Network


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

Hotspot


Dive into the research topics where Kari R. Fonseca is active.

Publication


Featured researches published by Kari R. Fonseca.


Science Translational Medicine | 2012

Negative Allosteric Modulation of the mGluR5 Receptor Reduces Repetitive Behaviors and Rescues Social Deficits in Mouse Models of Autism

Jill L. Silverman; Daniel G. Smith; Stacey J. Sukoff Rizzo; Michael N. Karras; Sarah M. Turner; Seda S. Tolu; Dianne K. Bryce; Deborah L. Smith; Kari R. Fonseca; Robert H. Ring; Jacqueline N. Crawley

Autism-like behaviors in mice were reversed by a negative modulator of a metabotropic glutamate receptor, suggesting a treatment for symptoms of autism spectrum disorders. Treatment of Autism Symptoms in Mice When they are 2 to 5 years old, children with autism start to show unusual social interactions and impaired communication. They may fail to develop relationships with their peers and be unable to interpret nuances of speech and body language. Most show repetitive motor behaviors and restricted interests and can have associated seizures, anxiety, or intellectual impairment. A large number of genes can put people at risk for this disorder, each in a small number of cases, and these genes point to connections between neurons as a vulnerable point in autism. Now, Silverman and colleagues have used two inbred strains of mice that display well-replicated behavioral abnormalities relevant to the diagnostic symptoms of autism and shown that some of these symptoms can be improved with a drug directed at a central glutamate receptor of the brain, mGluR5. The authors used two inbred strains of mice that display robust behaviors relevant to the diagnostic symptoms of autism. BTBR mice show deficits in many types of social interactions and high levels of repetitive self-grooming. C58 repetitively jumps. They used GRN-529, a compound developed by Pfizer that reduces the actions of glutamate, the main excitatory neurotransmitter in the brain. Other mGluR antagonists are showing promise in clinical trials for people with the fragile X mutation, who have both intellectual impairments and autism, so the authors reasoned that an mGluR5 compound might help autistic symptoms. GRN-529 reduced both the repetitive self-grooming in BTBR and the repetitive jumping in C58. Most intriguingly, GRN-529 also improved social behaviors in BTBR in two assays, one for social approach to an unfamiliar mouse and one for social interactions between freely moving pairs of mice. A particular strength of this study is that the authors replicated these beneficial actions of the mGluR5 compound in several separate groups of mice, in two laboratories. Although the path from target identification to effective human treatment is a long and winding road, the discovery of therapeutic efficacy for an mGluR5 negative allosteric modulator in both the repetitive and the social domains in two distinct mouse models is a promising beginning. This single biological target may offer a useful entry point to develop a pharmacological therapy that alleviates many symptoms of autism spectrum disorders. Neurodevelopmental disorders such as autism and fragile X syndrome were long thought to be medically untreatable, on the assumption that brain dysfunctions were immutably hardwired before diagnosis. Recent revelations that many cases of autism are caused by mutations in genes that control the ongoing formation and maturation of synapses have challenged this dogma. Antagonists of metabotropic glutamate receptor subtype 5 (mGluR5), which modulate excitatory neurotransmission, are in clinical trials for fragile X syndrome, a major genetic cause of intellectual disabilities. About 30% of patients with fragile X syndrome meet the diagnostic criteria for autism. Reasoning by analogy, we considered the mGluR5 receptor as a potential target for intervention in autism. We used BTBR T+tf/J (BTBR) mice, an established model with robust behavioral phenotypes relevant to the three diagnostic behavioral symptoms of autism—unusual social interactions, impaired communication, and repetitive behaviors—to probe the efficacy of a selective negative allosteric modulator of the mGluR5 receptor, GRN-529. GRN-529 reduced repetitive behaviors in three cohorts of BTBR mice at doses that did not induce sedation in control assays of open field locomotion. In addition, the same nonsedating doses reduced the spontaneous stereotyped jumping that characterizes a second inbred strain of mice, C58/J. Further, GRN-529 partially reversed the striking lack of sociability in BTBR mice on some parameters of social approach and reciprocal social interactions. These findings raise the possibility that a single targeted pharmacological intervention may alleviate multiple diagnostic behavioral symptoms of autism.


Journal of Medicinal Chemistry | 2009

Identification of a Brain Penetrant PDE9A Inhibitor Utilizing Prospective Design and Chemical Enablement as a Rapid Lead Optimization Strategy

Patrick Robert Verhoest; Caroline Proulx-Lafrance; Michael Corman; Lois K. Chenard; Christopher John Helal; Xinjun Hou; Robin J. Kleiman; Shenping Liu; Eric S. Marr; Frank S. Menniti; Christopher J. Schmidt; Michelle Vanase-Frawley; Anne W. Schmidt; Robert Williams; Frederick R. Nelson; Kari R. Fonseca; Spiros Liras

By use of chemical enablement and prospective design, a novel series of selective, brain penetrant PDE9A inhibitors have been identified that are capable of producing in vivo elevations of brain cGMP.


The Journal of Neuroscience | 2014

Reduction of Brain Kynurenic Acid Improves Cognitive Function

Rouba Kozak; Brian M. Campbell; Christine A. Strick; Weldon Horner; William E. Hoffmann; Tamás Kiss; Douglas S. Chapin; Dina McGinnis; Amanda L. Abbott; Brooke M. Roberts; Kari R. Fonseca; Victor Guanowsky; Damon Young; Patricia A. Seymour; Amy B. Dounay; Mihály Hajós; Graham V. Williams; Stacy A. Castner

The elevation of kynurenic acid (KYNA) observed in schizophrenic patients may contribute to core symptoms arising from glutamate hypofunction, including cognitive impairments. Although increased KYNA levels reduce excitatory neurotransmission, KYNA has been proposed to act as an endogenous antagonist at the glycine site of the glutamate NMDA receptor (NMDAR) and as a negative allosteric modulator at the α7 nicotinic acetylcholine receptor. Levels of KYNA are elevated in CSF and the postmortem brain of schizophrenia patients, and these elevated levels of KYNA could contribute to NMDAR hypofunction and the cognitive deficits and negative symptoms associated with this disease. However, the impact of endogenously produced KYNA on brain function and behavior is less well understood due to a paucity of pharmacological tools. To address this issue, we identified PF-04859989, a brain-penetrable inhibitor of kynurenine aminotransferase II (KAT II), the enzyme responsible for most brain KYNA synthesis. In rats, systemic administration of PF-04859989 dose-dependently reduced brain KYNA to as little as 28% of basal levels, and prevented amphetamine- and ketamine-induced disruption of auditory gating and improved performance in a sustained attention task. It also prevented ketamine-induced disruption of performance in a working memory task and a spatial memory task in rodents and nonhuman primates, respectively. Together, these findings support the hypotheses that endogenous KYNA impacts cognitive function and that inhibition of KAT II, and consequent lowering of endogenous brain KYNA levels, improves cognitive performance under conditions considered relevant for schizophrenia.


Journal of Pharmacology and Experimental Therapeutics | 2012

Phosphodiesterase 9A regulates central cGMP and modulates responses to cholinergic and monoaminergic perturbation in vivo.

Robin J. Kleiman; Douglas S. Chapin; Curt Christoffersen; Jody Freeman; Kari R. Fonseca; Kieran F. Geoghegan; Sarah Grimwood; Victor Guanowsky; Mihály Hajós; John F. Harms; Christopher John Helal; William E. Hoffmann; Geralyn P. Kocan; Mark J. Majchrzak; Dina McGinnis; Stafford McLean; Frank S. Menniti; Fredrick R. Nelson; Robin Roof; Anne W. Schmidt; Patricia A. Seymour; Diane Stephenson; Francis David Tingley; Michelle Vanase-Frawley; Patrick Robert Verhoest; Christopher J. Schmidt

Cyclic nucleotides are critical regulators of synaptic plasticity and participate in requisite signaling cascades implicated across multiple neurotransmitter systems. Phosphodiesterase 9A (PDE9A) is a high-affinity, cGMP-specific enzyme widely expressed in the rodent central nervous system. In the current study, we observed neuronal staining with antibodies raised against PDE9A protein in human cortex, cerebellum, and subiculum. We have also developed several potent, selective, and brain-penetrant PDE9A inhibitors and used them to probe the function of PDE9A in vivo. Administration of these compounds to animals led to dose-dependent accumulation of cGMP in brain tissue and cerebrospinal fluid, producing a range of biological effects that implied functional significance for PDE9A-regulated cGMP in dopaminergic, cholinergic, and serotonergic neurotransmission and were consistent with the widespread distribution of PDE9A. In vivo effects of PDE9A inhibition included reversal of the respective disruptions of working memory by ketamine, episodic and spatial memory by scopolamine, and auditory gating by amphetamine, as well as potentiation of risperidone-induced improvements in sensorimotor gating and reversal of the stereotypic scratching response to the hallucinogenic 5-hydroxytryptamine 2A agonist mescaline. The results suggested a role for PDE9A in the regulation of monoaminergic circuitry associated with sensory processing and memory. Thus, PDE9A activity regulates neuronal cGMP signaling downstream of multiple neurotransmitter systems, and inhibition of PDE9A may provide therapeutic benefits in psychiatric and neurodegenerative diseases promoted by the dysfunction of these diverse neurotransmitter systems.


ACS Medicinal Chemistry Letters | 2012

Discovery of Brain-Penetrant, Irreversible Kynurenine Aminotransferase II Inhibitors for Schizophrenia.

Amy B. Dounay; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Michelle Marie Claffey; Artem G. Evdokimov; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Matthew Merrill Hayward; Weldon Horner; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vanessa Paradis; Vinod D. Parikh; Matthew R. Reese; Suobao Rong; Michelle A. Salafia; Katherine Schuyten; Christine A. Strick; Jamison B. Tuttle; James Valentine; Hong Wang; Laura E. Zawadzke; Patrick Robert Verhoest

Kynurenine aminotransferase (KAT) II has been identified as a potential new target for the treatment of cognitive impairment associated with schizophrenia and other psychiatric disorders. Following a high-throughput screen, cyclic hydroxamic acid PF-04859989 was identified as a potent and selective inhibitor of human and rat KAT II. An X-ray crystal structure and (13)C NMR studies of PF-04859989 bound to KAT II have demonstrated that this compound forms a covalent adduct with the enzyme cofactor, pyridoxal phosphate (PLP), in the active site. In vivo pharmacokinetic and efficacy studies in rat show that PF-04859989 is a brain-penetrant, irreversible inhibitor and is capable of reducing brain kynurenic acid by 50% at a dose of 10 mg/kg (sc). Preliminary structure-activity relationship investigations have been completed and have identified the positions on this scaffold best suited to modification for further optimization of this novel series of KAT II inhibitors.


Journal of Medicinal Chemistry | 2012

Design and Discovery of 6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943), a Selective Brain Penetrant PDE9A Inhibitor for the Treatment of Cognitive Disorders

Patrick Robert Verhoest; Kari R. Fonseca; Xinjun Hou; Caroline Proulx-Lafrance; Michael Corman; Christopher John Helal; Michelle Marie Claffey; Jamison B. Tuttle; Karen J. Coffman; Shenpinq Liu; Frederick R. Nelson; Robin J. Kleiman; Frank S. Menniti; Christopher J. Schmidt; Michelle Vanase-Frawley; Spiros Liras

6-[(3S,4S)-4-Methyl-1-(pyrimidin-2-ylmethyl)pyrrolidin-3-yl]-1-(tetrahydro-2H-pyran-4-yl)-1,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one (PF-04447943) is a novel PDE9A inhibitor identified using parallel synthetic chemistry and structure-based drug design (SBDD) and has advanced into clinical trials. Selectivity for PDE9A over other PDE family members was achieved by targeting key residue differences between the PDE9A and PDE1C catalytic site. The physicochemical properties of the series were optimized to provide excellent in vitro and in vivo pharmacokinetics properties in multiple species including humans. It has been reported to elevate central cGMP levels in the brain and CSF of rodents. In addition, it exhibits procognitive activity in several rodent models and synaptic stabilization in an amyloid precursor protein (APP) transgenic mouse model. Recent disclosures from clinical trials confirm that it is well tolerated in humans and elevates cGMP in cerebral spinal fluid of healthy volunteers, confirming that it is a quality pharmacological tool for testing clinical hypotheses in disease states associated with impairment of cGMP signaling or cognition.


Journal of Medicinal Chemistry | 2011

Use of structure-based design to discover a potent, selective, in vivo active phosphodiesterase 10A inhibitor lead series for the treatment of schizophrenia.

Christopher John Helal; Zhijun Kang; Xinjun Hou; Jayvardhan Pandit; Thomas A. Chappie; John M. Humphrey; Eric S. Marr; Kimberly F. Fennell; Lois K. Chenard; Carol B. Fox; Christopher J. Schmidt; Robert Williams; Douglas S. Chapin; Judith A. Siuciak; Lorraine A. Lebel; Frank S. Menniti; Julia Cianfrogna; Kari R. Fonseca; Frederick R. Nelson; Rebecca O'connor; Mary Macdougall; Laura McDowell; Spiros Liras

Utilizing structure-based virtual library design and scoring, a novel chimeric series of phosphodiesterase 10A (PDE10A) inhibitors was discovered by synergizing binding site interactions and ADME properties of two chemotypes. Virtual libraries were docked and scored for potential binding ability, followed by visual inspection to prioritize analogs for parallel and directed synthesis. The process yielded highly potent and selective compounds such as 16. New X-ray cocrystal structures enabled rational design of substituents that resulted in the successful optimization of physical properties to produce in vivo activity and to modulate microsomal clearance and permeability.


Journal of Medicinal Chemistry | 2012

Application of structure-based drug design and parallel chemistry to identify selective, brain penetrant, in vivo active phosphodiesterase 9A inhibitors.

Michelle Marie Claffey; Christopher John Helal; Patrick Robert Verhoest; Zhijun Kang; Kristina S. Fors; Stanley Jung; Jiaying Zhong; Mark W. Bundesmann; Xinjun Hou; Shenping Lui; Robin J. Kleiman; Michelle Vanase-Frawley; Anne W. Schmidt; Frank S. Menniti; Christopher J. Schmidt; William E. Hoffman; Mihály Hajós; Laura McDowell; Rebecca E. O’Connor; Mary MacDougall-Murphy; Kari R. Fonseca; Stacey L. Becker; Frederick R. Nelson; Spiros Liras

Phosphodiesterase 9A inhibitors have shown activity in preclinical models of cognition with potential application as novel therapies for treating Alzheimers disease. Our clinical candidate, PF-04447943 (2), demonstrated acceptable CNS permeability in rats with modest asymmetry between central and peripheral compartments (free brain/free plasma = 0.32; CSF/free plasma = 0.19) yet had physicochemical properties outside the range associated with traditional CNS drugs. To address the potential risk of restricted CNS penetration with 2 in human clinical trials, we sought to identify a preclinical candidate with no asymmetry in rat brain penetration and that could advance into development. Merging the medicinal chemistry strategies of structure-based design with parallel chemistry, a novel series of PDE9A inhibitors was identified that showed improved selectivity over PDE1C. Optimization afforded preclinical candidate 19 that demonstrated free brain/free plasma ≥ 1 in rat and reduced microsomal clearance along with the ability to increase cyclic guanosine monophosphosphate levels in rat CSF.


Journal of Medicinal Chemistry | 2014

Discovery and preclinical characterization of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo-[3,4-b]pyrazine (PF470): a highly potent, selective, and efficacious metabotropic glutamate receptor 5 (mGluR5) negative allosteric modulator.

Lei Zhang; Gayatri Balan; Gabriela Barreiro; Brian P. Boscoe; Lois K. Chenard; Julie Cianfrogna; Michelle Marie Claffey; Laigao Chen; Karen J. Coffman; Susan E. Drozda; Joshua R. Dunetz; Kari R. Fonseca; Paul Galatsis; Sarah Grimwood; John T. Lazzaro; Jessica Y. Mancuso; Emily L. Miller; Matthew R. Reese; Bruce N. Rogers; Isao Sakurada; Marc B. Skaddan; Deborah L. Smith; Antonia F. Stepan; Patrick Trapa; Jamison B. Tuttle; Patrick Robert Verhoest; Daniel P. Walker; Ann S. Wright; Margaret M. Zaleska; Kenneth Zasadny

A novel series of pyrazolopyrazines is herein disclosed as mGluR5 negative allosteric modulators (NAMs). Starting from a high-throughput screen (HTS) hit (1), a systematic structure-activity relationship (SAR) study was conducted with a specific focus on balancing pharmacological potency with physicochemical and pharmacokinetic (PK) properties. This effort led to the discovery of 1-methyl-3-(4-methylpyridin-3-yl)-6-(pyridin-2-ylmethoxy)-1H-pyrazolo[3,4-b]pyrazine (PF470, 14) as a highly potent, selective, and orally bioavailable mGluR5 NAM. Compound 14 demonstrated robust efficacy in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-rendered Parkinsonian nonhuman primate model of l-DOPA-induced dyskinesia (PD-LID). However, the progression of 14 to the clinic was terminated because of a potentially mechanism-mediated finding consistent with a delayed-type immune-mediated type IV hypersensitivity in a 90-day NHP regulatory toxicology study.


ACS Medicinal Chemistry Letters | 2013

Structure-Based Design of Irreversible Human KAT II Inhibitors: Discovery of New Potency-Enhancing Interactions

Jamison B. Tuttle; Marie Anderson; Bruce M. Bechle; Brian M. Campbell; Cheng Chang; Amy B. Dounay; Edelweiss Evrard; Kari R. Fonseca; Xinmin Gan; Somraj Ghosh; Weldon Horner; Larry C. James; Ji-Young Kim; Laura A. McAllister; Jayvardhan Pandit; Vinod D. Parikh; Brian Rago; Michelle A. Salafia; Christine A. Strick; Laura E. Zawadzke; Patrick Robert Verhoest

A series of aryl hydroxamates recently have been disclosed as irreversible inhibitors of kynurenine amino transferase II (KAT II), an enzyme that may play a role in schizophrenia and other psychiatric and neurological disorders. The utilization of structure-activity relationships (SAR) in conjunction with X-ray crystallography led to the discovery of hydroxamate 4, a disubstituted analogue that has a significant potency enhancement due to a novel interaction with KAT II. The use of k inact/K i to assess potency was critical for understanding the SAR in this series and for identifying compounds with improved pharmacodynamic profiles.

Researchain Logo
Decentralizing Knowledge