Network


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

Hotspot


Dive into the research topics where Mark Butt is active.

Publication


Featured researches published by Mark Butt.


Regulatory Toxicology and Pharmacology | 2011

A multiple-dose toxicity study of tanezumab in cynomolgus monkeys.

Mark Zorbas; Susan Hurst; David L. Shelton; Mark Evans; Deborah Finco; Mark Butt

Nerve growth factor (NGF) is an important mediator of pain and hyperalgesia and has become a target of novel analgesic therapeutics. Tanezumab is a humanized IgG(2) antibody that binds NGF with high affinity and specificity. In a study to assess the toxicity and pharmacokinetic properties of tanezumab in adult, male and female, cynomolgus monkeys following weekly intravenous administration of 1, 10, or 30 mg/kg for up to 26 weeks (followed by an 8-week recovery period), tanezumab was well tolerated with no macroscopic or microscopic effects on those brain, spinal cord, nerve, or ganglia sections evaluated. One fifth of tanezumab-treated monkeys developed an antibody response to tanezumab that prevented maintenance of tanezumab exposure between dosing. In the antibody-negative animals, accumulation of tanezumab was observed; steady state was achieved approximately 8 weeks after the first dose of study drug, and exposure to tanezumab was approximately dose proportional with no observed difference between male and female animals. One monkey died during the study; this monkey had findings suggestive of hypersensitivity reaction. The favorable toxicity and pharmacokinetic profile of tanezumab seen in this study supports its further evaluation for the treatment of pain in clinical practice.


Regulatory Toxicology and Pharmacology | 2014

Dietary administration of paraquat for 13 weeks does not result in a loss of dopaminergic neurons in the substantia nigra of C57BL/6J mice.

Daniel J. Minnema; Kim Z. Travis; Charles B. Breckenridge; Nicholas C. Sturgess; Mark Butt; Jeffrey C. Wolf; Dan Zadory; Melissa J. Beck; James M. Mathews; Merrill O. Tisdel; Andrew R. Cook; Philip A. Botham; Lewis L. Smith

Several investigations have reported that mice administered paraquat dichloride (PQ·Cl2) by intraperitoneal injection exhibit a loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). In this study, male and female C57BL/6J mice were administered PQ·Cl2 in the diet at concentrations of 0 (control), 10, and 50ppm for a duration of 13weeks. A separate group of mice were administered 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) during week 12 as positive controls to produce a loss of dopaminergic neurons in the SNpc. The comparative effects of PQ and MPTP on the SNpc and/or striatum were assessed using neurochemical, neuropathological, and stereological endpoints. Morphological and stereological assessments were performed by investigators blinded to the origin of the tissue. Neither dose of PQ·Cl2 (10 or 50 ppm in the diet) caused a loss of striatal dopamine or dopamine metabolite concentrations in the brains of mice. Pathological assessments of the SNpc and striatum showed no evidence of neuronal degeneration or astrocytic/microglial activation. Furthermore, the number of tyrosine hydroxylase-positive (TH(+)) neurons in the SNpc was not reduced in PQ-treated mice. In contrast, MPTP caused a decrease in striatal dopamine concentration, a reduction in TH(+) neurons in the SNpc, and significant pathological changes including astrocytic and microglial activation in the striatum and SNpc. The MPTP-induced effects were greater in males than in females. It is concluded that 13weeks of continuous dietary exposure of C57BL/6J mice to 50ppm PQ·Cl2 (equivalent to 10.2 and 15.6mg PQ ion/kg body weight/day for males and females, respectively) does not result in the loss of, or damage to, dopaminergic neurons in the SNpc.


Toxicological Sciences | 2014

Morphologic, Stereologic, and Morphometric Evaluation of the Nervous System in Young Cynomolgus Monkeys (Macaca fascicularis) Following Maternal Administration of Tanezumab, a Monoclonal Antibody to Nerve Growth Factor

Mark Butt; Mark Evans; Christopher J. Bowman; Thomas Cummings; Satoru Oneda; David L. Shelton; Mark Zorbas

Tanezumab, an antibody to nerve growth factor, was administered to pregnant cynomolgus monkeys at 0, 0.5, 4, and 30u2009mg/kg weekly, beginning gestation day (GD) 20 through parturition (∼GD165). Maternal tanezumab administration appeared to increase stillbirths and infant mortality, but no consistent pattern of gross and/or microscopic change was detected to explain the mortality. Offspring exposed in utero were evaluated at 12 months of age using light microscopy (all tissues), stereology (basal forebrain cholinergic and dorsal root ganglia neurons), and morphometry (sural nerve). Light microscopy revealed decreased number of neurons in sympathetic ganglia (superior mesenteric, cervicothoracic, and ganglia in the thoracic sympathetic trunk). Stereologic assessment indicated an overall decrease in dorsal root ganglion (thoracic) volume and number of neurons in animals exposed to tanezumab 4u2009mg/kg (nu2009=u20099) and 30u2009mg/kg (nu2009=u20091). At all tanezumab doses, the sural nerve was small due to decreases in myelinated and unmyelinated axons. Existing axons/myelin sheaths appeared normal when viewed with light and transmission electron microscopy. There was no indication of tanezumab-related, active neuron/nerve fiber degeneration/necrosis in any tissue, indicating decreased sensory/sympathetic neurons and axonal changes were due to hypoplasia or atrophy. These changes in the sensory and sympathetic portions of the peripheral nervous system suggest some degree of developmental neurotoxicity, although what effect, if any, the changes had on normal function and survival was not apparent. Overall, these changes were consistent with published data from rodent studies.


Reproductive Toxicology | 2014

Developmental toxicity assessment of tanezumab, an anti-nerve growth factor monoclonal antibody, in cynomolgus monkeys (Macaca fascicularis).

Christopher J. Bowman; Mark Evans; Thomas Cummings; Satoru Oneda; Mark Butt; Susan Hurst; Jessica-lyn Gremminger; David L. Shelton; Cris Kamperschroer; Mark Zorbas

Two intravenous studies with tanezumab, an anti-nerve growth factor monoclonal antibody, were conducted in pregnant cynomolgus monkeys to assess potential effects on pregnancy and pre- and postnatal development. Study 1 evaluated infants up to 12 months of age following weekly maternal dosing (0, 0.5, 4 or 30 mg/kg; 18 per group) from gestation day (GD) 20 through parturition. Study 2 evaluated infants 2 months postnatally following weekly maternal dosing (0, 0.5 or 30 mg/kg; 20-21 per group) from GD 20 through 48. In the absence of maternal toxicity, tanezumab increased stillbirth and post-birth infant mortality/morbidity, decreased infant growth and resulted in microscopic changes in the peripheral sympathetic and sensory nervous system of the infants at all doses. Decreased primary antibody responses and increased incidences in skin changes in infants were also observed. The no-observed-adverse-effect-level for maternal toxicity was 30 mg/kg and <0.5 mg/kg for developmental toxicity.


Toxicological Sciences | 2017

From the Cover: Evaluation of the Effects of Tanezumab, a Monoclonal Antibody Against Nerve Growth Factor, on the Sympathetic Nervous System in Adult Cynomolgus Monkeys (Macaca fascicularis): A Stereologic, Histomorphologic, and Cardiofunctional Assessment

Patrice Bélanger; Paul Butler; Mark Butt; Siddhartha Bhatt; Stephen Foote; David L. Shelton; Mark Evans; Rosalinda Arends; Susan Hurst; Carlin Okerberg; Thomas Cummings; David Potter; Jill Steidl-Nichols; Mark Zorbas

Abstract Tanezumab, a humanized monoclonal antibody against nerve growth factor is in development for treatment of chronic pain. Three nonclinical studies assessed effects of clinically relevant and supratherapeutic doses of tanezumab on the sympathetic nervous system (SNS) of adult nonhuman primates. Study 1 evaluated potential effects of subcutaneous (SC) tanezumab (1.2u2009mg/kg every 8u2009weeks [Q8W]) on SNS in cynomolgus monkeys for 3 or 6u2009months and reversibility or persistence of any effects through a nondosing/recovery period. Study 2 evaluated whether neuronal cell death occurs shortly after a single SC tanezumab injection (1.2u2009mg/kg). Assessments for these two studies included evaluations of superior cervical and cervicothoracic ganglia for neuronal cell death and morphology. Study 3 evaluated effects of SC tanezumab (1.2u2009mg/kg Q8W and 30u2009mg/kg/week) over 6u2009months on sympathetic control of cardiovascular function. Tanezumab exposure was associated with stereologic changes in sympathetic ganglia, including smaller ganglion volume, and smaller average neuron size/area beginning at 2u2009weeks and reaching maximal levels by 1u2009month with no further progression through 6u2009months. These changes were not associated with clinical signs, completely reversed upon tanezumab withdrawal, and were not considered adverse. Tanezumab had no adverse effects on sympathetic control of cardiovascular function. These data support the conclusion that tanezumab administration for up to 6u2009months has no adverse effects on SNS morphology or function and does not cause neuronal cell death in adult nonhuman primates.


Toxicological Sciences | 2016

Safety Evaluation of CNS Administered Biologics—Study Design, Data Interpretation, and Translation to the Clinic

Brian R. Vuillemenot; Sven Korte; Teresa Leah Wright; Eric L. Adams; Robert B. Boyd; Mark Butt

Many central nervous system (CNS) diseases are inadequately treated by systemically administered therapies due to the blood brain barrier (BBB), which prevents achieving adequate drug concentrations at sites of action. Due to the increasing prevalence of neurodegenerative diseases and the inability of most systemically administered therapies to cross the BBB, direct CNS delivery will likely play an increasing role in treatment. Administration of large molecules, cells, viral vectors, oligonucleotides, and other novel therapies directly to the CNS via the subarachnoid space, ventricular system, or parenchyma overcomes this obstacle. Clinical experience with direct CNS administration of small molecule therapies suggests that this approach may be efficacious for the treatment of neurodegenerative disorders using biological therapies. Risks of administration into the brain tissue or cerebrospinal fluid include local damage from implantation of the delivery system and/or administration of the therapeutic and reactions affecting the CNS. Preclinical safety studies on CNS administered compounds must differentiate between the effects of the test article, the delivery device, and/or the vehicle, and assess exacerbations of reactions due to combinations of effects. Animal models characterized for safety assessment of CNS administered therapeutics have enabled human trials, but interpretation can be challenging. This manuscript outlines the challenges of preclinical intrathecal/intracerebroventricular/intraparenchymal studies, evaluation of results, considerations for special endpoints, and translation of preclinical findings to enable first-in-human trials. Recommendations will be made based on the authors collective experience with conducting these studies to enable clinical development of CNS-administered biologics.


Materials Research Express | 2015

Preclinical characterization and safety of a novel hydrogel for augmenting dural repair

Michael J. Strong; Michael A Carnahan; Keith D’Alessio; Jared Daniel Goodnow Butlin; Mark Butt; Anthony L. Asher

Cerebrospinal fluid (CSF) leakage is a potentially serious complication in surgical procedures involving opening of the dura mater. Although several materials have been developed to help achieve watertight dural closures, CSF leakages persist. The goal of this study was to evaluate the performance of a novel hydrogel designed to provide augmentation to standard methods of dural repair. Performance measures such as polymerization time, dimensional swelling, burst strength, and elasticity were examined in laboratory situations. Additionally, biocompatibility in an in vivo rat model was examined. The results demonstrate that this novel hydrogel has superior mechanical strength and tissue adherence with enhanced flexibility, reduced swelling, and quicker set time compared with existing hydrogel dural sealants approved for intra-cranial use. Furthermore, biocompatibility studies demonstrate that this compound is both non-toxic and non-immunogenic.


Journal of Pharmacology and Experimental Therapeutics | 2017

Reveglucosidase alfa (BMN 701), an IGF 2 Tagged rhAcid α-Glucosidase, Improves Respiratory Functional Parameters in a Murine Model of Pompe Disease

Jeff Peng; Jill Dalton; Mark Butt; Kristin Tracy; Derek Kennedy; Peter E. Haroldsen; Rhea Cahayag; Stephen Zoog; Charles A. O'Neill; Laurie Tsuruda

Pompe disease is a rare neuromuscular disorder caused by an acid α-glucosidase (GAA) deficiency resulting in glycogen accumulation in muscle, leading to myopathy and respiratory weakness. Reveglucosidase alfa (BMN 701) is an insulin-like growth factor 2–tagged recombinant human acid GAA (rhGAA) that enhances rhGAA cellular uptake via a glycosylation-independent insulin-like growth factor 2 binding region of the cation-independent mannose-6-phosphate receptor (CI-MPR). The studies presented here evaluated the effects of Reveglucosidase alfa treatment on glycogen clearance in muscle relative to rhGAA, as well as changes in respiratory function and glycogen clearance in respiratory-related tissue in a Pompe mouse model (GAAtm1Rabn/J). In a comparison of glycogen clearance in muscle with Reveglucosidase alfa and rhGAA, Reveglucosidase alfa was more effective than rhGAA with 2.8–4.7 lower EC50 values, probably owing to increased cellular uptake. The effect of weekly intravenous administration of Reveglucosidase alfa on respiratory function was monitored in Pompe and wild-type mice using whole body plethysmography. Over 12 weeks of 20-mg/kg Reveglucosidase alfa treatment in Pompe mice, peak inspiratory flow (PIF) and peak expiratory flow (PEF) stabilized with no compensation in respiratory rate and inspiratory time during hypercapnic and recovery conditions compared with vehicle-treated Pompe mice. Dose-related decreases in glycogen levels in both ambulatory and respiratory muscles generally correlated to changes in respiratory function. Improvement of murine PIF and PEF were similar in magnitude to increases in maximal inspiratory and expiratory pressure observed clinically in late onset Pompe patients treated with Reveglucosidase alfa (Byrne et al., manuscript in preparation).


Archive | 2010

Adherus Dural and Spinal Sealant as Adjuncts to Sutured Dural Repair in a Canine Lumbar Durotomy Repair Model

Anthony L. Asher; Michael A Carnahan; Robert B. Boyd; Eric L. Adams; Mark Butt; Spine Associates


Molecular Genetics and Metabolism | 2018

Pharmacology of BMN 250 administered via intracerebroventricular infusion once every 2 weeks for twenty-six weeks or longer in a canine model of mucopolysaccharidosis type IIIB

N. Matthew Ellinwood; Bethann Valentine; Andrew Hess; Jackie K. Jens; Elizabeth M. Snella; Wendy A. Ware; Shannon J. Hostetter; Gil Ben-Shlomo; Nick D. Jeffery; Sina Safayi; Jodi D. Smith; Suzanne T. Millman; Rebecca L. Parsons; Mark Butt; Jonathan D. Cooper; Igor Nestrasil; Heather Prill; Xiao Liu; Huiyu Zhou; Roger Lawrence; Brett E. Crawford; Anita Grover; Andrew Melton; Anu Cherukuri; Jill C.M. Wait; Jason Pinkstaff; Emma McCullagh

Collaboration


Dive into the Mark Butt's collaboration.

Top Co-Authors

Avatar

Anthony L. Asher

Carolinas Healthcare System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anita Grover

BioMarin Pharmaceutical

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge