Network


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

Hotspot


Dive into the research topics where Dinah Misner is active.

Publication


Featured researches published by Dinah Misner.


Pharmacology & Therapeutics | 2015

Inhibition of nicotinamide phosphoribosyltransferase (NAMPT) as a therapeutic strategy in cancer.

Deepak Sampath; Tanja S. Zabka; Dinah Misner; Thomas O’Brien; Peter S. Dragovich

NAD is a metabolite that is an important cofactor and second messenger for a number of cellular processes such as genomic stability and metabolism that are essential for survival. NAD is generated de novo from tryptophan or recycled from NAM through the NAMPT-dependent salvage pathway. Alternatively, cells can convert NA to NAD through the NAPRT1-dependent salvage pathway. Tumor cells rapidly turn over NAD but do not efficiently utilize the de novo synthesis pathway. Hence, they are more reliant on the NAMPT salvage pathway for NAD regeneration making this enzyme an attractive therapeutic target for cancer. NAMPT is over-expressed in a number of cancer types such as colorectal, ovarian, breast, gastric, prostate, gliomas as well as B-cell lymphomas. A number of novel, potent and selective NAMPT small molecule inhibitors have been synthesized to date that have displayed robust anti-tumor activity in tumor models in vitro and in vivo. These inhibitors efficiently suppress NAD production in a time dependent manner and sustained reduction of NAD levels leads to loss of ATP and ultimately cell death. This review will summarize the chemical properties of these unique NAMPT inhibitors as well as their mechanism of action, pharmacodynamic activity and efficacy in tumor models in vitro and in vivo. An overview of biomarkers that predict response to treatment and mechanisms of resistance to NAMPT inhibitors will also be provided. Additionally, NAMPT inhibitors that have advanced into clinical trials will be reviewed along with experimental strategies tested to potentially increase the therapeutic index of these inhibitors.


Toxicological Sciences | 2015

Retinal Toxicity, in vivo and in vitro, Associated with Inhibition of Nicotinamide Phosphoribosyltransferase

Tanja S. Zabka; Jatinder Singh; Preeti Dhawan; Bianca M. Liederer; Jason Oeh; Mara A. Kauss; Yang Xiao; Mark Zak; Tori Lin; Bobbi McCray; Nghi La; Trung Nguyen; Joseph Beyer; Cynthia Farman; Hirdesh Uppal; Peter S. Dragovich; Thomas O’Brien; Deepak Sampath; Dinah Misner

Nicotinamide phosphoribosyltransferase (NAMPT) is a pleiotropic protein with intra- and extra-cellular functions as an enzyme, cytokine, growth factor, and hormone. NAMPT is of interest for oncology, because it catalyzes the rate-limiting step in the salvage pathway to generate nicotinamide adenine dinucleotide (NAD), which is considered a universal energy- and signal-carrying molecule involved in cellular energy metabolism and many homeostatic functions. This manuscript describes NAMPT inhibitor-induced retinal toxicity that was identified in rodent safety studies. This toxicity had a rapid onset and progression and initially targeted the photoreceptor and outer nuclear layers. Using in vivo safety and efficacy rodent studies, human and mouse cell line potency data, human and rat retinal pigmented epithelial cell in vitro systems, and rat mRNA expression data of NAMPT, nicotinic acid phosphoribosyltransferase, and nicotinamide mononucleotide adenylyltransferease (NMNAT) in several tissues from rat including retina, we demonstrate that the retinal toxicity is on-target and likely human relevant. We demonstrate that this toxicity is not mitigated by coadministration of nicotinic acid (NA), which can enable NAD production through the NAMPT-independent pathway. Further, modifying the physiochemical properties of NAMPT inhibitors could not sufficiently reduce retinal exposure. Our work highlights opportunities to leverage appropriately designed efficacy studies to identify known and measurable safety findings to screen compounds more rapidly and reduce animal use. It also demonstrates that in vitro systems with the appropriate cell composition and relevant biology and toxicity endpoints can provide tools to investigate mechanism of toxicity and the human translation of nonclinical safety concerns.


Chemical Research in Toxicology | 2016

Safety Lead Optimization and Candidate Identification: Integrating New Technologies into Decision-Making.

Donna Dambach; Dinah Misner; Mathew Brock; Aaron M. Fullerton; William R. Proctor; Jonathan Maher; Dong Lee; Kevin A. Ford

Discovery toxicology focuses on the identification of the most promising drug candidates through the development and implementation of lead optimization strategies and hypothesis-driven investigation of issues that enable rational and informed decision-making. The major goals are to [a] identify and progress the drug candidate with the best overall drug safety profile for a therapeutic area, [b] remove the most toxic drugs from the portfolio prior to entry into humans to reduce clinical attrition due to toxicity, and [c] establish a well-characterized hazard and translational risk profile to enable clinical trial designs. This is accomplished through a framework that balances the multiple considerations to identify a drug candidate with the overall best drug characteristics and provides a cogent understanding of mechanisms of toxicity. The framework components include establishing a target candidate profile for each program that defines the qualities of a successful candidate based on the intended therapeutic area, including the risk tolerance for liabilities; evaluating potential liabilities that may result from engaging the therapeutic target (pharmacology-mediated or on-target) and that are chemical structure-mediated (off-target); and characterizing identified liabilities. Lead optimization and investigation relies upon the integrated use of a variety of technologies and models (in silico, in vitro, and in vivo) that have achieved a sufficient level of qualification or validation to provide confidence in their use. We describe the strategic applications of various nonclinical models (established and new) for a holistic and integrated risk assessment that is used for rational decision-making. While this review focuses on strategies for small molecules, the overall concepts, approaches, and technologies are generally applicable to biotherapeutics.


Toxicology Mechanisms and Methods | 2015

Preclinical models of nicotinamide phosphoribosyltransferase inhibitor-mediated hematotoxicity and mitigation by co-treatment with nicotinic acid

Jacqueline M. Tarrant; Preeti Dhawan; Jatinder Singh; Tanja S. Zabka; Emer Clarke; Garry DosSantos; Peter S. Dragovich; Deepak Sampath; Tori Lin; Bobbi McCray; Nghi La; Trung Nguyen; Ariel Kauss; Donna Dambach; Dinah Misner; Hirdesh Uppal

Abstract Nicotinamide adenine dinucleotide (NAD) is an essential co-factor in glycolysis and is a key molecule involved in maintaining cellular energy metabolism. Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the rate-limiting step of an important salvage pathway in which nicotinamide is recycled into NAD. NAMPT is up-regulated in many types of cancer and NAMPT inhibitors (NAMPTi) have potential therapeutic benefit in cancer by impairing tumor metabolism. Clinical trials with NAMPTi APO-866 and GMX-1778, however, failed to reach projected efficacious exposures due to dose-limiting thrombocytopenia. We evaluated preclinical models for thrombocytopenia that could be used in candidate drug selection and risk mitigation strategies for NAMPTi-related toxicity. Rats treated with a suite of structurally diverse and potent NAMPTi at maximum tolerated doses had decreased reticulocyte and lymphocyte counts, but no thrombocytopenia. We therefore evaluated and qualified a human colony forming unit-megakaryocyte (CFU-MK) as in vitro predictive model of NAMPTi-induced MK toxicity and thrombocytopenia. We further demonstrate that the MK toxicity is on-target based on the evidence that nicotinic acid (NA), which is converted to NAD via a NAMPT-independent pathway, can mitigate NAMPTi toxicity to human CFU-MK in vitro and was also protective for the hematotoxicity in rats in vivo. Finally, assessment of CFU-MK and human platelet bioenergetics and function show that NAMPTi was toxic to MK and not platelets, which is consistent with the clinically observed time-course of thrombocytopenia.


Toxicological Sciences | 2016

Therapeutic Antibody-Induced Vascular Toxicity Due to Off-Target Activation of Nitric Oxide in Cynomolgus Monkeys.

Rama Pai; Ning Ma; Anu V. Connor; Dimitry M. Danilenko; Jacqueline M. Tarrant; Dany Salvail; Lisa Wong; Dylan P. Hartley; Dinah Misner; Eric Stefanich; Yan Wu; Yongmei Chen; Hong Wang; Donna Dambach

PRO304186, a humanized monoclonal antibody targeting soluble interleukin-17 A and F, was developed for autoimmune and inflammatory disease indications. When administered to cynomolgus monkeys PRO304186 induced unexpected adverse effects characterized by clinical signs of hematemesis, hematochezia, and moribundity. Pathology findings included hemorrhage throughout the gastrointestinal tract without any evidence of vascular wall damage or inflammatory cellular infiltration. Mechanistic investigation of these effects revealed mild elevations of serum MCP-1 and IL-12/23 but without a classical proinflammatory profile in PRO304186-treated animals. In vitro studies demonstrated off-target effects on vascular endothelial cells including activation of nitric oxide synthase leading to production of nitric oxide (NO) accompanied by increased mitochondrial membrane depolarization, glutathione depletion, and increased paracellular permeability. Additionally, endothelial cell-PRO304186-conditioned medium reduced myosin light chain phosphorylation in vascular smooth muscle cells. Furthermore, an ex vivo study utilizing segments from cynomolgus aorta and femoral artery confirmed PRO304186-induced endothelium-dependent smooth muscle relaxation and vasodilation mediated via NO. Finally, a single dose of PRO304186 in cynomolgus monkeys induced a rapid and pronounced increase in NO in the portal circulation that preceded a milder elevation of NO in the systemic circulation and corresponded temporally with systemic hypotension; findings consistent with NO-mediated vasodilation leading to hypotension. These changes were associated with non-inflammatory, localized hemorrhage in the gastrointestinal tract consistent with hemodynamic vascular injury associated with intense local vasodilation. Together, these data demonstrate that PRO304186-associated toxicity in monkeys was due to an off-target effect on endothelium that involved regional NO release resulting in severe systemic vasodilation, hypotension, and hemorrhage.


Journal of Pharmacology and Experimental Therapeutics | 2016

Bruton’s Tyrosine Kinase Small Molecule Inhibitors Induce a Distinct Pancreatic Toxicity in Rats

Rebecca Erickson; Leah Schutt; Jacqueline M. Tarrant; Michelle Mcdowell; Lichuan Liu; Adam R. Johnson; Sock-Cheng Lewin-Koh; Maj Hedehus; Jed Ross; Richard A. D. Carano; Karin Staflin; Fiona Zhong; James J. Crawford; Shelly Zhong; Karin Reif; Arna Katewa; Harvey Wong; Wendy B. Young; Donna Dambach; Dinah Misner

Bruton’s tyrosine kinase (BTK) is a member of the Tec family of cytoplasmic tyrosine kinases involved in B-cell and myeloid cell signaling. Small molecule inhibitors of BTK are being investigated for treatment of several hematologic cancers and autoimmune diseases. GDC-0853 ((S)-2-(3′-(hydroxymethyl)-1-methyl-5-((5-(2-methyl-4-(oxetan-3-yl)piperazin-1-yl)pyridin-2-yl)amino)-6-oxo-1,6-dihydro-[3,4′-bipyridin]-2′-yl)-7,7-dimethyl-3,4,7,8-tetrahydro-2H-cyclopenta[4,5]pyrrolo[1,2-a]pyrazin-1(6H)-one) is a selective and reversible oral small-molecule BTK inhibitor in development for the treatment of rheumatoid arthritis and systemic lupus erythematosus. In Sprague-Dawley (SD) rats, administration of GDC-0853 and other structurally diverse BTK inhibitors for 7 days or longer caused pancreatic lesions consisting of multifocal islet-centered hemorrhage, inflammation, fibrosis, and pigment-laden macrophages with adjacent lobular exocrine acinar cell atrophy, degeneration, and inflammation. Similar findings were not observed in mice or dogs at much higher exposures. Hemorrhage in the peri-islet vasculature emerged between four and seven daily doses of GDC-0853 and was histologically similar to spontaneously occurring changes in aging SD rats. This suggests that GDC-0853 could exacerbate a background finding in younger animals. Glucose homeostasis was dysregulated following a glucose challenge; however, this occurred only after 28 days of administration and was not directly associated with onset or severity of pancreatic lesions. There were no changes in other common serum biomarkers assessing endocrine and exocrine pancreatic function. Additionally, these lesions were not readily detectable via Doppler ultrasound, computed tomography, or magnetic resonance imaging. Our results indicate that pancreatic lesions in rats are likely a class effect of BTK inhibitors, which may exacerbate an islet-centered pathology that is unlikely to be relevant to humans.


Journal of Medicinal Chemistry | 2018

Discovery of GDC-0853: A Potent, Selective, and Noncovalent Bruton’s Tyrosine Kinase Inhibitor in Early Clinical Development

James J. Crawford; Adam R. Johnson; Dinah Misner; Lisa D. Belmont; Georgette Castanedo; Regina Choy; Melis Coraggio; Liming Dong; Charles Eigenbrot; Rebecca Erickson; Nico Ghilardi; Jonathan Hau; Arna Katewa; Pawan Bir Kohli; Wendy Lee; Joseph W. Lubach; Brent S. McKenzie; Daniel F. Ortwine; Leah Schutt; Suzanne Tay; Binqing Wei; Karin Reif; Lichuan Liu; Harvey Wong; Wendy B. Young

Brutons tyrosine kinase (Btk) is a nonreceptor cytoplasmic tyrosine kinase involved in B-cell and myeloid cell activation, downstream of B-cell and Fcγ receptors, respectively. Preclinical studies have indicated that inhibition of Btk activity might offer a potential therapy in autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. Here we disclose the discovery and preclinical characterization of a potent, selective, and noncovalent Btk inhibitor currently in clinical development. GDC-0853 (29) suppresses B cell- and myeloid cell-mediated components of disease and demonstrates dose-dependent activity in an in vivo rat model of inflammatory arthritis. It demonstrates highly favorable safety, pharmacokinetic (PK), and pharmacodynamic (PD) profiles in preclinical and Phase 2 studies ongoing in patients with rheumatoid arthritis, lupus, and chronic spontaneous urticaria. On the basis of its potency, selectivity, long target residence time, and noncovalent mode of inhibition, 29 has the potential to be a best-in-class Btk inhibitor for a wide range of immunological indications.


Toxicology in Vitro | 2018

In vitro assessment of chemotherapy-induced neuronal toxicity

Chelsea Snyder; Lanlan Yu; Tin Ngo; Daniel Sheinson; Yuda Zhu; Min Tseng; Dinah Misner; Karin Staflin

Neurotoxicity is a major concern during drug development, and together with liver and cardio-toxicity, it is one of the main causes of clinical drug attrition. Current pre-clinical models may not sufficiently identify and predict the risk for central or peripheral nervous system toxicity. One such example is clinically dose-limiting neuropathic effects after the administration of chemotherapeutic agents. Thus, the need to establish novel in vitro tools to evaluate the risk of neurotoxicities, such as neuropathy, remains unmet in drug discovery. Though in vitro studies have been conducted using primary and immortalized cell lines, some limitations include the utility for higher throughput methodologies, method reproducibility, and species extrapolation. As a novel alternative, human induced-pluripotent stem cell (iPSC)-derived neurons appear promising for testing new drug candidates. These iPSC-derived neurons are readily available and can be manipulated as required. Here, we describe a novel approach to assess neurotoxicity caused by different classes of chemotherapeutics using kinetic monitoring of neurite dynamic changes and apoptosis in human iPSC-neurons. These studies show promising changes in neurite dynamics in response to clinical inducers of neuropathy, as well as the ability to rank-order and gather mechanistic insight into class-specific compound induced neurotoxicity. This platform can be utilized in early drug development, as part of a weight of evidence approach, to screen drug candidates, and potentially reduce clinical attrition due to neurotoxicity.


Toxicology Research | 2014

For a series of methylindole analogs, reactive metabolite formation is a poor predictor of intrinsic cytotoxicity in human hepatocytes

Chenghong Zhang; Shuguang Ma; Erlie Marie Delarosa; Suzanne Tay; Jasleen Sodhi; Vivek Musinipally; Patricia Chang; Rama Pai; Jason S. Halladay; Dinah Misner; Jane R. Kenny; Cornelis E. C. A. Hop; S. Cyrus Khojasteh

The formation of reactive metabolites (RMs) can lead to in vitro cytotoxicity, and, therefore, intrinsic cytotoxicity studies in human hepatocytes are often conducted during drug discovery. We studied seven methylindole (MI) analogs (1MI–7MI) to determine whether cytotoxicity in human hepatocytes can be predicted on the basis of formation of glutathione (GSH) conjugates and/or time-dependent inhibition (TDI) against major cytochrome P450 (P450) enzymes in human liver microsomes. In GSH trapping studies, the bioactivation of 5MI resulted in formation of the highest level of GSH-related conjugates on the basis of mass spectrometric response. The remaining analogs ranked in the following order (high to low): 7MI > 4MI > 6MI > 2MI > 3MI > 1MI. Another noteworthy observation was the loss of pyroglutamic acid, derived from the glutamate residue of the GSH conjugate, for most MI analogs. With regard to TDI, all the MI analogs were weak inhibitors of the CYP3A enzyme. For CYP3A, the ratio of TDI parameters (kinact/KI; mL min−1 μmol−1) from the highest to the lowest were 0.71 for 5MI, 0.65 for 7MI, 0.48 for 2MI, 0.45 for 6MI, 0.29 for 4MI, 0.19 for 3MI and 0.15 for 1MI. Only 2MI caused cytotoxicity with a half maximal inhibitory concentration (IC50) of 306 μM in human hepatocytes. However, when cellular GSH levels were depleted by buthionine sulphoximine in hepatocytes, the cytotoxicity IC50 of 3MI significantly decreased from greater than 500 to 33.6 μM. On the basis of these results, there is a reasonable correlation between the relative levels of GSH-related conjugates formed and kinact/KI values of CYP3A; however, no apparent link exists between these values from human liver microsomes and in vitro cytotoxicity in hepatocytes for MI analogs.


Cardiovascular Toxicology | 2017

Cardiotoxicity Associated with Nicotinamide Phosphoribosyltransferase Inhibitors in Rodents and in Rat and Human-Derived Cells Lines

Dinah Misner; M. A. Kauss; Jatinder Singh; Hirdesh Uppal; A. Bruening-Wright; Bianca M. Liederer; Tori Lin; Bobbi McCray; Nghi La; Trung Nguyen; Deepak Sampath; Peter S. Dragovich; Thomas O’Brien; Tanja S. Zabka

Collaboration


Dive into the Dinah Misner's collaboration.

Researchain Logo
Decentralizing Knowledge