics (TACT): an innovative de-risking model to foster orphan drug development.

Emma Heslop; Cristina Csimma; Volker Straub; John M. McCall; Kanneboyina Nagaraju; Kathryn R. Wagner; Didier Caizergues; Rudolf Korinthenberg; Kevin M. Flanigan; Petra Kaufmann; Elizabeth McNeil; Sharon E. Hesterlee; Dominic J. Wells; Kate Bushby


Inflammation Research | 2016

VBP15, a novel dissociative steroid compound, reduces NFκB-induced expression of inflammatory cytokines in vitro and symptoms of murine trinitrobenzene sulfonic acid-induced colitis.

Jesse M. Damsker; Laurie S. Conklin; Soheil Sadri; Blythe C. Dillingham; Karuna Panchapakesan; Christopher R. Heier; John M. McCall; Anthony D. Sandler


Archive | 2009

Non-hormonal steroid modulators of NF-κB for treatment of disease

John M. McCall; Eric I. Hoffman; Kanneboyina Nagaraju

Collaboration


Dive into the John M. McCall's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jesse M. Damsker

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erica K.M. Reeves

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Blythe C. Dillingham

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher R. Heier

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arpana Sali

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Edward M. Connor

George Washington University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurie S. Conklin

George Washington University

View shared research outputs
Researchain Logo
Decentralizing Knowledge