267
Views
0
CrossRef citations to date
0
Altmetric
Review

Preclinical target validation for non-addictive therapeutics development for pain

, , ORCID Icon, ORCID Icon, , , , , , , , , , , , , , , ORCID Icon, , , & show all
Pages 811-822 | Received 16 Jun 2022, Accepted 09 Nov 2022, Published online: 24 Nov 2022

References

  • Dahlhamer J, Lucas J, Zelaya C, et al. Prevalence of chronic pain and high-impact chronic pain among adults — United States, 2016. MMWR Morb Mortal Wkly Rep. 2018 Sep 14 67(36):1001–1006.
  • Von Korff M, Scher AI, Helmick C, et al. United States national pain strategy for population research: concepts, definitions, and pilot data. J Pain. 2016 Oct;17(10):1068–1080.
  • Collins FS, Koroshetz WJ, Volkow ND. Helping to end addiction over the long-term: the research plan for the NIH HEAL initiative. Jama. 2018 Jul 10;320(2):129–130.
  • Thomas D, Wessel C. The State of Innovation in highly prevalent chronic diseases: biotechnology innovation organization (BIO). Pain Addict Ther. 2018;2:1–17.
  • Woolf CJ. Capturing novel non-opioid pain targets. Biol Psychiatry. 2020 Jan 1;87(1):74–81. .
  • Volkow ND, Icaza MEM, Poznyak V, et al. Addressing the opioid crisis globally. World Psychiatry. 2019 Jun;18(2):231–232.
  • Alsaloum M, Higerd GP, Effraim PR, et al. Status of peripheral sodium channel blockers for non-addictive pain treatment. Nat Rev Neurol. 2020 Dec;16(12):689–705.
  • Barbanti P, Aurilia C, Fofi L, et al. The role of anti-CGRP antibodies in the pathophysiology of primary headaches. Neurol Sci. 2017 May;38(Suppl 1):31–35.
  • Baker RG, Koroshetz WJ, Volkow ND. The helping to end addiction long-term (HEAL) initiative of the national institutes of health. Jama. 2021 Sep 21;326(11):1005–1006. .
  • Emmerich CH, Gamboa LM, Hofmann MCJ, et al. Improving target assessment in biomedical research: the GOT-IT recommendations. Nat Rev Drug Discov. 2021 Jan;20(1):64–81.
  • Prinz F, Schlange T, Asadullah K. Believe it or not: how much can we rely on published data on potential drug targets? Nat Rev Drug Discov. 2011 Aug 31; 10(9):712.
  • Begley CG, Ellis LM. Drug development: raise standards for preclinical cancer research. Nature. 2012 Mar 28; 483(7391):531–533.
  • Landis SC, Amara SG, Asadullah K, et al. A call for transparent reporting to optimize the predictive value of preclinical research. Nature. 2012 Oct 11;490(7419):187–191.
  • Percie du Sert N, Hurst V, Ahluwalia A, et al. The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. PLoS Biol. 2020 Jul;18(7):e3000410.
  • Carvalho-Silva D, Pierleoni A, Pignatelli M, et al. Open targets platform: new developments and updates two years on. Nucleic Acids Res. 2019 Jan 8 47(D1):D1056–d1065.
  • Davis KD, Aghaeepour N, Ahn AH, et al. Discovery and validation of biomarkers to aid the development of safe and effective pain therapeutics: challenges and opportunities. Nat Rev Neurol. 2020 Jul;16(7):381–400.
  • King EA, Davis JW, Degner JF. Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval. PLoS Genet. 2019 Dec;15(12):e1008489.
  • Fillingim RB, Wallace MR, Herbstman DM, et al. Genetic contributions to pain: a review of findings in humans. Oral Dis. 2008 Nov;14(8):673–682.
  • Minde J, Toolanen G, Andersson T, et al. Familial insensitivity to pain (HSAN V) and a mutation in the NGFB gene. A neurophysiological and pathological study. Muscle Nerve. 2004 Dec;30(6):752–760.
  • Mardy S, Miura Y, Endo F, et al. Congenital insensitivity to pain with anhidrosis (CIPA): effect of TRKA (NTRK1) missense mutations on autophosphorylation of the receptor tyrosine kinase for nerve growth factor. Hum Mol Genet. [2001 Feb 1];10(3):179–188.
  • Drenth JP, Waxman SG. Mutations in sodium-channel gene SCN9A cause a spectrum of human genetic pain disorders. J Clin Invest. 2007 Dec;117(12):3603–3609.
  • Sudlow C, Gallacher J, Allen N, et al. UK biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med. 2015 Mar;12(3):e1001779.
  • FinnGen [Internet]. [cited October 19, 2020]. Available from: https://www.finngen.fi.
  • Singh T, Poterba T, Curtis D et al, , Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature. 2022 Apr;604(7906):509–516.
  • Meloto CB, Benavides R, Lichtenwalter RN, et al. Human pain genetics database: a resource dedicated to human pain genetics research. Pain. 2018 Apr;159(4):749–763.
  • Ray P, Torck A, Quigley L, et al. Comparative transcriptome profiling of the human and mouse dorsal root ganglia: an RNA-seq-based resource for pain and sensory neuroscience research. Pain. 2018 Jul;159(7):1325–1345.
  • Raina PS, Wolfson C, Kirkland SA, et al. The Canadian longitudinal study on aging (CLSA). Can J Aging. 2009 Sep;28(3):221–229.
  • Renthal W, Chamessian A, Curatolo M, et al. Human cells and networks of pain: transforming pain target identification and therapeutic development. Neuron. [2021 May 5];109(9):1426–1429.
  • Kaye AD, Garcia AJ, Hall OM, et al. Update on the pharmacogenomics of pain management. Pharmgenomics Pers Med. 2019;12:125–143.
  • Haberberger RV, Barry C, Dominguez N, et al. Human Dorsal Root Ganglia. Front Cell Neurosci. 2019;13:271.
  • Arendt-Nielsen L. Pain sensitisation in osteoarthritis. Clin Exp Rheumatol. 2017 Sep-Oct;35(Suppl 1075):68–74.
  • Backonja MM, Attal N, Baron R, et al. Value of quantitative sensory testing in neurological and pain disorders: neuPSIG consensus. Pain. 2013 Sep;154(9):1807–1819.
  • Treede RD. The role of quantitative sensory testing in the prediction of chronic pain. Pain. 2019 May;160 Suppl 1:S66–s69.
  • Reimer M, Forstenpointner J, Hartmann A, et al. Sensory bedside testing: a simple stratification approach for sensory phenotyping. Pain Rep. 2020 May-Jun;5(3):e820.
  • Arendt-Nielsen L. Central sensitization in humans: assessment and pharmacology. Handb Exp Pharmacol. 2015;227:79–102.
  • Baron R, Maier C, Attal N, et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain. 2017 Feb;158(2):261–272.
  • Binder A, May D, Baron R, et al. Transient receptor potential channel polymorphisms are associated with the somatosensory function in neuropathic pain patients. PLoS One. [2011 Mar 29];6(3):e17387.
  • Sachau J, Bruckmueller H, Gierthmühlen J, et al. The serotonin receptor 2A (HTR2A) rs6313 variant is associated with higher ongoing pain and signs of central sensitization in neuropathic pain patients. Eur J Pain. 2021 Mar;25(3):595–611.
  • Demant DT, Lund K, Vollert J, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014 Nov;155(11):2263–2273.
  • Olesen SS, Graversen C, Bouwense SA, et al. Quantitative sensory testing predicts pregabalin efficacy in painful chronic pancreatitis. PLoS One. 2013;8(3):e57963.
  • Gustorff B, Poole C, Kloimstein H, et al. Treatment of neuropathic pain with the capsaicin 8% patch: quantitative sensory testing (QST) in a prospective observational study identifies potential predictors of response to capsaicin 8% patch treatment. Scand J Pain. [2013 Jul 1];4(3):138–145.
  • Zis P, Liampas A, Artemiadis A, et al. EEG recordings as biomarkers of pain perception: where do we stand and where to go? Pain Ther. 2022 Jun;11(2):369–380.
  • Tracey I, Woolf CJ, Andrews NA. Composite pain biomarker signatures for objective assessment and effective treatment. Neuron. 2019 Mar 6;101(5):783–800.
  • Namer B, Schmidt D, Eberhardt E, et al. Pain relief in a neuropathy patient by lacosamide: proof of principle of clinical translation from patient-specific iPS cell-derived nociceptors. EBioMedicine. 2019;39:401–408.
  • Ackerley R, Watkins RH. Microneurography as a tool to study the function of individual C-fiber afferents in humans: responses from nociceptors, thermoreceptors, and mechanoreceptors. J Neurophysiol. 2018 Dec 1;120(6):2834–2846.
  • Garcia-Perez E, Schönberger T, Sumalla M, et al. Behavioural, morphological and electrophysiological assessment of the effects of type 2 diabetes mellitus on large and small nerve fibres in Zucker diabetic fatty, Zucker lean and Wistar rats. Eur J Pain. 2018 Sep;22(8):1457–1472.
  • Serra J. Re-emerging microneurography. J Physiol. 2009 Jan 15;587(2):295–296.
  • Kiernan MC, Bostock H, Park SB, et al. Measurement of axonal excitability: consensus guidelines. Clin Neurophysiol. 2020 Jan;131(1):308–323.
  • Palomés-Borrajo G, Badia J, Navarro X, et al. Nerve excitability and neuropathic pain is reduced by BET protein inhibition after spared nerve injury. J Pain. 2021 Jun 19; 22:1617–1630.
  • Parker JL, Shariati NH, Karantonis DM. Electrically evoked compound action potential recording in peripheral nerves. Bioelectro Med. 2018;1(1):71–83.
  • Morton DL, Sandhu JS, Jones AK. Brain imaging of pain: state of the art. J Pain Res. 2016;9:613–624.
  • Panchision DM. Concise review: progress and challenges in using human stem cells for biological and therapeutics discovery: neuropsychiatric disorders. Stem Cells. 2016 Mar;34(3):523–536.
  • Hirose M, Kuroda Y, Murata E. NGF/TrkA signaling as a therapeutic target for pain. Pain Pract. 2016 Feb;16(2):175–182.
  • Han L, Liu Y, Xiong H, et al. CGRP monoclonal antibody for preventive treatment of chronic migraine: an update of meta-analysis. Brain Behav. 2019 Feb;9(2):e01215.
  • Naka A. An iPSC model reveals mechanisms of interindividual differences in pain. J Neurosci. 2019 Jul 10;39(28):5422–5423.
  • Chrysostomidou L, Cooper AH, Weir GA. Cellular models of pain: new technologies and their potential to progress preclinical research. Neurobiol Pain. 2021 [2021/Aug/01];10:100063.
  • Broberg M, Hästbacka J, Helle E. From stem cells to populations-using hiPSC, next-generation sequencing, and GWAS to explore the genetic and molecular mechanisms of congenital heart defects. Genes (Basel). 2021 Jun 16. 12(6):921.
  • Kanfer G, Sarraf SA, Maman Y, et al. Image-based pooled whole-genome CRISPRi screening for subcellular phenotypes. J Cell Biol. 2021 Feb 1;220:(2):e202006180.
  • Wheeler EC, Vu AQ, Einstein JM, et al. Pooled CRISPR screens with imaging on microraft arrays reveals stress granule-regulatory factors. Nat Methods. 2020 Jun;17(6):636–642.
  • Wilkinson IVL, Terstappen GC, Russell AJ Combining experimental strategies for successful target deconvolution. Drug Discov Today. 2020 Sep 21;25:1998–2005.
  • A critical evaluation of animal pain models 2019 [ updated January 30, 2019 - Januray 31, 2019, cited 2020 Oct 19]. Available from: https://www.ninds.nih.gov/News-Events/Events-Proceedings/Events/NIH-Workshop-Critical-Evaluation-Animal-Pain-Models
  • Whiteside GT, Adedoyin A, Leventhal L. Predictive validity of animal pain models? A comparison of the pharmacokinetic-pharmacodynamic relationship for pain drugs in rats and humans. Neuropharmacology. 2008 Apr;54(5):767–775.
  • Malfait AM, Little CB. On the predictive utility of animal models of osteoarthritis. Arthritis Res Ther. 2015 Sep 14;17:225.
  • Abboud C, Duveau A, Bouali-Benazzouz R, et al. Animal models of pain: diversity and benefits. J Neurosci Methods. 2021 Jan 15;348:108997.
  • Fabry ME. Transgenic animal models of sickle cell disease. Experientia. 1993 Jan 15;49(1):28–36.
  • Parisien M, Samoshkin A, Tansley SN, et al. Genetic pathway analysis reveals a major role for extracellular matrix organization in inflammatory and neuropathic pain. Pain. 2019 Apr;160(4):932–944.
  • Rice ASC, Finnerup NB, Kemp HI, et al. Sensory profiling in animal models of neuropathic pain: a call for back-translation. Pain. 2018 May;159(5):819–824.
  • Da Silva JT, Seminowicz DA. Neuroimaging of pain in animal models: a review of recent literature. Pain Rep. 2019 Jul-Aug;4(4):e732.
  • Liang R, Broussard GJ, Tian L. Imaging chemical neurotransmission with genetically encoded fluorescent sensors. ACS Chem Neurosci. 2015 Jan 21;6(1):84–93.
  • Xie JY, Qu C, Patwardhan A, et al. Activation of mesocorticolimbic reward circuits for assessment of relief of ongoing pain: a potential biomarker of efficacy. Pain. 2014 Aug;155(8):1659–1666.
  • Sikandar S, Ronga I, Iannetti GD, et al. Neural coding of nociceptive stimuli-from rat spinal neurones to human perception. Pain. 2013 Aug;154(8):1263–1273.
  • Buntinx L, Vermeersch S, de Hoon J. Development of anti-migraine therapeutics using the capsaicin-induced dermal blood flow model. Br J Clin Pharmacol. 2015 Nov;80(5):992–1000.
  • Nowaczewska M, Straburzyński M, Meder G, et al. Changes in cerebral blood flow after erenumab treatment in good and non-responders-a pilot study of migraine patients. J Clin Med. 2021 Jun 7;10:(11):2523.
  • Joshi SK, Hernandez G, Mikusa JP, et al. Comparison of antinociceptive actions of standard analgesics in attenuating capsaicin and nerve-injury-induced mechanical hypersensitivity. Neuroscience. [2006 Dec 1];143(2):587–596.
  • Bishop T, Marchand F, Young AR, et al. Ultraviolet-B-induced mechanical hyperalgesia: a role for peripheral sensitisation. Pain. 2010 Jul;150(1):141–152.
  • Bishop T, Hewson DW, Yip PK, et al. Characterisation of ultraviolet-B-induced inflammation as a model of hyperalgesia in the rat. Pain. 2007 Sep;131(1–2):70–82.
  • Bishop T, Ballard A, Holmes H, et al. Ultraviolet-B induced inflammation of human skin: characterisation and comparison with traditional models of hyperalgesia. Eur J Pain. 2009 May;13(5):524–532.
  • Dawes JM, Antunes-Martins A, Perkins JR, et al. Genome-wide transcriptional profiling of skin and dorsal root ganglia after ultraviolet-B-induced inflammation. PLoS One. 2014;9(4):e93338.
  • Mogil JS. Animal models of pain: progress and challenges. Nat Rev Neurosci. 2009 Apr;10(4):283–294.
  • Gregory NS, Harris AL, Robinson CR, et al. An overview of animal models of pain: disease models and outcome measures. J Pain. 2013 Nov;14(11):1255–1269.
  • Burma NE, Leduc-Pessah H, Fan CY, et al. Animal models of chronic pain: advances and challenges for clinical translation. J Neurosci Res. 2017;95(6):1242–1256.
  • Berge OG. Predictive validity of behavioural animal models for chronic pain. Br J Pharmacol. 2011 Oct;164(4):1195–1206.
  • Jackson SJ, Authier S, Brohmann H, et al. Neurofunctional test batteries in safety pharmacology - Current and emerging considerations for the drug development process. J Pharmacol Toxicol Methods. 2019;100:106602.
  • Yaksh TL, Woller SA, Ramachandran R, et al. The search for novel analgesics: targets and mechanisms. F1000Prime Rep. 2015;7:56.
  • Xie JY, Qu C, Munro G, et al. Antihyperalgesic effects of Meteorin in the rat chronic constriction injury model: a replication study. Pain. 2019 Aug;160(8):1847–1855.
  • Wodarski R, Delaney A, Ultenius C, et al. Cross-centre replication of suppressed burrowing behaviour as an ethologically relevant pain outcome measure in the rat: a prospective multicentre study. Pain. 2016 Oct;157(10):2350–2365.
  • Maher DP, Wong CH, Siah KW, et al. Estimates of probabilities of successful development of pain medications: an analysis of pharmaceutical clinical development programs from 2000 to 2020. Anesthesiology. [2022 Aug 1];137(2):243–251.
  • Thomas DWB, Justin A, John C, et al. Clinical development success rates 2006-2015. Biotechnol Innovation Organ (BIO). 2016:1–28.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.