Discovery and validation of biomarkers to aid the development of safe and effective pain therapeutics: challenges and opportunities

Karen D Davis, Nima Aghaeepour, Andrew H Ahn, Martin S Angst, David Borsook, Ashley Brenton, Michael E Burczynski, Christopher Crean, Robert Edwards, Brice Gaudilliere, Georgene W Hergenroeder, Michael J Iadarola, Smriti Iyengar, Yunyun Jiang, Jiang-Ti Kong, Sean Mackey, Carl Y Saab, Christine N Sang, Joachim Scholz, Marta Segerdahl, Irene Tracey, Christin Veasley, Jing Wang, Tor D Wager, Ajay D Wasan, Mary Ann Pelleymounter, Karen D Davis, Nima Aghaeepour, Andrew H Ahn, Martin S Angst, David Borsook, Ashley Brenton, Michael E Burczynski, Christopher Crean, Robert Edwards, Brice Gaudilliere, Georgene W Hergenroeder, Michael J Iadarola, Smriti Iyengar, Yunyun Jiang, Jiang-Ti Kong, Sean Mackey, Carl Y Saab, Christine N Sang, Joachim Scholz, Marta Segerdahl, Irene Tracey, Christin Veasley, Jing Wang, Tor D Wager, Ajay D Wasan, Mary Ann Pelleymounter

Abstract

Pain medication plays an important role in the treatment of acute and chronic pain conditions, but some drugs, opioids in particular, have been overprescribed or prescribed without adequate safeguards, leading to an alarming rise in medication-related overdose deaths. The NIH Helping to End Addiction Long-term (HEAL) Initiative is a trans-agency effort to provide scientific solutions to stem the opioid crisis. One component of the initiative is to support biomarker discovery and rigorous validation in collaboration with industry leaders to accelerate high-quality clinical research into neurotherapeutics and pain. The use of objective biomarkers and clinical trial end points throughout the drug discovery and development process is crucial to help define pathophysiological subsets of pain, evaluate target engagement of new drugs and predict the analgesic efficacy of new drugs. In 2018, the NIH-led Discovery and Validation of Biomarkers to Develop Non-Addictive Therapeutics for Pain workshop convened scientific leaders from academia, industry, government and patient advocacy groups to discuss progress, challenges, gaps and ideas to facilitate the development of biomarkers and end points for pain. The outcomes of this workshop are outlined in this Consensus Statement.

Conflict of interest statement

S.I., M.J.I. and M.A.P. are NIH employees. A.H.A. is an employee of Teva Pharmaceuticals. D.B. consults for Biogen. A.B. is the Chief Science Officer of Mycroft Bioanalytics, a precision medicine company focused on pain. C.Y.S. is Chief Scientific Officer at neurotecnix, a start-up that develops EEG-based pain biomarkers; a consultant for Asahi Kasei Pharmaceuticals, Japan and PainQX, USA; has received remuneration for symposia funded by the International Association for the Study of Pain, Medtronic, Boston Scientific and NIH/NINDS; and is an inventor on US patents 61/328,583, 9,486,632, 62/203,798 and 62/329,345, all of which relate to the detection or treatment of pain. C.N.S. has received funding from Merck, Helixmith and the Utley Foundation; has acted as a consultant for Lilly Research Laboratories, Covance, Genentech, Alkermes, Arena, Nevaker and Heron Therapeutics; is an inventor on European patent 01942162, US patent 8,309,507 and CA patent 2,499,987, all of which relate to the treatment of central neuropathic pain; and has non-financial competing interests as Director of Rick Hansen Institute, Chairman of Medical and Scientific Advisory Committee, United Spinal Association and membership on the Interagency Pain Research Coordinating Committee (NIH/HHS). J.S. is an employee and stock owner with Biogen. T.D.W. is an inventor on US patent US 2018/0055407. A.D.W. is a consultant for Analgesic Solutions and Pfizer, and has received an Investigator-Initiated Grant from Collegium Pharmaceuticals. The other authors declare no competing interests.

Figures

Fig. 1. Preclinical and human pain biomarkers.
Fig. 1. Preclinical and human pain biomarkers.
a | Development of preclinical pain biomarkers starts with induction of different modalities of pain that are clinically relevant. In the absence of a ground truth for pain in animals, a critical first step relies on converging lines of evidence from behavioural, electrophysiological and other overt signs. The next step is to demonstrate reversal of these signs using analgesic compounds with proven efficacy in humans. b | Development of human pain biomarkers starts with the individual’s self-report, also known as the ground truth (asterisks), and a set of signs and symptoms, with the goal of defining objective methods and criteria, as well as end points for assessing, predicting and/or classifying pain and analgesia. Thus, biomarkers are obtained to indicate chronic pain predisposition, pain mechanisms, diagnostic stratification, chronification, recovery and treatment outcome (response or failure).
Fig. 2. Steps to identify and develop…
Fig. 2. Steps to identify and develop biomarkers for clinical use.
The process starts with recognition of the need for a biomarker followed by discovery of candidate biomarkers. Assay development ensues. The type of assay selected is based on the properties of the biomarker or analyte. Specific detection of the analyte is required to move forward to the assay development phase. The analyte must be measurable and the detection method must be reliable and reproducible. During development, a prototype assay is tested with a test set of samples, including both positive and negative controls. As the assay is developed, conditions are optimized, and the prototype assay is then refined, tested and retested to ensure reliable, reproducible results. For an omics assay, this process may include optimizing the pH, reducing the background signal or filtering the biological fluid to remove signal interference (for example, from haemoglobin). Once the prototype assay is optimized and produces reliable, verifiable results on test sets of samples, it must be validated using a naive sample set. Validation must be performed without knowledge of patient status to eliminate any bias in interpretation of results. If specific detection of the analyte is demonstrated, prospective validation is performed. Reproducible, reliable, sensitive and specific biomarker detection positions a biomarker for clinical use.

References

    1. Treede RD, et al. Chronic pain as a symptom or a disease: the IASP classification of chronic pain for the International Classification of Diseases (ICD-11) Pain. 2019;160:19–27.
    1. Von Korff M, et al. United States National Pain Strategy for population research: concepts, definitions, and pilot data. J. Pain. 2016;17:1068–1080.
    1. GBD 2017 Disease and Injury Incidence and Prevalence Collaborators Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392:1789–1858.
    1. Nahin RL, Sayer B, Stussman BJ, Feinberg TM. Eighteen-year trends in the prevalence of, and health care use for, noncancer pain in the United States: data from the Medical Expenditure Panel Survey. J. Pain. 2019;20:796–809.
    1. US Institute of Medicine. Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research (National Academies, 2011).
    1. Dahlhamer J, et al. Prevalence of chronic pain and high-impact chronic pain among adults — United States, 2016. MMWR Morb. Mortal. Wkly. Rep. 2018;67:1001–1006.
    1. Gatchel RJ, et al. Research agenda for the prevention of pain and its impact: report of the Work Group on the Prevention of Acute and Chronic Pain of the Federal Pain Research Strategy. J. Pain. 2018;19:837–851.
    1. World Health Organization. Management of substance abuse. Information sheet on opioid overdose (WHO, 2018).
    1. US Substance Abuse and Mental Health Services Administration. Facing addiction in America: the Surgeon General’s report on alcohol, drugs, and health (US Department of Health and Human Services, 2016).
    1. Mackey S, Kao MC. Managing twin crises in chronic pain and prescription opioids. BMJ. 2019;364:l917.
    1. Pitcher MH, Von Korff M, Bushnell MC, Porter L. Prevalence and profile of high-impact chronic pain in the United States. J. Pain. 2019;20:146–160.
    1. FDA Center for Drug Evaluation and Research. Advancing health through innovation: 2018 new drug therapy approvals (FDA, 2019).
    1. Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contemp. Clin. Trials Commun. 2018;11:156–164.
    1. Thomas D, Wessel C. The state of innovation in highly prevalent chronic disease. BIO Ind. Anal. 2018;II:1–15.
    1. Ferber G. Biomarkers and proof of concept. Methods Find. Exp. Clin. Pharmacol. 2002;24(Suppl. C):35–40.
    1. Morgan P, et al. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat. Rev. Drug Discov. 2018;17:167–181.
    1. Thomas, D. W. et al. Clinical development success rates 2006–2015 (BIO, 2016).
    1. Nagakura Y. The need for fundamental reforms in the pain research field to develop innovative drugs. Expert Opin. Drug Discov. 2017;12:39–46.
    1. Niculescu AB, et al. Towards precision medicine for pain: diagnostic biomarkers and repurposed drugs. Mol. Psychiat. 2019;24:501–522.
    1. Wideman TH, et al. The multimodal assessment model of pain: a novel framework for further integrating the subjective pain experience within research and practice. Clin. J. Pain. 2019;35:212–221.
    1. Treede RD. The International Association for the Study of Pain definition of pain: as valid in 2018 as in 1979, but in need of regularly updated footnotes. Pain. Rep. 2018;3:e643.
    1. Bonafe FSS, de Campos LA, Maroco J, Campos J. Brief pain inventory: a proposal to extend its clinical application. Eur. J. Pain. 2019;23:565–576.
    1. Main CJ. Pain assessment in context: a state of the science review of the McGill pain questionnaire 40 years on. Pain. 2016;157:1387–1399.
    1. Bullock L, et al. Pain assessment and pain treatment for community-dwelling people with dementia: a systematic review and narrative synthesis. Int. J. Geriatr. Psychiat. 2019;34:807–821.
    1. Birnie KA, Hundert AS, Lalloo C, Nguyen C, Stinson JN. Recommendations for selection of self-report pain intensity measures in children and adolescents: a systematic review and quality assessment of measurement properties. Pain. 2019;160:5–18.
    1. Dansie EJ, Turk DC. Assessment of patients with chronic pain. Br. J. Anaesth. 2013;111:19–25.
    1. Smith SM, et al. Pain intensity rating training: results from an exploratory study of the ACTTION PROTECCT system. Pain. 2016;157:1056–1064.
    1. Vollert J, et al. Quantitative sensory testing using DFNS protocol in Europe: an evaluation of heterogeneity across multiple centers in patients with peripheral neuropathic pain and healthy subjects. Pain. 2016;157:750–758.
    1. Vollert J, et al. Stratifying patients with peripheral neuropathic pain based on sensory profiles: algorithm and sample size recommendations. Pain. 2017;158:1446–1455.
    1. Haanpaa M, et al. NeuPSIG guidelines on neuropathic pain assessment. Pain. 2011;152:14–27.
    1. Group BDW. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001;69:89–95.
    1. US Food and Drug Administration–National Institutes of Health Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource (FDA, 2016).
    1. European Medicines Agency. Guideline on the clinical investigation of medicines for the treatment of Alzheimer’s disease (EMA, 2018).
    1. FDA Center for Drug Evaluation and Research. Biomarker qualification: evidentiary framework (FDA, 2018).
    1. Wager TD, et al. An fMRI-based neurologic signature of physical pain. N. Engl. J. Med. 2013;368:1388–1397.
    1. Woo C-W, Wager TD. Neuroimaging-based biomarker discovery and validation. Pain. 2015;156:1379–1381.
    1. Davis KD, et al. Brain imaging tests for chronic pain: medical, legal and ethical issues and recommendations. Nat. Rev. Neurol. 2017;13:624–638.
    1. Kragel PA, Koban L, Barrett LF, Wager TD. Representation, pattern information, and brain signatures: from neurons to neuroimaging. Neuron. 2018;99:257–273.
    1. Woo C-W, Chang LJ, Lindquist MA, Wager TD. Building better biomarkers: brain models in translational neuroimaging. Nat. Neurosci. 2017;20:365–377.
    1. Kohoutová L, et al. Toward a unified framework for interpreting machine-learning models in neuroimaging. Nat. Protoc. 2020;15:1399–1435.
    1. Varoquaux G. Cross-validation failure: small sample sizes lead to large error bars. Neuroimage. 2018;180:68–77.
    1. Hastie, T., Tibshirani, R. & Friedman, J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction (Springer, 2013).
    1. Chang LJ, Gianaros PJ, Manuck SB, Krishnan A. A sensitive and specific neural signature for picture-induced negative affect. PLOS Biol. 2015;13:e1002180.
    1. Woo CW, et al. Quantifying cerebral contributions to pain beyond nociception. Nat. Commun. 2017;8:14211.
    1. Zunhammer M, Bingel U, Wager TD. Placebo effects on the neurologic pain signature: a meta-analysis of individual participant functional magnetic resonance imaging data. JAMA Neurol. 2018;75:1321–1330.
    1. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain and analgesia. Part 1: the need, reality, challenges, and solutions. Discov. Med. 2011;11:197–207.
    1. Borsook D, Becerra L, Hargreaves R. Biomarkers for chronic pain and analgesia. Part 2: how, where, and what to look for using functional imaging. Discov. Med. 2011;11:209–219.
    1. Bair E, et al. Identification of clusters of individuals relevant to temporomandibular disorders and other chronic pain conditions: the OPPERA study. Pain. 2016;157:1266–1278.
    1. Rolke R, et al. Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): standardized protocol and reference values. Pain. 2006;123:231–243.
    1. Diatchenko L, Fillingim RB, Smith SB, Maixner W. The phenotypic and genetic signatures of common musculoskeletal pain conditions. Nat. Rev. Rheumatol. 2013;9:340–350.
    1. Smith SM, et al. The potential role of sensory testing, skin biopsy, and functional brain imaging as biomarkers in chronic pain clinical trials: IMMPACT considerations. J. Pain. 2017;18:757–777.
    1. Ashraf AB, et al. The painful face — pain expression recognition using active appearance models. Image Vis. Comput. 2009;27:1788–1796.
    1. Bartlett MS, Littlewort GC, Frank MG, Lee K. Automatic decoding of facial movements reveals deceptive pain expressions. Curr. Biol. 2014;24:738–743.
    1. LaChapelle DL, Hadjistavropoulos T, Craig KD. Pain measurement in persons with intellectual disabilities. Clin. J. Pain. 1999;15:13–23.
    1. Sikka K, et al. Automated assessment of children’s postoperative pain using computer vision. Pediatrics. 2015;136:e124–e131.
    1. Branco A, Fekete SMW, Rugolo LMSS, Rehder MI. The newborn pain cry: descriptive acoustic spectrographic analysis. Int. J. Pediatr. Otorhinolaryngol. 2007;71:539–546.
    1. Cohn, J. F. et al. Detecting depression from facial actions and vocal prosody. Int. Conf. Affect. Comput. Intell. Interact. Workshops10.1109/ACII.2009.5349358 (2009).
    1. Gholami B, Haddad WM, Tannenbaum AR. Relevance vector machine learning for neonate pain intensity assessment using digital imaging. IEEE Trans. Biomed. Eng. 2010;57:1457–1466.
    1. Yang M, et al. A machine learning approach to assessing gait patterns for complex regional pain syndrome. Med. Eng. Phys. 2012;34:740–746.
    1. Nguyen QC, et al. Social media indicators of the food environment and state health outcomes. Public Health. 2017;148:120–128.
    1. Olausson H, Wessberg J, Morrison I, McGlone F, Vallbo A. The neurophysiology of unmyelinated tactile afferents. Neurosci. Biobehav. Rev. 2010;34:185–191.
    1. Serra J. Microneurography: towards a biomarker of spontaneous pain. Pain. 2012;153:1989–1990.
    1. Serra J, et al. Hyperexcitable C nociceptors in fibromyalgia. Ann. Neurol. 2014;75:196–208.
    1. Waxman, S. G. Chasing Men on Fire: The Story of the Search for a Pain Gene (MIT Press, 2018).
    1. Ploner M, Sorg C, Gross J. Brain rhythms of pain. Trends Cogn. Sci. 2017;21:100–110.
    1. Ploner M, May ES. Electroencephalography and magnetoencephalography in pain research — current state and future perspectives. Pain. 2018;159:206–211.
    1. Pinheiro ES, et al. Electroencephalographic patterns in chronic pain: a systematic review of the literature. PLOS ONE. 2016;11:e0149085.
    1. Peng W, et al. Brain oscillations reflecting pain-related behavior in freely moving rats. Pain. 2018;159:106–118.
    1. Nickel MM, et al. Brain oscillations differentially encode noxious stimulus intensity and pain intensity. Neuroimage. 2017;148:141–147.
    1. May ES, et al. Prefrontal gamma oscillations reflect ongoing pain intensity in chronic back pain patients. Hum. Brain Mapp. 2018;40:293–305.
    1. Leblanc BW, Lii TR, Silverman AE, Alleyne RT, Saab CY. Cortical theta is increased while thalamocortical coherence is decreased in rat models of acute and chronic pain. Pain. 2014;155:773–782.
    1. LeBlanc BW, Bowary PM, Chao YC, Lii TR, Saab CY. Electroencephalographic signatures of pain and analgesia in rats. Pain. 2016;157:2330–2340.
    1. LeBlanc BW, et al. T-type calcium channel blocker Z944 restores cortical synchrony and thalamocortical connectivity in a rat model of neuropathic pain. Pain. 2016;157:255–263.
    1. Koyama S, Xia J, Leblanc BW, Gu JW, Saab CY. Sub-paresthesia spinal cord stimulation reverses thermal hyperalgesia and modulates low frequency EEG in a rat model of neuropathic pain. Sci. Rep. 2018;8:7181.
    1. Koyama S, et al. An electroencephalography bioassay for preclinical testing of analgesic efficacy. Sci. Rep. 2018;6:16402.
    1. Llinas RR, Ribary U, Jeanmonod D, Kronberg E, Mitra PP. Thalamocortical dysrhythmia: a neurological and neuropsychiatric syndrome characterized by magnetoencephalography. Proc. Natl Acad. Sci. USA. 1999;96:15222–15227.
    1. Sarnthein J, Stern J, Aufenberg C, Rousson V, Jeanmonod D. Increased EEG power and slowed dominant frequency in patients with neurogenic pain. Brain. 2006;129:55–64.
    1. Stern J, Jeanmonod D, Sarnthein J. Persistent EEG overactivation in the cortical pain matrix of neurogenic pain patients. Neuroimage. 2006;31:721–731.
    1. Saab CY, Barrett LF. Thalamic bursts and the epic pain model. Front. Comput. Neurosci. 2016;10:147.
    1. LeBlanc BW, et al. Thalamic bursts down-regulate cortical theta and nociceptive behavior. Sci. Rep. 2017;7:2482.
    1. Mamas M, Dunn WB, Neyses L, Goodacre R. The role of metabolites and metabolomics in clinically applicable biomarkers of disease. Arch. Toxicol. 2011;85:5–17.
    1. Ramsden CE, et al. A systems approach for discovering linoleic acid derivatives that potentially mediate pain and itch. Sci. Signal. 2017;10:eaal5241.
    1. Dorsey SG, et al. Whole blood transcriptomic profiles can differentiate vulnerability to chronic low back pain. PLOS ONE. 2019;14:e0216539.
    1. Roses AD. Apolipoprotein E alleles as risk factors in Alzheimer’s disease. Annu. Rev. Med. 1996;47:387–400.
    1. Molinuevo JL, et al. Current state of Alzheimer’s fluid biomarkers. Acta Neuropathol. 2018;136:821–853.
    1. Blennow K, Mattsson N, Scholl M, Hansson O, Zetterberg H. Amyloid biomarkers in Alzheimer’s disease. Trends Pharmacol. Sci. 2015;36:297–309.
    1. Mattsson N, Cullen NC, Andreasson U, Zetterberg H, Blennow K. Association between longitudinal plasma neurofilament light and neurodegeneration in patients with Alzheimer disease. JAMA Neurol. 2019;76:791–799.
    1. McIntosh AM, et al. Genetic and environmental risk for chronic pain and the contribution of risk variants for major depressive disorder: a family-based mixed-model analysis. PLOS Med. 2016;13:e1002090.
    1. Gormley P, et al. Common variant burden contributes to the familial aggregation of migraine in 1,589 families. Neuron. 2018;99:1098.
    1. Zorina-Lichtenwalter K, Meloto CB, Khoury S, Diatchenko L. Genetic predictors of human chronic pain conditions. Neuroscience. 2016;338:36–62.
    1. Tracey I, Woolf CJ, Andrews NA. Composite pain biomarker signatures for objective assessment and effective treatment. Neuron. 2019;101:783–800.
    1. Sandoval J, Peiro-Chova L, Pallardo FV, Garcia-Gimenez JL. Epigenetic biomarkers in laboratory diagnostics: emerging approaches and opportunities. Exp.Rev. Mol. Diagn. 2013;13:457–471.
    1. Douglas SR, et al. Analgesic response to intravenous ketamine is linked to a circulating microRNA signature in female patients with complex regional pain syndrome. J. Pain. 2015;16:814–824.
    1. Ramanathan S, Ajit SK. MicroRNA-based biomarkers in pain. Adv. Pharmacol. 2016;75:35–62.
    1. Lopez-Gonzalez MJ, Landry M, Favereaux A. MicroRNA and chronic pain: from mechanisms to therapeutic potential. Pharmacol. Ther. 2017;180:1–15.
    1. Descalzi G, et al. Epigenetic mechanisms of chronic pain. Trends Neurosci. 2015;38:237–246.
    1. Raoof R, Willemen H, Eijkelkamp N. Divergent roles of immune cells and their mediators in pain. Rheumatology. 2018;57:429–440.
    1. Ji RR, Chamessian A, Zhang YQ. Pain regulation by non-neuronal cells and inflammation. Science. 2016;354:572–577.
    1. Tsai AS, et al. A year-long immune profile of the systemic response in acute stroke survivors. Brain. 2019;142:978–991.
    1. Aghaeepour N, et al. A proteomic clock of human pregnancy. Am. J. Obstet. Gynecol. 2018;218:347.e1–347.e14.
    1. Ghaemi MS, et al. Multiomics modeling of the immunome, transcriptome, microbiome, proteome and metabolome adaptations during human pregnancy. Bioinformatics. 2019;35:95–103.
    1. Ganz P, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA. 2016;315:2532–2541.
    1. Erez O, et al. The prediction of late-onset preeclampsia: results from a longitudinal proteomics study. PLOS ONE. 2017;12:e0181468.
    1. Aghaeepour N, et al. Deep immune profiling of an arginine-enriched nutritional intervention in patients undergoing surgery. J. Immunol. 2017;199:2171–2180.
    1. Gaudilliere B, et al. Clinical recovery from surgery correlates with single-cell immune signatures. Sci. Transl. Med. 2014;6:255ra131.
    1. Fragiadakis GK, et al. Patient-specific immune states before surgery are strong correlates of surgical recovery. Anesthesiology. 2015;123:1241–1255.
    1. Wallace DJ, Gavin IM, Karpenko O, Barkhordar F, Gillis BS. Cytokine and chemokine profiles in fibromyalgia, rheumatoid arthritis and systemic lupus erythematosus: a potentially useful tool in differential diagnosis. Rheumatol. Int. 2015;35:991–996.
    1. LaPaglia DM, et al. RNA-Seq investigations of human post-mortem trigeminal ganglia. Cephalalgia. 2018;38:912–932.
    1. Jacob M, Lopata AL, Dasouki M, Abdel Rahman AM. Metabolomics toward personalized medicine. Mass. Spectrom. Rev. 2017;38:221–238.
    1. Parker KS, et al. Urinary metabolomics identifies a molecular correlate of interstitial cystitis/bladder pain syndrome in a Multidisciplinary Approach to the Study of Chronic Pelvic Pain (MAPP) research network cohort. EBioMedicine. 2016;7:167–174.
    1. Chan DD, et al. In vivo articular cartilage deformation: noninvasive quantification of intratissue strain during joint contact in the human knee. Sci. Rep. 2016;6:19220.
    1. Lu G, Fei B. Medical hyperspectral imaging: a review. J. Biomed. Opt. 2014;19:10901.
    1. Marcu L, Boppart SA, Hutchinson MR, Popp J, Wilson BC. Biophotonics: the big picture. J. Biomed. Opt. 2017;23:1–7.
    1. Mackey S, Greely HT, Martucci K. Neuroimaging-based pain biomarkers: definitions, clinical and research applications, and evaluation frameworks to achieve personalized pain medicine. Pain. Rep. 2019;4:e762.
    1. van der Miesen MM, Lindquist MA, Wager TD. Neuroimaging-based biomarkers for pain: state of the field and current directions. Pain. Rep. 2019;4:e751.
    1. Vachon-Presseau E, et al. Corticolimbic anatomical characteristics predetermine risk for chronic pain. Brain. 2016;139:1958–1970.
    1. Fischer TZ, Waxman SG. Neuropathic pain in diabetes — evidence for a central mechanism. Nat. Rev. Neurol. 2010;6:462–466.
    1. Kuner R, Flor H. Structural plasticity and reorganisation in chronic pain. Nat. Rev. Neurosci. 2016;18:20–30.
    1. Reddan MC, Wager TD. Brain systems at the intersection of chronic pain and self-regulation. Neurosci. Lett. 2018;702:24–33.
    1. O’Muircheartaigh J, et al. Multivariate decoding of cerebral blood flow measures in a clinical model of on-going postsurgical pain. Hum. Brain Mapp. 2015;36:633–642.
    1. Marshall TM, et al. Activation of descending pain-facilitatory pathways from the rostral ventromedial medulla by cholecystokinin elicits release of prostaglandin-E2 in the spinal cord. Pain. 2012;153:86–94.
    1. Xie JY, et al. Cholecystokinin in the rostral ventromedial medulla mediates opioid-induced hyperalgesia and antinociceptive tolerance. J. Neurosci. 2005;25:409–416.
    1. Martucci KT, Weber KA, 2nd, Mackey SC. Altered cervical spinal cord resting-state activity in fibromyalgia. Arthritis Rheumatol. 2019;71:441–450.
    1. Weber KA, 2nd, et al. Thermal stimulation alters cervical spinal cord functional connectivity in humans. Neuroscience. 2018;369:40–50.
    1. Islam H, Law CSW, Weber KA, Mackey SC, Glover GH. Dynamic per slice shimming for simultaneous brain and spinal cord fMRI. Magn. Reson. Med. 2019;81:825–838.
    1. Davis KD, Moayedi M. Central mechanisms of pain revealed through functional and structural MRI. J. Neuroimmune Pharmacol. 2013;8:518–534.
    1. Geuter, S., et al. in Handbook of Psychophysiology (eds Cacioppo, J. T. et al.) 41–73 (Cambridge Univ. Press, 2017).
    1. Cherry SR. Fundamentals of positron emission tomography and applications in preclinical drug development. J. Clin. Pharmacol. 2001;41:482–491.
    1. Jones AKP, Watabe H, Cunningham VJ, Jones T. Cerebral decreases in opioid receptor binding in patients with central neuropathic pain measured by [11C] diprenorphine binding and PET. Eur. J. Pain. 2004;8:479–485.
    1. Zubieta JK, et al. Regional μ opioid receptor regulation of sensory and affective dimensions of pain. Science. 2001;293:311–315.
    1. Loggia ML, et al. Evidence for brain glial activation in chronic pain patients. Brain. 2015;138:604–615.
    1. Notter T, Coughlin JM, Sawa A, Meyer U. Reconceptualization of translocator protein as a biomarker of neuroinflammation in psychiatry. Mol. Psychiat. 2018;23:36–47.
    1. Gent YYJ, et al. Macrophage positron emission tomography imaging as a biomarker for preclinical rheumatoid arthritis: findings of a prospective pilot study. Arthritis Rheum. 2012;64:62–66.
    1. Brown JE, Chatterjee N, Younger J, Mackey S. Towards a physiology-based measure of pain: patterns of human brain activity distinguish painful from non-painful thermal stimulation. PLOS ONE. 2011;6:e24124.
    1. Marquand A, et al. Quantitative prediction of subjective pain intensity from whole-brain fMRI data using Gaussian processes. Neuroimage. 2010;49:2178–2189.
    1. López-Solà M, et al. Towards a neurophysiological signature for fibromyalgia. Pain. 2017;158:34–47.
    1. Mano H, et al. Classification and characterisation of brain network changes in chronic back pain: a multicenter study. Wellcome Open. Res. 2018;3:19.
    1. Mansour A, et al. Global disruption of degree rank order: a hallmark of chronic pain. Sci. Rep. 2016;6:34853.
    1. Cheng JC, et al. Multivariate machine learning distinguishes cross-network dynamic functional connectivity patterns in state and trait neuropathic pain. Pain. 2018;159:1764–1776.
    1. Nan J, et al. Whole-brain functional connectivity identification of functional dyspepsia. PLOS ONE. 2013;8:e65870.
    1. Callan D, Mills L, Nott C, England R, England S. A tool for classifying individuals with chronic back pain: using multivariate pattern analysis with functional magnetic resonance imaging data. PLOS ONE. 2014;9:e98007.
    1. Bagarinao E, et al. Preliminary structural MRI based brain classification of chronic pelvic pain: a MAPP network study. Pain. 2014;155:2502–2509.
    1. Ung H, et al. Multivariate classification of structural MRI data detects chronic low back pain. Cereb. Cortex. 2014;24:1037–1044.
    1. Baliki MN, et al. Corticostriatal functional connectivity predicts transition to chronic back pain. Nat. Neurosci. 2012;15:1117–1119.
    1. Kutch JJ, et al. Resting-state functional connectivity predicts longitudinal pain symptom change in urologic chronic pelvic pain syndrome: a MAPP network study. Pain. 2017;158:1069–1082.
    1. Hashmi JA, et al. Brain networks predicting placebo analgesia in a clinical trial for chronic back pain. Pain. 2012;153:2393–2402.
    1. Tetreault P, et al. Brain connectivity predicts placebo response across chronic pain clinical trials. PLOS Biol. 2016;14:e1002570.
    1. Bosma RL, et al. Brain dynamics and temporal summation of pain predicts neuropathic pain relief from ketamine infusion. Anesthesiology. 2018;129:1015–1024.
    1. Hung PS, Chen DQ, Davis KD, Zhong J, Hodaie M. Predicting pain relief: use of pre-surgical trigeminal nerve diffusion metrics in trigeminal neuralgia. Neuroimage Clin. 2017;15:710–718.
    1. Rosa MJ, Seymour B. Decoding the matrix: benefits and limitations of applying machine learning algorithms to pain neuroimaging. Pain. 2014;155:864–867.
    1. Davis KD. Is chronic pain a disease? Evaluating pain and nociception through self-report and neuroimaging. J. Pain. 2013;14:332–333.
    1. Hemington KS, Wu Q, Kucyi A, Inman RD, Davis KD. Abnormal cross-network functional connectivity in chronic pain and its association with clinical symptoms. Brain Struct. Funct. 2016;221:4203–4219.
    1. Marbach D, et al. Wisdom of crowds for robust gene network inference. Nat. Methods. 2012;9:796–804.
    1. Aghaeepour N, et al. Critical assessment of automated flow cytometry data analysis techniques. Nat. Methods. 2013;10:228–238.
    1. Aghaeepour N, et al. A benchmark for evaluation of algorithms for identification of cellular correlates of clinical outcomes. Cytometry A. 2016;89:16–21.
    1. Halilaj E, Hastie TJ, Gold GE, Delp SL. Physical activity is associated with changes in knee cartilage microstructure. Osteoarthr. Cartil. 2018;26:770–774.
    1. Tibshirani, R. & Friedman, J. A pliable lasso. Preprint at arXiv (2018).
    1. Choo J, Liu S. Visual analytics for explainable deep learning. IEEE Comput. Graph. Appl. 2018;38:84–92.
    1. Aghaeepour N, et al. GateFinder: projection-based gating strategy optimization for flow and mass cytometry. Bioinformatics. 2018;34:4131–4133.
    1. Taylor J, Tibshirani R. Post-selection inference for ℓ1-penalized likelihood models. Can. J. Stat. 2018;46:41–61.
    1. Cagney DN, et al. The FDA NIH Biomarkers, EndpointS, and other Tools (BEST) resource in neuro-oncology. Neuro Oncol. 2018;20:1162–1172.
    1. Dworkin RH, et al. Core outcome measures for chronic pain clinical trials: IMMPACT recommendations. Pain. 2005;113:9–19.
    1. Edwards RR, et al. Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. Pain. 2016;157:1851–1871.
    1. Bennett M. The LANSS pain scale: the Leeds Assessment of Neuropathic Symptoms and Signs. Pain. 2001;92:147–157.
    1. Bennett MI, et al. Using screening tools to identify neuropathic pain. Pain. 2007;127:199–203.
    1. Bouhassira D, et al. Neuropathic pain phenotyping as a predictor of treatment response in painful diabetic neuropathy: data from the randomized, double-blind, COMBO-DN study. Pain. 2014;155:2171–2179.
    1. Forstenpointner J, Rehm S, Gierthmuhlen J, Baron R. Stratification of neuropathic pain patients: the road to mechanism-based therapy? Curr. Opin. Anaesthesiol. 2018;31:562–568.
    1. Turk DC, et al. Identifying important outcome domains for chronic pain clinical trials: an IMMPACT survey of people with pain. Pain. 2008;137:276–285.
    1. Taylor AM, et al. Assessment of physical function and participation in chronic pain clinical trials: IMMPACT/OMERACT recommendations. Pain. 2016;157:1836–1850.
    1. Turk DC, Fillingim RB, Ohrbach R, Patel KV. Assessment of psychosocial and functional impact of chronic pain. J. Pain. 2016;17:T21–T49.
    1. Perrot S, Lanteri-Minet M. Patients’ global impression of change in the management of peripheral neuropathic pain: clinical relevance and correlations in daily practice. Eur. J. Pain. 2019;23:1117–1128.
    1. Jamison RN, Dorado K, Mei A, Edwards RR, Martel MO. Influence of opioid-related side effects on disability, mood, and opioid misuse risk among patients with chronic pain in primary care. Pain. Rep. 2017;2:e589.
    1. Lauria G, et al. European Federation of Neurological Societies/Peripheral Nerve Society guideline on the use of skin biopsy in the diagnosis of small fiber neuropathy. Report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society. Eur. J. Neurol. 2010;17:903–912.
    1. Devigili G, et al. The diagnostic criteria for small fibre neuropathy: from symptoms to neuropathology. Brain. 2008;131:1912–1925.
    1. Themistocleous AC, et al. The Pain in Neuropathy Study (PiNS): a cross-sectional observational study determining the somatosensory phenotype of painful and painless diabetic neuropathy. Pain. 2016;157:1132–1145.
    1. Zhou L, et al. Correlates of epidermal nerve fiber densities in HIV-associated distal sensory polyneuropathy. Neurology. 2007;68:2113–2119.
    1. von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron. 2012;73:638–652.
    1. Costigan M, Scholz J, Woolf CJ. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu. Rev. Neurosci. 2009;32:1–32.
    1. Zunhammer M, Bingel U, Wager TD, Placebo Imaging Consortium Placebo effects on the neurologic pain signature: a meta-analysis of individual participant functional magnetic resonance imaging data. JAMA Neurol. 2018;75:1321–1330.
    1. Campbell CM, et al. Randomized control trial of topical clonidine for treatment of painful diabetic neuropathy. Pain. 2012;153:1815–1823.
    1. Rowbotham MC, et al. Oral and cutaneous thermosensory profile of selective TRPV1 inhibition by ABT-102 in a randomized healthy volunteer trial. Pain. 2011;152:1192–1200.
    1. Serra J, et al. Effects of a T-type calcium channel blocker, ABT-639, on spontaneous activity in C-nociceptors in patients with painful diabetic neuropathy: a randomized controlled trial. Pain. 2015;156:2175–2183.
    1. Yarnitsky D, et al. Remote electrical neuromodulation (REN) relieves acute migraine: a randomized, double-blind, placebo-controlled, multicenter trial. Headache. 2019;59:1240–1252.
    1. US Food and Drug Administration. Statement by FDA Commissioner Scott Gottlieb, MD on the agency’s ongoing work to forcefully address the opioid crisis (FDA, 2018).
    1. Canadian Institutes of Health Research. Institute of Musculoskeletal Health and Arthritis IMHA Strategic Plan 2014–2018: enhancing musculoskeletal, skin and oral health. CIHR (2014).
    1. Heath Canada. Responding to Canada’s opioid crisis (Government of Canada, 2019).
    1. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use. E16 biomarkers related to drug or biotechnology product develoment: context, structure and format of quantification submissions. ICH (2010).
    1. European Medicines Agency. Essential considerations for successful qualification of novel methodologies (EMA, 2017).
    1. US Food and Drug Administration. Table of surrogate endpoints that were the basis of drug approval or licensure. FDA (2019).
    1. Innovative Medicines Initiative. Innovative Medicines Initiative IMI1 Final project report public summary: Europain. Understanding chronic pain and improving its treatment (IMI, 2015).
    1. Fitzgerald M, Walker SM. Infant pain management: a developmental neurobiological approach. Nat. Clin. Pract. Neurol. 2009;5:35–50.
    1. Goksan S, et al. fMRI reveals neural activity overlap between adult and infant pain. eLife. 2015;4:e06356.
    1. Hicks CL, von Baeyer CL, Spafford PA, van Korlaar I, Goodenough B. The faces pain scale — revised: toward a common metric in pediatric pain measurement. Pain. 2001;93:173–183.
    1. Zamzmi G, et al. A review of automated pain assessment in infants: features, classification tasks, and databases. IEEE Rev. Biomed. Eng. 2018;11:77–96.
    1. Boly M, et al. Perception of pain in the minimally conscious state with PET activation: an observational study. Lancet Neurol. 2008;7:1013–1020.
    1. Monti MM, et al. Willful modulation of brain activity in disorders of consciousness. N. Engl. J. Med. 2010;362:579–589.
    1. Cole LJ, et al. Pain sensitivity and fMRI pain-related brain activity in Alzheimer’s disease. Brain. 2006;129:2957–2965.
    1. de Knegt N, Scherder E. Pain in adults with intellectual disabilities. Pain. 2011;152:971–974.
    1. de Knegt NC, et al. Behavioral pain indicators in people with intellectual disabilities: a systematic review. J. Pain. 2013;14:885–896.
    1. Fanurik D, Koh JL, Dale Harrison R, Conrad TM, Tomerun C. Pain assessment in children with cognitive impairment. Clin.Nurs. Res. 1998;7:103–119.
    1. Wolff BB, Langley S. Cultural factors and the response to pain: a review. Am. Anthropol. 1968;70:494–501.
    1. Zborowski M. Cultural components in responses to pain. J. Soc. Issues. 1952;8:16–30.
    1. Anderson SR, Reynolds Losin EA. A sociocultural neuroscience approach to pain. Cult. Brain. 2017;5:14–35.
    1. Loeb S, et al. Overdiagnosis and overtreatment of prostate cancer. Eur. Urol. 2014;65:1046–1055.
    1. Cannon A, Kurklinsky S, Guthrie KJ, Riegert-Johnson DL. Advanced genetic testing comes to the pain clinic to make a diagnosis of paroxysmal extreme pain disorder. Case Rep. Neurol. Med. 2016;2016:9212369.
    1. Drenth JP, Waxman SG. Mutations in sodium-channel gene SCN9A cause a spectrum of human genetic pain disorders. J. Clin. Invest. 2007;117:3603–3609.
    1. Carey TS, Garrett JM. The relation of race to outcomes and the use of health care services for acute low back pain. Spine. 2003;28:390–394.
    1. Quartana PJ, Campbell CM, Edwards RR. Pain catastrophizing: a critical review. Expert Rev. Neurother. 2009;9:745–758.
    1. Clarke TK, et al. Low frequency genetic variants in the μ-opioid receptor (OPRM1) affect risk for addiction to heroin and cocaine. Neurosci. Lett. 2013;542:71–75.
    1. Petersen KK, Arendt-Nielsen L, Simonsen O, Wilder-Smith O, Laursen MB. Presurgical assessment of temporal summation of pain predicts the development of chronic postoperative pain 12 months after total knee replacement. Pain. 2015;156:55–61.
    1. Lauria G, et al. Intraepidermal nerve fiber density at the distal leg: a worldwide normative reference study. J. Peripher. Nerv. Syst. 2010;15:202–207.
    1. Freeman R, Baron R, Bouhassira D, Cabrera J, Emir B. Sensory profiles of patients with neuropathic pain based on the neuropathic pain symptoms and signs. Pain. 2014;155:367–376.
    1. Reimer M, et al. Prediction of response to tapentadol in chronic low back pain. Eur. J. Pain. 2017;21:322–333.
    1. Starkey Lewis PJ, et al. Circulating microRNAs as potential markers of human drug-induced liver injury. Hepatology. 2011;54:1767–1776.
    1. Serra J, et al. Microneurographic identification of spontaneous activity in C-nociceptors in neuropathic pain states in humans and rats. Pain. 2012;153:42–55.
    1. 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;120:2834–2846.
    1. Pascal MMV, et al. DOLORisk: study protocol for a multi-centre observational study to understand the risk factors and determinants of neuropathic pain. Wellcome Open. Res. 2019;3:63.
    1. Levitt J, Saab CY. What does a pain ‘biomarker’ mean and can a machine be taught to measure pain? Neurosci. Lett. 2019;702:40–43.
    1. Schulman J, Ramirez R, Zonenshayn M, Ribary U, Llinas RR. Thalamocortical dysrhythmia syndrome: MEG imaging of neuropathic pain. Thalamus Relat. Syst. 2005;31:33–39.
    1. Juottonen K, et al. Altered central sensorimotor processing in patients with complex regional pain syndrome. Pain. 2002;98:315–323.
    1. Kim JA, et al. Neuropathic pain and pain interference are linked to alpha-band slowing and reduced beta-band magnetoencephalography activity within the dynamic pain connectome in patients with multiple sclerosis. Pain. 2019;160:187–197.
    1. Scuteri D, et al. New trends in migraine pharmacology: targeting calcitonin gene-related peptide (CGRP) with monoclonal antibodies. Front. Pharmacol. 2019;10:363.
    1. Goadsby PJ, et al. Pathophysiology of migraine: a disorder of sensory processing. Physiol. Rev. 2017;97:553–622.
    1. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain. 2013;154:2310–2316.
    1. Vlckova-Moravcova E, Bednarik J, Dusek L, Toyka KV, Sommer C. Diagnostic validity of epidermal nerve fiber densities in painful sensory neuropathies. Muscle Nerve. 2008;37:50–60.
    1. Carragee EJ, Alamin TF, Miller JL, Carragee JM. Discographic, MRI and psychosocial determinants of low back pain disability and remission: a prospective study in subjects with benign persistent back pain. Spine J. 2005;5:24–35.
    1. Szabo N, et al. White matter microstructural alterations in migraine: a diffusion-weighted MRI study. Pain. 2012;153:651–656.
    1. Woodworth D, et al. Unique microstructural changes in the brain associated with urological chronic pelvic pain syndrome (UCPPS) revealed by diffusion tensor MRI, super-resolution track density imaging, and statistical parameter mapping: a MAPP network neuroimaging study. PLOS ONE. 2015;10:e0140250.
    1. Griebel AJ, Trippel SB, Emery NC, Neu CP. Noninvasive assessment of osteoarthritis severity in human explants by multicontrast MRI. Magn. Res. Med. 2014;71:807–814.
    1. Staikopoulos V, et al. Hyperspectral imaging of endogenous fluorescent metabolic molecules to identify pain states in central nervous system tissue. Proc. SPIE. 2016;10013:1001306.
    1. Aarnio M, et al. Visualization of painful inflammation in patients with pain after traumatic ankle sprain using [11C]-d-deprenyl PET/CT. Scand. J. Pain. 2017;17:418–424.
    1. Uceyler N, et al. Increased cortical activation upon painful stimulation in fibromyalgia syndrome. BMC Neurol. 2015;15:210.
    1. Vrana A, Meier ML, Hotz-Boendermaker S, Humphreys BK, Scholkmann F. Cortical sensorimotor processing of painful pressure in patients with chronic lower back pain — an optical neuroimaging study using fNIRS. Front. Hum. Neurosci. 2016;10:578.
    1. Demant DT, 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;155:2263–2273.
    1. Geuter S, Gamer M, Onat S, Büchel C. Parametric trial-by-trial prediction of pain by easily available physiological measures. Pain. 2014;155:994–1001.
    1. Kalliomaki J, et al. Evaluation of a novel chemokine receptor 2 (CCR2)-antagonist in painful diabetic polyneuropathy. Scand. J. Pain. 2013;4:77–83.
    1. Kalliomaki J, et al. A randomized, double-blind, placebo-controlled trial of a chemokine receptor 2 (CCR2) antagonist in posttraumatic neuralgia. Pain. 2013;154:761–767.
    1. Quiding H, et al. TRPV1 antagonistic analgesic effect: a randomized study of AZD1386 in pain after third molar extraction. Pain. 2013;154:808–812.
    1. Miller F, Bjornsson M, Svensson O, Karlsten R. Experiences with an adaptive design for a dose-finding study in patients with osteoarthritis. Contemp. Clin. Trials. 2014;37:189–199.
    1. Gimbel JS, et al. Long-term safety and effectiveness of tanezumab as treatment for chronic low back pain. Pain. 2014;155:1793–1801.
    1. Juhasz G, et al. Sumatriptan causes parallel decrease in plasma calcitonin gene-related peptide (CGRP) concentration and migraine headache during nitroglycerin induced migraine attack. Cephalalgia. 2005;25:179–183.
    1. Yarnitsky D, Granot M, Nahman-Averbuch H, Khamaisi M, Granovsky Y. Conditioned pain modulation predicts duloxetine efficacy in painful diabetic neuropathy. Pain. 2012;153:1193–1198.
    1. Yarnitsky D, et al. Nonpainful remote electrical stimulation alleviates episodic migraine pain. Neurology. 2017;88:1250–1255.
    1. Nahman-Averbuch H, et al. Waning of “conditioned pain modulation”: a novel expression of subtle pronociception in migraine. Headache. 2013;53:1104–1115.
    1. Yarnitsky D. Conditioned pain modulation (the diffuse noxious inhibitory control-like effect): its relevance for acute and chronic pain states. Curr. Opin. Anaesthesiol. 2010;23:611–615.
    1. Petropoulos IN, et al. Corneal confocal microscopy: ready for prime time. Clin. Exp. Optom. 2019;103:265–277.

Source: PubMed

3
订阅