Towards precision medicine for pain: diagnostic biomarkers and repurposed drugs

A B Niculescu, H Le-Niculescu, D F Levey, K Roseberry, K C Soe, J Rogers, F Khan, T Jones, S Judd, M A McCormick, A R Wessel, A Williams, S M Kurian, F A White, A B Niculescu, H Le-Niculescu, D F Levey, K Roseberry, K C Soe, J Rogers, F Khan, T Jones, S Judd, M A McCormick, A R Wessel, A Williams, S M Kurian, F A White

Abstract

We endeavored to identify objective blood biomarkers for pain, a subjective sensation with a biological basis, using a stepwise discovery, prioritization, validation, and testing in independent cohorts design. We studied psychiatric patients, a high risk group for co-morbid pain disorders and increased perception of pain. For discovery, we used a powerful within-subject longitudinal design. We were successful in identifying blood gene expression biomarkers that were predictive of pain state, and of future emergency department (ED) visits for pain, more so when personalized by gender and diagnosis. MFAP3, which had no prior evidence in the literature for involvement in pain, had the most robust empirical evidence from our discovery and validation steps, and was a strong predictor for pain in the independent cohorts, particularly in females and males with PTSD. Other biomarkers with best overall convergent functional evidence for involvement in pain were GNG7, CNTN1, LY9, CCDC144B, and GBP1. Some of the individual biomarkers identified are targets of existing drugs. Moreover, the biomarker gene expression signatures were used for bioinformatic drug repurposing analyses, yielding leads for possible new drug candidates such as SC-560 (an NSAID), and amoxapine (an antidepressant), as well as natural compounds such as pyridoxine (vitamin B6), cyanocobalamin (vitamin B12), and apigenin (a plant flavonoid). Our work may help mitigate the diagnostic and treatment dilemmas that have contributed to the current opioid epidemic.

Conflict of interest statement

ABN is listed as inventor on a patent application being filed by Indiana University, and is a co-founder of MindX Sciences. The other authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
Steps 1–3: Discovery, prioritization, and validation. a Cohorts used in study, depicting flow of discovery, prioritization, and validation of biomarkers from each step. b Discovery cohort longitudinal within-subject analysis. Phchp### is study ID for each subject. V# denotes visit number. c Discovery of possible subtypes of Pain based on High Pain visits in the discovery cohort. Subjects were clustered using measures of mood and anxiety (Simplified Affective State Scale (SASS)), as well as psychosis (PANNS Positive). d Differential gene expression in the Discovery cohort—number of genes identified with differential expression (DE) and absent-present (AP) methods with an internal score of 2 and above. Red—increased in expression in High Pain, blue—decreased in expression in High Pain. At the discovery step probesets are identified based on their score for tracking pain with a maximum of internal points of 6 (33% (2 pt), 50% (4 pt), and 80% (6 pt)). e Prioritization with CFG for prior evidence of involvement in pain. In the prioritization step, probesets are converted to their associated genes using Affymetrix annotation and GeneCards. Genes are prioritized and scored using CFG for pain evidence with a maximum of 12 external points. Genes scoring at least six points out of a maximum possible of 18 total internal and external scores points are carried to the validation step. f Validation in an independent cohort of psychiatric patients with co-morbid pain disorders and severe subjective and functional pain ratings. In the validation step biomarkers are assessed for stepwise change from the discovery groups of subjects with Low Pain, to High Pain, to Clinically Severe Pain disorder, using ANOVA. N = number of testing visits. Five biomarkers were nominally significant, MFAP3 and PIK3CD were the most significant, and 68 biomarkers were stepwise changed
Fig. 2
Fig. 2
Best single biomarkers predictors. From the long list (n = 65). Those on short list (n = 5) are bolded. Bar graph shows best predictive biomarkers in each group. *Nominally significant p < 0.05. **Bonferroni significant for the 65 biomarkers tested. Table underneath the figures displays the actual number of biomarkers for each group whose ROC AUC p-values (a, b) and Cox odds ratio p-values (c) are at least nominally significant. Some female diagnostic group are missing from the graph as they did not have any significant biomarkers. Cross-sectional is based on levels at one visit. Longitudinal is based on levels at multiple visits (integrates levels at most recent visit, maximum levels, slope into most recent visit, and maximum slope). Dividing lines represent the cutoffs for a test performing at chance levels (white), and at the same level as the best biomarkers for all subjects in cross-sectional (gray) and longitudinal (black) based predictions. All biomarkers perform better than chance. Biomarkers performed better when personalized by gender and diagnosis
Fig. 3
Fig. 3
Biological roles. STRING interaction network for the top biomarkers for pain (65 probesets, 60 genes)

References

    1. Woolf CJ. What is this thing called pain? J Clin Invest. 2010;120:3742–4. doi: 10.1172/JCI45178.
    1. Baron R, Maier C, Attal N, Binder A, Bouhassira D, Cruccu G, et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain. 2017;158:261–72. doi: 10.1097/j.pain.0000000000000753.
    1. Fond G, Boyer L, Andrianarisoa M, Godin O, Bulzacka E, Berna F, et al. Self-reported pain in patients with schizophrenia. Results from the national first-step FACE-SZ cohort. Prog Neuropsychopharmacol Biol Psychiatry. 2018;85:62–8. doi: 10.1016/j.pnpbp.2018.04.007.
    1. Costigan M, Belfer I, Griffin RS, Dai F, Barrett LB, Coppola G, et al. Multiple chronic pain states are associated with a common amino acid-changing allele in KCNS1. Brain: J Neurol. 2010;133:2519–27. doi: 10.1093/brain/awq195.
    1. Rodieux F, Piguet V, Berney P, Desmeules J, Besson M. Pharmacogenetics and analgesic effects of antidepressants in chronic pain management. Pers Med. 2015;12:163–75. doi: 10.2217/pme.14.61.
    1. Le-Niculescu H, Levey DF, Ayalew M, Palmer L, Gavrin LM, Jain N, et al. Discovery and validation of blood biomarkers for suicidality. Mol Psychiatry. 2013;18:1249–64. doi: 10.1038/mp.2013.95.
    1. Niculescu AB, Levey DF, Phalen PL, Le-Niculescu H, Dainton HD, Jain N, et al. Understanding and predicting suicidality using a combined genomic and clinical risk assessment approach. Mol Psychiatry. 2015;20:1266–85. doi: 10.1038/mp.2015.112.
    1. Levey DF, Niculescu EM, Le-Niculescu H, Dainton HL, Phalen PL, Ladd TB, et al. Towards understanding and predicting suicidality in women: biomarkers and clinical risk assessment. Mol Psychiatry. 2016;21:768–85. doi: 10.1038/mp.2016.31.
    1. Niculescu AB, Le-Niculescu H, Levey DF, Phalen PL, Dainton HL, Roseberry K, et al. Precision medicine for suicidality: from universality to subtypes and personalization. Mol Psychiatry. 2017;22:1250–73. doi: 10.1038/mp.2017.128.
    1. Niculescu AB, Levey D, Le-Niculescu H, Niculescu E, Kurian SM, Salomon D. Psychiatric blood biomarkers: avoiding jumping to premature negative or positive conclusions. Mol Psychiatry. 2015;20:286–8. doi: 10.1038/mp.2014.180.
    1. Niculescu AB, Le-Niculescu H. Dissecting suicidality using a combined genomic and clinical approach. Neuropsychopharmacol: Off Publ Am Coll Neuropsychopharmacol. 2017;42:360. doi: 10.1038/npp.2016.228.
    1. Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, Chen R, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–307. doi: 10.1016/j.cell.2012.02.009.
    1. Niculescu AB, Le-Niculescu H. Convergent functional genomics: what we have learned and can learn about genes, pathways, and mechanisms. Neuropsychopharmacol: Off Publ Am Coll Neuropsychopharmacol. 2010;35:355–6. doi: 10.1038/npp.2009.107.
    1. Gandal MJ, Haney JR, Parikshak NN, Leppa V, Ramaswami G, Hartl C, et al. Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap. Science. 2018;359:693–7. doi: 10.1126/science.aad6469.
    1. Petrosky Emiko, Harpaz Rafael, Fowler Katherine A., Bohm Michele K., Helmick Charles G., Yuan Keming, Betz Carter J. Chronic Pain Among Suicide Decedents, 2003 to 2014: Findings From the National Violent Death Reporting System. Annals of Internal Medicine. 2018;169(7):448. doi: 10.7326/M18-0830.
    1. Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, Miriami E, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–307. doi: 10.1016/j.cell.2012.02.009.
    1. Piening BD, Zhou W, Contrepois K, Rost H, Gu Urban GJ, Mishra T, et al. Integrative personal omics profiles during periods of weight gain and loss. Cell Syst. 2018;6:157–70 e158. doi: 10.1016/j.cels.2017.12.013.
    1. Karczewski KJ, Snyder MP. Integrative omics for health and disease. Nat Rev Genet. 2018;19:299–310. doi: 10.1038/nrg.2018.4.
    1. Lillie EO, Patay B, Diamant J, Issell B, Topol EJ, Schork NJ. The n-of-1 clinical trial: the ultimate strategy for individualizing medicine? Pers Med. 2011;8:161–73. doi: 10.2217/pme.11.7.
    1. Schork NJ. Personalized medicine: Time for one-person trials. Nature. 2015;520:609–11. doi: 10.1038/520609a.
    1. Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, Jagadish HV, et al. The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain: J Neurol. 2011;134(Pt 11):3222–35. doi: 10.1093/brain/awr228.
    1. Gruber HE, Hoelscher GL, Ingram JA, Hanley EN., Jr Genome-wide analysis of pain-, nerve- and neurotrophin -related gene expression in the degenerating human annulus. Mol Pain. 2012;8:63. doi: 10.1186/1744-8069-8-63.
    1. Bell RL, Kimpel MW, McClintick JN, Strother WN, Carr LG, Liang T, et al. Gene expression changes in the nucleus accumbens of alcohol-preferring rats following chronic ethanol consumption. Pharmacol Biochem Behav. 2009;94:131–47. doi: 10.1016/j.pbb.2009.07.019.
    1. Le-Niculescu H, Balaraman Y, Patel S, Tan J, Sidhu K, Jerome RE, et al. Towards understanding the schizophrenia code: an expanded convergent functional genomics approach. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:129–58. doi: 10.1002/ajmg.b.30481.
    1. Le-Niculescu H, McFarland MJ, Ogden CA, Balaraman Y, Patel S, Tan J, et al. Phenomic, convergent functional genomic, and biomarker studies in a stress-reactive genetic animal model of bipolar disorder and co-morbid alcoholism. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:134–66. doi: 10.1002/ajmg.b.30707.
    1. Liu CJ, Dib-Hajj SD, Black JA, Greenwood J, Lian Z, Waxman SG. Direct interaction with contactin targets voltage-gated sodium channel Na(v)1.9/NaN to the cell membrane. J Biol Chem. 2001;276:46553–61. doi: 10.1074/jbc.M108699200.
    1. Olausson P, Ghafouri B, Backryd E, Gerdle B. Clear differences in cerebrospinal fluid proteome between women with chronic widespread pain and healthy women - a multivariate explorative cross-sectional study. J Pain Res. 2017;10:575–90. doi: 10.2147/JPR.S125667.
    1. Miura Y, Devaux JJ, Fukami Y, Manso C, Belghazi M, Wong AH, et al. Contactin 1 IgG4 associates to chronic inflammatory demyelinating polyneuropathy with sensory ataxia. Brain: a J Neurol. 2015;138(Pt 6):1484–91. doi: 10.1093/brain/awv054.
    1. Focking M, Lopez LM, English JA, Dicker P, Wolff A, Brindley E, et al. Proteomic and genomic evidence implicates the postsynaptic density in schizophrenia. Mol Psychiatry. 2015;20:424–32. doi: 10.1038/mp.2014.63.
    1. Vawter MP, Ferran E, Galke B, Cooper K, Bunney WE, Byerley W. Microarray screening of lymphocyte gene expression differences in a multiplex schizophrenia pedigree. Schizophr Res. 2004;67:41–52. doi: 10.1016/S0920-9964(03)00151-8.
    1. Cao-Lei L, Massart R, Suderman MJ, Machnes Z, Elgbeili G, Laplante DP, et al. DNA methylation signatures triggered by prenatal maternal stress exposure to a natural disaster: Project Ice Storm. PLoS ONE. 2014;9:e107653. doi: 10.1371/journal.pone.0107653.
    1. Hammamieh R, Chakraborty N, Gautam A, Miller SA, Muhie S, Meyerhoff J, et al. Transcriptomic analysis of the effects of a fish oil enriched diet on murine brains. PLoS ONE. 2014;9:e90425. doi: 10.1371/journal.pone.0090425.
    1. Parisien M, Khoury S, Chabot-Dore AJ, Sotocinal SG, Slade GD, Smith SB, et al. Effect of human genetic variability on gene expression in dorsal root ganglia and association with pain phenotypes. Cell Rep. 2017;19:1940–52. doi: 10.1016/j.celrep.2017.05.018.
    1. Descalzi G, Mitsi V, Purushothaman I, Gaspari S, Avrampou K, Loh YE, et al. Neuropathic pain promotes adaptive changes in gene expression in brain networks involved in stress and depression. Sci Signal. 2017;10.
    1. Smith SB, Maixner DW, Fillingim RB, Slade G, Gracely RH, Ambrose K, et al. Large candidate gene association study reveals genetic risk factors and therapeutic targets for fibromyalgia. Arthritis Rheum. 2012;64:584–93. doi: 10.1002/art.33338.
    1. Hoyo-Becerra C, Huebener A, Trippler M, Lutterbeck M, Liu ZJ, Truebner K, et al. Concomitant interferon alpha stimulation and TLR3 activation induces neuronal expression of depression-related genes that are elevated in the brain of suicidal persons. PLoS ONE. 2013;8:e83149. doi: 10.1371/journal.pone.0083149.
    1. Chang X, Liu Y, Hahn CG, Gur RE, Sleiman PMA, Hakonarson H. RNA-seq analysis of amygdala tissue reveals characteristic expression profiles in schizophrenia. Transl Psychiatry. 2017;7:e1203. doi: 10.1038/tp.2017.154.
    1. Saetre P, Emilsson L, Axelsson E, Kreuger J, Lindholm E, Jazin E. Inflammation-related genes up-regulated in schizophrenia brains. BMC Psychiatry. 2007;7:46. doi: 10.1186/1471-244X-7-46.
    1. Breen MS, Maihofer AX, Glatt SJ, Tylee DS, Chandler SD, Tsuang MT, et al. Gene networks specific for innate immunity define post-traumatic stress disorder. Mol Psychiatry. 2015;20:1538–45. doi: 10.1038/mp.2015.9.
    1. Le-Niculescu H, Case NJ, Hulvershorn L, Patel SD, Bowker D, Gupta J, et al. Convergent functional genomic studies of omega-3 fatty acids in stress reactivity, bipolar disorder and alcoholism. Transl Psychiatry. 2011;1:e4. doi: 10.1038/tp.2011.1.
    1. Liu J, Lewohl JM, Harris RA, Iyer VR, Dodd PR, Randall PK, et al. Patterns of gene expression in the frontal cortex discriminate alcoholic from nonalcoholic individuals. Neuropsychopharmacology. 2006;31:1574–82. doi: 10.1038/sj.npp.1300947.
    1. Andrus BM, Blizinsky K, Vedell PT, Dennis K, Shukla PK, Schaffer DJ, et al. Gene expression patterns in the hippocampus and amygdala of endogenous depression and chronic stress models. Mol Psychiatry. 2012;17:49–61. doi: 10.1038/mp.2010.119.
    1. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–35. doi: 10.1126/science.1132939.
    1. Lamb J. The Connectivity Map: a new tool for biomedical research. Nat Rev Cancer. 2007;7:54–60. doi: 10.1038/nrc2044.

Source: PubMed

3
Suscribir