David Immke
Amgen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by David Immke.
Molecular Pain | 2005
Derek C. Molliver; David Immke; Leonardo Fierro; Michel Paré; Frank L. Rice; Edwin W. McCleskey
BackgroundASIC3, the most sensitive of the acid-sensing ion channels, depolarizes certain rat sensory neurons when lactic acid appears in the extracellular medium. Two functions have been proposed for it: 1) ASIC3 might trigger ischemic pain in heart and muscle; 2) it might contribute to some forms of touch mechanosensation. Here, we used immunocytochemistry, retrograde labelling, and electrophysiology to ask whether the distribution of ASIC3 in rat sensory neurons is consistent with either of these hypotheses.ResultsLess than half (40%) of dorsal root ganglion sensory neurons react with anti-ASIC3, and the population is heterogeneous. They vary widely in cell diameter and express different growth factor receptors: 68% express TrkA, the receptor for nerve growth factor, and 25% express TrkC, the NT3 growth factor receptor. Consistent with a role in muscle nociception, small (<25 μm) sensory neurons that innervate muscle are more likely to express ASIC3 than those that innervate skin (51% of small muscle afferents vs. 28% of small skin afferents). Over 80% of ASIC3+ muscle afferents co-express CGRP (a vasodilatory peptide). Remarkably few (9%) ASIC3+ cells express P2X3 receptors (an ATP-gated ion channel), whereas 31% express TRPV1 (the noxious heat and capsaicin-activated ion channel also known as VR1). ASIC3+/CGRP+ sensory nerve endings were observed on muscle arterioles, the blood vessels that control vascular resistance; like the cell bodies, the endings are P2X3- and can be TRPV1+. The TrkC+/ASIC3+ cell bodies are uniformly large, possibly consistent with non-nociceptive mechanosensation. They are not proprioceptors because they fail two other tests: ASIC3+ cells do not express parvalbumin and they are absent from the mesencephalic trigeminal nucleus.ConclusionOur data indicates that: 1) ASIC3 is expressed in a restricted population of nociceptors and probably in some non-nociceptors; 2) co-expression of ASIC3 and CGRP, and the absence of P2X3, are distinguishing properties of a class of sensory neurons, some of which innervate blood vessels. We suggest that these latter afferents may be muscle metaboreceptors, neurons that sense the metabolic state of muscle and can trigger pain when there is insufficient oxygen.
Journal of Pharmacology and Experimental Therapeutics | 2007
Narender R. Gavva; Anthony W. Bannon; David N. Hovland; Sonya G. Lehto; Lana Klionsky; Sekhar Surapaneni; David Immke; Charles Henley; Leyla Arik; Annette Bak; James O. Davis; Nadia Ernst; Gal Hever; Rongzhen Kuang; Licheng Shi; Rami Tamir; Jue Wang; Weiya Wang; Gary Zajic; Dawn Zhu; Mark H. Norman; Jean-Claude Louis; Ella Magal; James J. S. Treanor
Capsaicin, the active ingredient in some pain-relieving creams, is an agonist of a nonselective cation channel known as the transient receptor potential vanilloid type 1 (TRPV1). The pain-relieving mechanism of capsaicin includes desensitization of the channel, suggesting that TRPV1 antagonism may be a viable pain therapy approach. In agreement with the above notion, several TRPV1 antagonists have been reported to act as antihyperalgesics. Here, we report the in vitro and in vivo characterization of a novel and selective TRPV1 antagonist, N-(4-[6-(4-trifluoromethyl-phenyl)-pyrimidin-4-yloxy]-benzothiazol-2-yl)-acetamide I (AMG 517), and compare its pharmacology with that of a closely related analog, tert-butyl-2-(6-([2-(acetylamino)-1,3-benzothiazol-4-yl]oxy)pyrimidin-4-yl)-5-(trifluoromethyl)phenylcarbamate (AMG8163). Both AMG 517 and AMG8163 potently and completely antagonized capsaicin, proton, and heat activation of TRPV1 in vitro and blocked capsaicin-induced flinch in rats in vivo. To support initial clinical investigations, AMG 517 was evaluated in a comprehensive panel of toxicology studies that included in vivo assessments in rodents, dogs, and monkeys. The toxicology studies indicated that AMG 517 was generally well tolerated; however, transient increases in body temperature (hyperthermia) were observed in all species after AMG 517 dosing. To further investigate this effect, we tested and showed that the antipyretic, acetaminophen, suppressed the hyperthermia caused by TRPV1 blockade. We also showed that repeated administration of TRPV1 antagonists attenuated the hyperthermia response, whereas the efficacy in capsaicin-induced flinch model was maintained. In conclusion, these studies suggest that the transient hyperthermia elicited by TRPV1 blockade may be manageable in the development of TRPV1 antagonists as therapeutic agents. However, the impact of TRPV1 antagonist-induced hyperthermia on their clinical utility is still unknown.
Journal of Medicinal Chemistry | 2011
Howard Bregman; Loren Berry; John L. Buchanan; April Chen; Bingfan Du; Elma Feric; Markus Hierl; Liyue Huang; David Immke; Brett Janosky; Danielle Johnson; Xingwen Li; Joseph Ligutti; Dong Liu; Annika B. Malmberg; David J. Matson; Jeff S. McDermott; Peter Miu; Hanh Nho Nguyen; Vinod F. Patel; Daniel Waldon; Ben Wilenkin; Xiao Mei Zheng; Anruo Zou; Erin F. DiMauro
Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.
Journal of Pharmacology and Experimental Therapeutics | 2015
Licheng Shi; Sonya G. Lehto; Dawn Zhu; Hong Sun; Jianhua Zhang; Brian Smith; David Immke; Kenneth D. Wild; Cen Xu
Therapeutic agents that block the calcitonin gene–related peptide (CGRP) signaling pathway are a highly anticipated and promising new drug class for migraine therapy, especially after reports that small-molecule CGRP-receptor antagonists are efficacious for both acute migraine treatment and migraine prevention. Using XenoMouse technology, we successfully generated AMG 334, a fully human monoclonal antibody against the CGRP receptor. Here we show that AMG 334 competes with [125I]-CGRP binding to the human CGRP receptor, with a Ki of 0.02 nM. AMG 334 fully inhibited CGRP-stimulated cAMP production with an IC50 of 2.3 nM in cell-based functional assays (human CGRP receptor) and was 5000-fold more selective for the CGRP receptor than other human calcitonin family receptors, including adrenomedullin, calcitonin, and amylin receptors. The potency of AMG 334 at the cynomolgus monkey (cyno) CGRP receptor was similar to that at the human receptor, with an IC50 of 5.7 nM, but its potency at dog, rabbit, and rat receptors was significantly reduced (>5000-fold). Therefore, in vivo target coverage of AMG 334 was assessed in cynos using the capsaicin-induced increase in dermal blood flow model. AMG 334 dose-dependently prevented capsaicin-induced increases in dermal blood flow on days 2 and 4 postdosing. These results indicate AMG 334 is a potent, selective, full antagonist of the CGRP receptor and show in vivo dose-dependent target coverage in cynos. AMG 334 is currently in clinical development for the prevention of migraine.
Journal of Medicinal Chemistry | 2012
Essa Hu; Ji Ma; Christopher Biorn; Dianna Lester-Zeiner; Robert Cho; Shannon Rumfelt; Roxanne Kunz; Thomas Nixey; Klaus Michelsen; Silke Miller; Jianxia Shi; Jamie Wong; Geraldine Hill Della Puppa; Jessica Able; Santosh Talreja; Dah-Ren Hwang; Stephen A. Hitchcock; Amy Porter; David Immke; Jennifer R. Allen; James J. S. Treanor; Hang Chen
A radiolabeled tracer for imaging therapeutic targets in the brain is a valuable tool for lead optimization in CNS drug discovery and for dose selection in clinical development. We report the rapid identification of a novel phosphodiesterase 10A (PDE10A) tracer candidate using a LC-MS/MS technology. This structurally distinct PDE10A tracer, AMG-7980 (5), has been shown to have good uptake in the striatum (1.2% ID/g tissue), high specificity (striatum/thalamus ratio of 10), and saturable binding in vivo. The PDE10A affinity (K(D)) and PDE10A target density (B(max)) were determined to be 0.94 nM and 2.3 pmol/mg protein, respectively, using [(3)H]5 on rat striatum homogenate. Autoradiography on rat brain sections indicated that the tracer signal was consistent with known PDE10A expression pattern. The specific binding of [(3)H]5 to rat brain was blocked by another structurally distinct, published PDE10A inhibitor, MP-10. Lastly, our tracer was used to measure in vivo PDE10A target occupancy of a PDE10A inhibitor in rats using LC-MS/MS technology.
Bioorganic & Medicinal Chemistry Letters | 2010
Vijay Keshav Gore; Vu Van Ma; Ruoyuan Yin; Joe Ligutti; David Immke; Elizabeth M. Doherty; Mark H. Norman
The membrane bound large-conductance, calcium-activated potassium channel (BKCa) is an important regulator of neuronal activity. Here we describe the identification and structure-activity relationship of a novel class of potent tetrahydroquinoline BKCa agonists. An example from this class of BKCa agonists was shown to depress the spontaneous neuronal discharges in an electrophysiological model of migraine.
International Journal of Neuroscience | 2016
Ruoyuan Yin; Dong Liu; Mark Chhoa; Chi-Ming Li; Yi Luo; Maosheng Zhang; Sonya G. Lehto; David Immke; Bryan D. Moyer
The nine members of the voltage-gated sodium channel (Nav) family mediate inward sodium currents that depolarize neurons and lead to action potential firing. Increased Nav expression and function in sensory ganglia may drive ectopic action potentials and result in neuropathic pain. Using patch-clamp electrophysiology and molecular biology techniques, experiments were performed to elucidate the contribution of Nav channels to sodium currents in rat dorsal root ganglion (DRG) neurons following the L5/L6 spinal nerve ligation (SNL) model of neuropathic pain. The abundance of DRG neurons with fast, tetrodotoxin sensitive (TTX-S) currents was seven-fold higher whereas the abundance of DRG neurons with slow, tetrodotoxin resistant (TTX-R) currents was nearly thirty-fold lower when comparing ipsilateral (injured) to contralateral (uninjured) neurons. TTX-S currents were elevated in larger neurons while TTX-R currents were reduced in both small and large neurons. Among Nav transcripts encoding TTX-R channels, Scn10a (Nav1.8) and Scn11a (Nav1.9) expression was twenty- to thirty-fold lower, while among Nav transcripts encoding TTX-S channels, Scn3a (Nav1.3) expression was four-fold higher in injured compared to uninjured DRG by qRT-PCR analysis. In summary, the SNL model of neuropathic pain induced a phenotypic switch in Nav expression from TTX-R to TTX-S channels in injured DRG neurons. Transcriptional reprogramming of Nav genes may drive ectopic action potential firing and contribute to neuropathic pain.
Journal of Pharmacology and Experimental Therapeutics | 2015
Hang Chen; Dianna Lester-Zeiner; Jianxia Shi; Silke Miller; Charles Glaus; Essa Hu; N. Chen; Jessica Able; Christopher Biorn; J. Wong; Ji Ma; Klaus Michelsen; G. Hill Della Puppa; Tim Kazules; H. H. Dou; S. Talreja; Xiaoning Zhao; A. Chen; Shannon Rumfelt; Roxanne Kunz; H. Ye; O. R. Thiel; Toni Williamson; C. Davis; Amy Porter; David Immke; J. R. Allen; James J. S. Treanor
Phosphodiesterase 10A (PDE10A) inhibitors have therapeutic potential for the treatment of psychiatric and neurologic disorders, such as schizophrenia and Huntington’s disease. One of the key requirements for successful central nervous system drug development is to demonstrate target coverage of therapeutic candidates in brain for lead optimization in the drug discovery phase and for assisting dose selection in clinical development. Therefore, we identified AMG 580 [1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)-2-fluoropropan-1-one], a novel, selective small-molecule antagonist with subnanomolar affinity for rat, primate, and human PDE10A. We showed that AMG 580 is suitable as a tracer for lead optimization to determine target coverage by novel PDE10A inhibitors using triple-stage quadrupole liquid chromatography–tandem mass spectrometry technology. [3H]AMG 580 bound with high affinity in a specific and saturable manner to both striatal homogenates and brain slices from rats, baboons, and human in vitro. Moreover, [18F]AMG 580 demonstrated prominent uptake by positron emission tomography in rats, suggesting that radiolabeled AMG 580 may be suitable for further development as a noninvasive radiotracer for target coverage measurements in clinical studies. These results indicate that AMG 580 is a potential imaging biomarker for mapping PDE10A distribution and ensuring target coverage by therapeutic PDE10A inhibitors in clinical studies.
Journal of Headache and Pain | 2014
Cen Xu; Licheng Shi; S Rao; C King; Hong Sun; Dawn Zhu; Sonya G. Lehto; Kenneth D. Wild; David Immke
Results AMG 334 is a potent inhibitor of [125I]-CGRP binding to the human CGRP receptor with a Ki of 0.02 nM. It exhibited full inhibition of CGRP-stimulated cAMP production with an IC50 of 2.3 nM in cell-based functional assays. Potency of AMG 334 at the cyno CGRP receptor is similar to that at the human receptor, but with significant reduced potency at dog, rabbit and rat receptors. AMG 334 also demonstrates > 5000-fold selectivity over other closely related receptors in the family. The receptor kinetics studies using [125I]-AMG 334 reveals a dissociation t1/2 off of 67 min. In the cyno study, AMG 334 produces a significant and sustained inhibitory effect on capsaicin-induced increase in dermal blood flow.
European Journal of Pain | 2007
Sonya G. Lehto; Rami Tamir; Dawn Zhu; A. Le; B. Youngblood; David Immke; Mark H. Norman; Ella Magal; Jean Claude Louis; James J. S. Treanor; Narender R. Gavva
was evaluated in the streptozotocin (STZ) model for diabetic neuropathic pain, the vincristine model for chemotherapy-induced neuropathic pain, a bone cancer model, the monosodium iodo acetate (MIA) model for osteoarthritic pain and the tumour necrosis factor alpha (TNFa) model for chronic muscle pain. In each model various endpoints were assessed including thermal and tactile allodynia and thermal and tactile hyperalgesia. In the STZ model lacosamide was active on all pain parameters. Moreover, when compared to clinically used analgesics such as amitryptiline, pregabalin, gabapentin, levetiracetam, lamotrigine or venlafaxine lacosamide was the compound with the broadest efficacy. Lacosamide was also active in models for cancer pain, as evidenced by potent effects against vincristineinduced hyperalgesia and bone cancer pain. Furthermore, muscle hyperalgesia induced by TNFa was more potently reduced by lacosamide as compared to pregabalin. Finally, lacosamide attenuated arthritic pain induced by MIA in rats. These results suggest that lacosamide may specifically have antihyperalgesic activity under conditions of chronic neuropathic, cancer, inflammatory and musculoskeletal pain.