1
|
Vardigan JD, Pall PS, McDevitt DS, Huang C, Clements MK, Li Y, Kraus RL, Breslin MJ, Bungard CJ, Nemenov MI, Klukinov M, Burgey CS, Layton ME, Stachel SJ, Lange HS, Savitz AT, Santarelli VP, Henze DA, Uslaner JM. Analgesia and peripheral c-fiber modulation by selective Nav1.8 inhibition in rhesus. Pain 2024:00006396-990000000-00732. [PMID: 39382325 DOI: 10.1097/j.pain.0000000000003404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/01/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT Voltage-gated sodium (Nav) channels present untapped therapeutic value for better and safer pain medications. The Nav1.8 channel isoform is of particular interest because of its location on peripheral pain fibers and demonstrated role in rodent preclinical pain and neurophysiological assays. To-date, no inhibitors of this channel have been approved as drugs for treating painful conditions in human, possibly because of challenges in developing a sufficiently selective drug-like molecule with necessary potency not only in human but also across preclinical species critical to the preclinical development path of drug discovery. In addition, the relevance of rodent pain assays to the human condition is under increasing scrutiny as a number of mechanisms (or at the very least molecules) that are active in rodents have not translated to humans, and direct impact on pain fibers has not been confirmed in vivo. In this report, we have leveraged numerous physiological end points in nonhuman primates to evaluate the analgesic and pharmacodynamic activity of a novel, potent, and selective Nav1.8 inhibitor compound, MSD199. These pharmacodynamic biomarkers provide important confirmation of the in vivo impact of Nav1.8 inhibition on peripheral pain fibers in primates and have high translational potential to the clinical setting. These findings may thus greatly improve success of translational drug discovery efforts toward better and safer pain medications, as well as the understanding of primate biology of Nav1.8 inhibition broadly.
Collapse
Affiliation(s)
- Joshua D Vardigan
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Parul S Pall
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Dillon S McDevitt
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - ChienJung Huang
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Michelle K Clements
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Yuxing Li
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Richard L Kraus
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Michael J Breslin
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | | | | | | | - Chritopher S Burgey
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Mark E Layton
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Shawn J Stachel
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Henry S Lange
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Alan T Savitz
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Vincent P Santarelli
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Darrell A Henze
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Jason M Uslaner
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| |
Collapse
|
2
|
Breslin MJ, Schubert JW, Wang D, Huang C, Clements MK, Li Y, Zhou X, Vardigan JD, Kraus RL, Santarelli VP, Uslaner JM, Coleman PJ, Stachel SJ. 2-Aminopyridines as Potent and Selective Na v1.8 Inhibitors Exhibiting Efficacy in a Nonhuman Primate Pain Model. ACS Med Chem Lett 2024; 15:917-923. [PMID: 38894930 PMCID: PMC11181479 DOI: 10.1021/acsmedchemlett.4c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Herein we describe the discovery of a 2-aminopyridine scaffold as a potent and isoform selective inhibitor of the Nav1.8 sodium channel. Parallel library synthesis, guided by in silico predictions, rapidly transformed initial hits into a novel 2-aminopyridine lead class possessing good ADME and pharmacokinetic profiles that were able to display activity in a clinically translatable nonhuman primate capsaicin-sensitized thermode pharmacodynamic assay. Progress toward the lead identification, optimization, and in vivo efficacy of these compounds will be discussed.
Collapse
Affiliation(s)
- Michael J. Breslin
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Jeffrey W. Schubert
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Deping Wang
- Modeling
and Informatics, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Chienjung Huang
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Michelle K. Clements
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Yuxing Li
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Xiaoping Zhou
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Joshua D. Vardigan
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Richard L. Kraus
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Vincent P. Santarelli
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Jason M. Uslaner
- Preclinical
and Translational Medicine Discovery, Merck
& Co., Inc., 770
Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Paul J. Coleman
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Shawn J. Stachel
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| |
Collapse
|
3
|
Ichinose H, Natsume T, Yano M, Awaga Y, Hanada M, Takamatsu H, Matsuyama Y. Evaluation of brain activation related to resting pain using functional magnetic resonance imaging in cynomolgus macaques undergoing knee surgery. J Orthop 2024; 52:12-16. [PMID: 38404703 PMCID: PMC10881445 DOI: 10.1016/j.jor.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 02/09/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Functional magnetic resonance imaging (fMRI) visualizes hemodynamic responses associated with brain and spinal cord activation. Various types of pain have been objectively assessed using fMRI as considerable brain activations. This study aimed to develop a pain model in cynomolgus macaques undergoing knee surgery and confirm brain activation due to resting pain after knee surgery. Methods An osteochondral graft surgery on the femoral condyle in the unilateral knee was performed on four cynomolgus macaques (Macaca fascicularis). Resting pain was evaluated as changes in brain fMRI findings with a 3.0-T MRI scanner preoperatively, postoperatively, and after postoperative administration of morphine. In the fMRI analysis, Z-values >1.96 were considered statistically significant. Results Brain activation without stimulation after surgery in the cingulate cortex (3.09) and insular cortex (3.06) on the opposite side of the surgery was significantly greater than that before surgery (1.05 and 1.03, respectively) according to fMRI. After the administration of morphine, activation due to resting pain decreased in the cingulate cortex (1.38) and insular cortex (1.21). Conclusion Osteochondral graft surgery on the femoral condyle can lead to postoperative resting pain. fMRI can reveal activation in pain-related brain areas and evaluate resting pain due to knee surgery.
Collapse
Affiliation(s)
- Hatsumi Ichinose
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takahiro Natsume
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Mizuho Yano
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Yuji Awaga
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Mitsuru Hanada
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hiroyuki Takamatsu
- Pharmacology Group, Hamamatsu Pharma Research, Inc., Hamamatsu, Shizuoka, Japan
| | - Yukihiro Matsuyama
- Department of Orthopaedic Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
4
|
Tram M, Ibrahim T, Hovhannisyan A, Akopian A, Ruparel S. Lingual innervation in male and female marmosets. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2023; 14:100134. [PMID: 38099285 PMCID: PMC10719518 DOI: 10.1016/j.ynpai.2023.100134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 12/17/2023]
Abstract
Several gaps in knowledge exists in our understanding of orofacial pain. Some of these include type of peripheral sensory innervation in specific tissues, differences in innervation between sexes and validation of rodent studies in higher order species. The current study addresses these gaps by validating mouse studies for sensory innervation of tongue tissue in non-human primates as well as assesses sex-specific differences. Tongue and trigeminal ganglia were collected from naïve male and female marmosets and tested for nerve fibers using specific markers by immunohistochemistry and number of fibers quantified. We also tested whether specific subgroups of nerve fibers belonged to myelinating or non-myelinating axons. We observed that similar to findings in mice, marmoset tongue was innervated with nerve filaments expressing nociceptor markers like CGRP and TRPV1 as well as non-nociceptor markers like TrkB, parvalbumin (PV) and tyrosine hydroxylase (TH). Furthermore, we found that while portion of TrkB and PV may be sensory fibers, TH-positive fibers were primarily sympathetic nerve fibers. Moreover, number of CGRP, TrkB and TH-positive nerve fibers were similar in both sexes. However, we observed a higher proportion of myelinated TRPV1 positive fibers in females than in males as well as increased number of PV + fibers in females. Taken together, the study for the first time characterizes sensory innervation in non-human primates as well as evaluates sex-differences in innervation of tongue tissue, thereby laying the foundation for future orofacial pain research with new world smaller NHPs like the common marmoset.
Collapse
Affiliation(s)
- Meilinn Tram
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Tarek Ibrahim
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Anahit Hovhannisyan
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Armen Akopian
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| | - Shivani Ruparel
- Department of Endodontics, School of Dentistry, University of Texas Health San Antonio, USA
| |
Collapse
|
5
|
Paterson EA, Turner PV. Challenges with Assessing and Treating Pain in Research Primates: A Focused Survey and Literature Review. Animals (Basel) 2022; 12:2304. [PMID: 36078024 PMCID: PMC9455027 DOI: 10.3390/ani12172304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Research primates may undergo surgical procedures making effective pain management essential to ensure good animal welfare and unbiased scientific data. Adequate pain mitigation is dependent on whether veterinarians, technicians, researchers, and caregivers can recognize and assess pain, as well as the availability of efficacious therapeutics. A survey was conducted to evaluate primate veterinary approaches to pain assessment and alleviation, as well as expressed challenges for adequately managing primate pain. The survey (n = 93 respondents) collected information regarding institutional policies and procedures for pain recognition, methods used for pain relief, and perceived levels of confidence in primate pain assessment. Results indicated that 71% (n = 60) of respondents worked at institutions that were without formal experimental pain assessment policies. Pain assessment methods were consistent across respondents with the majority evaluating pain based on changes in general activity levels (100%, n = 86) and food consumption (97%, n = 84). Self-reported confidence in recognizing and managing pain ranged from slightly confident to highly confident, and there was a commonly expressed concern about the lack of objective pain assessment tools and science-based evidence regarding therapeutic recommendations of analgesics for research primates. These opinions correspond with significant gaps in the primate pain management literature, including limited specific pharmacokinetic data and efficacy testing for commonly used analgesics in research primate species as well as limited research on objective and specific measures of pain in research primates. These results demonstrate that there are inconsistencies in institutional policies and procedures surrounding pain management in research primates and a lack of objective pain assessment methods. Demonstrating the gaps and challenges in primate pain management can inform guideline development and suggest areas for future research.
Collapse
Affiliation(s)
- Emilie A. Paterson
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 0C4, Canada
| | - Patricia V. Turner
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 0C4, Canada
- Global Animal Welfare and Training, Charles River, Wilmington, MA 01887, USA
| |
Collapse
|
6
|
Kitano Y, Shinozuka T. Inhibition of Na V1.7: the possibility of ideal analgesics. RSC Med Chem 2022; 13:895-920. [PMID: 36092147 PMCID: PMC9384491 DOI: 10.1039/d2md00081d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/25/2022] [Indexed: 08/03/2023] Open
Abstract
The selective inhibition of NaV1.7 is a promising strategy for developing novel analgesic agents with fewer adverse effects. Although the potent selective inhibition of NaV1.7 has been recently achieved, multiple NaV1.7 inhibitors failed in clinical development. In this review, the relationship between preclinical in vivo efficacy and NaV1.7 coverage among three types of voltage-gated sodium channel (VGSC) inhibitors, namely conventional VGSC inhibitors, sulphonamides and acyl sulphonamides, is discussed. By demonstrating the PK/PD discrepancy of preclinical studies versus in vivo models and clinical results, the potential reasons behind the disconnect between preclinical results and clinical outcomes are discussed together with strategies for developing ideal analgesic agents.
Collapse
Affiliation(s)
- Yutaka Kitano
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| | - Tsuyoshi Shinozuka
- R&D Division, Daiichi Sankyo Co., Ltd. 1-2-58 Hiromachi Shinagawa-ku Tokyo 140-8710 Japan
| |
Collapse
|
7
|
Ballard JE, Pall PS, Vardigan J, Zhao F, Holahan MA, Zhou X, Jochnowitz N, Kraus RL, Klein RM, Henze DA, Houghton AK, Burgey CS, Gibson C, Struyk A. Translational Pharmacokinetic–Pharmacodynamic Modeling of NaV1.7 Inhibitor MK-2075 to Inform Human Efficacious Dose. Front Pharmacol 2021; 12:786078. [PMID: 35002718 PMCID: PMC8740778 DOI: 10.3389/fphar.2021.786078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
MK-2075 is a small-molecule selective inhibitor of the NaV1.7 channel investigated for the treatment of postoperative pain. A translational strategy was developed for MK-2075 to quantitatively interrelate drug exposure, target modulation, and the desired pharmacological response in preclinical animal models for the purpose of human translation. Analgesics used as a standard of care in postoperative pain were evaluated in preclinical animal models of nociceptive behavior (mouse tail flick latency and rhesus thermode heat withdrawal) to determine the magnitude of pharmacodynamic (PD) response at plasma concentrations associated with efficacy in the clinic. MK-2075 was evaluated in those same animal models to determine the concentration of MK-2075 required to achieve the desired level of response. Translation of MK-2075 efficacious concentrations in preclinical animal models to a clinical PKPD target in humans was achieved by accounting for species differences in plasma protein binding and in vitro potency against the NaV1.7 channel. Estimates of human pharmacokinetic (PK) parameters were obtained from allometric scaling of a PK model from preclinical species and used to predict the dose required to achieve the clinical exposure. MK-2075 exposure–response in a preclinical target modulation assay (rhesus olfaction) was characterized using a computational PKPD model which included a biophase compartment to account for the observed hysteresis. Translation of this model to humans was accomplished by correcting for species differences in PK NaV1.7 potency, and plasma protein binding while assuming that the kinetics of distribution to the target site is the same between humans and rhesus monkeys. This enabled prediction of the level of target modulation anticipated to be achieved over the dosing interval at the projected clinical efficacious human dose. Integration of these efforts into the early development plan informed clinical study design and decision criteria.
Collapse
Affiliation(s)
- Jeanine E. Ballard
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, NJ, United States
- *Correspondence: Jeanine E. Ballard,
| | - Parul S. Pall
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Joshua Vardigan
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Fuqiang Zhao
- Translational Imaging Biomarkers, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Marie A. Holahan
- Translational Imaging Biomarkers, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Xiaoping Zhou
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Nina Jochnowitz
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Richard L. Kraus
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Rebecca M. Klein
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Darrell A. Henze
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Andrea K. Houghton
- Neuroscience Pharmacology, Merck & Co. Inc., Kenilworth, NJ, United States
| | | | - Christopher Gibson
- Pharmacokinetics Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, NJ, United States
| | - Arie Struyk
- Translational Medicine, Merck & Co. Inc., Kenilworth, NJ, United States
| |
Collapse
|
8
|
Kraus RL, Zhao F, Pall PS, Zhou D, Vardigan JD, Danziger A, Li Y, Daley C, Ballard JE, Clements MK, Klein RM, Holahan MA, Greshock TJ, Kim RM, Layton ME, Burgey CS, Serra J, Henze DA, Houghton AK. Na v1.7 target modulation and efficacy can be measured in nonhuman primate assays. Sci Transl Med 2021; 13:13/594/eaay1050. [PMID: 34011626 DOI: 10.1126/scitranslmed.aay1050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/03/2020] [Accepted: 02/20/2021] [Indexed: 12/13/2022]
Abstract
Humans with loss-of-function mutations in the Nav1.7 channel gene (SCN9A) show profound insensitivity to pain, whereas those with gain-of-function mutations can have inherited pain syndromes. Therefore, inhibition of the Nav1.7 channel with a small molecule has been considered a promising approach for the treatment of various human pain conditions. To date, clinical studies conducted using selective Nav1.7 inhibitors have not provided analgesic efficacy sufficient to warrant further investment. Clinical studies to date used multiples of in vitro IC50 values derived from electrophysiological studies to calculate anticipated human doses. To increase the chance of clinical success, we developed rhesus macaque models of action potential propagation, nociception, and olfaction, to measure Nav1.7 target modulation in vivo. The potent and selective Nav1.7 inhibitors SSCI-1 and SSCI-2 dose-dependently blocked C-fiber nociceptor conduction in microneurography studies and inhibited withdrawal responses to noxious heat in rhesus monkeys. Pharmacological Nav1.7 inhibition also reduced odor-induced activation of the olfactory bulb (OB), measured by functional magnetic resonance imaging (fMRI) studies consistent with the anosmia reported in Nav1.7 loss-of-function patients. These data demonstrate that it is possible to measure Nav1.7 target modulation in rhesus macaques and determine the plasma concentration required to produce a predetermined level of inhibition. The calculated plasma concentration for preclinical efficacy could be used to guide human efficacious exposure estimates. Given the translatable nature of the assays used, it is anticipated that they can be also used in phase 1 clinical studies to measure target modulation and aid in the interpretation of phase 1 clinical data.
Collapse
Affiliation(s)
- Richard L Kraus
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA.
| | - Fuqiang Zhao
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Parul S Pall
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Dan Zhou
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Joshua D Vardigan
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Andrew Danziger
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Yuxing Li
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Christopher Daley
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Jeanine E Ballard
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Michelle K Clements
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Rebecca M Klein
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Marie A Holahan
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Thomas J Greshock
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Ronald M Kim
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Mark E Layton
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Christopher S Burgey
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Jordi Serra
- Department of Clinical Neurophysiology, Ruskin Wing, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Darrell A Henze
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| | - Andrea K Houghton
- Merck & Co. Inc., WP-14, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, USA
| |
Collapse
|
9
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
10
|
Application of Pharmacokinetic-Pharmacodynamic Modeling to Inform Translation of In Vitro NaV1.7 Inhibition to In Vivo Pharmacological Response in Non-human Primate. Pharm Res 2020; 37:181. [PMID: 32888082 PMCID: PMC7473964 DOI: 10.1007/s11095-020-02914-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/18/2020] [Indexed: 12/25/2022]
Abstract
Purpose This work describes a staged approach to the application of pharmacokinetic-pharmacodynamic (PK-PD) modeling in the voltage-gated sodium ion channel (NaV1.7) inhibitor drug discovery effort to address strategic questions regarding in vitro to in vivo translation of target modulation. Methods PK-PD analysis was applied to data from a functional magnetic resonance imaging (fMRI) technique to non-invasively measure treatment mediated inhibition of olfaction signaling in non-human primates (NHPs). Initial exposure-response was evaluated using single time point data pooled across 27 compounds to inform on in vitro to in vivo correlation (IVIVC). More robust effect compartment PK-PD modeling was conducted for a subset of 10 compounds with additional PD and PK data to characterize hysteresis. Results The pooled compound exposure-response facilitated an early exploration of IVIVC with a limited dataset for each individual compound, and it suggested a 2.4-fold in vitro to in vivo scaling factor for the NaV1.7 target. Accounting for hysteresis with an effect compartment PK-PD model as compounds advanced towards preclinical development provided a more robust determination of in vivo potency values, which resulted in a statistically significant positive IVIVC with a slope of 1.057 ± 0.210, R-squared of 0.7831, and p value of 0.006. Subsequent simulations with the PK-PD model informed the design of anti-nociception efficacy studies in NHPs. Conclusions A staged approach to PK-PD modeling and simulation enabled integration of in vitro NaV1.7 potency, plasma protein binding, and pharmacokinetics to describe the exposure-response profile and inform future study design as the NaV1.7 inhibitor effort progressed through drug discovery. Electronic supplementary material The online version of this article (10.1007/s11095-020-02914-9) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Coussens NP, Sittampalam GS, Jonson SG, Hall MD, Gorby HE, Tamiz AP, McManus OB, Felder CC, Rasmussen K. The Opioid Crisis and the Future of Addiction and Pain Therapeutics. J Pharmacol Exp Ther 2019; 371:396-408. [PMID: 31481516 DOI: 10.1124/jpet.119.259408] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/29/2019] [Indexed: 12/26/2022] Open
Abstract
Opioid misuse and addiction are a public health crisis resulting in debilitation, deaths, and significant social and economic impact. Curbing this crisis requires collaboration among academic, government, and industrial partners toward the development of effective nonaddictive pain medications, interventions for opioid overdose, and addiction treatments. A 2-day meeting, The Opioid Crisis and the Future of Addiction and Pain Therapeutics: Opportunities, Tools, and Technologies Symposium, was held at the National Institutes of Health (NIH) to address these concerns and to chart a collaborative path forward. The meeting was supported by the NIH Helping to End Addiction Long-TermSM (HEAL) Initiative, an aggressive, trans-agency effort to speed scientific solutions to stem the national opioid crisis. The event was unique in bringing together two research disciplines, addiction and pain, in order to create a forum for crosscommunication and collaboration. The output from the symposium will be considered by the HEAL Initiative; this article summarizes the scientific presentations and key takeaways. Improved understanding of the etiology of acute and chronic pain will enable the discovery of novel targets and regulatable pain circuits for safe and effective therapeutics, as well as relevant biomarkers to ensure adequate testing in clinical trials. Applications of improved technologies including reagents, assays, model systems, and validated probe compounds will likely increase the delivery of testable hypotheses and therapeutics to enable better health outcomes for patients. The symposium goals were achieved by increasing interdisciplinary collaboration to accelerate solutions for this pressing public health challenge and provide a framework for focused efforts within the research community. SIGNIFICANCE STATEMENT: This article summarizes key messages and discussions resulting from a 2-day symposium focused on challenges and opportunities in developing addiction- and pain-related medications. Speakers and attendees came from 40 states in the United States and 15 countries, bringing perspectives from academia, industry, government, and healthcare by researchers, clinicians, regulatory experts, and patient advocates.
Collapse
Affiliation(s)
- Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - G Sitta Sittampalam
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Samantha G Jonson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Heather E Gorby
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Amir P Tamiz
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Owen B McManus
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Christian C Felder
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| | - Kurt Rasmussen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland (N.P.C., G.S.S., S.G.J., M.D.H.); Orvos Communications, LLC (H.E.G.); National Institute of Neurologic Disorders and Stroke (A.P.T.) and National Institute on Drug Abuse (K.R.), National Institutes of Health, Bethesda, Maryland; Q-State Biosciences, Cambridge, Massachusetts (O.B.M.); and VP Discovery Research, Karuna Therapeutics, Boston, Massachusetts (C.C.F.)
| |
Collapse
|