288
Views
5
CrossRef citations to date
0
Altmetric
Review

A Review of DNA Risk Alleles to Determine Epigenetic Repair of mRNA Expression to Prove Therapeutic Effectiveness in Reward Deficiency Syndrome (RDS): Embracing “Precision Behavioral Management”

ORCID Icon, , , , ORCID Icon, ORCID Icon, , , , ORCID Icon, ORCID Icon & show all
Pages 2115-2134 | Published online: 17 Dec 2021

References

  • SAMHSA. Available from: SAMHSA.gov/data/report/results-2013-national-survey-drug-use-and-health-detailed-tables. Accessed November 24, 2021
  • National Institutes on Drug Abuse, Substance Abuse and Mental Health Services Administration. National survey on drug use and health, 2014–2020. Available from: SAMHSA.gov/data/data-we-collect/NSDUH-National-Survey-drug-use-and-health. Accessed November 24, 2021.
  • Hingson R, Heeren T, Winter M, et al. Magnitude of alcohol-related mortality and morbidity among U.S. college students ages 18–24: changes from 1998–2001. Annu Rev Public Health. 2005;26:2590279. PMID: 15760289. doi:10.1146/annurev.publhealth.26.021304.144652
  • National Institutes of Health, National Institute of Drug Abuse. Monitoring the future national survey results on drug use, 1975–2019, volume 1: secondary school students; 2019.
  • Volkow ND. Pandemic worsens opioid crisis: NIDA director; July 2020. Available from: https://drugfree.org/drug-and-alcohol-news/pandemic-worsens-opioid-crisis-NIDA-Director/. Accessed November 24, 2021.
  • Davenport S, Weaver A, Caverly M. Economic impact of non-medical opioid use in the United States: annual estimates and projections for 2015 through 2019. Society of Actuaries. Schaumberg, Illinois; 2019. Available from: www.SOA.org. Accessed November 24, 2021.
  • Abijo T, Blum K, Gondré-Lewis MC. Neuropharmacological and neurogenetic correlates of Opioid Use Disorder (OUD) as a function of ethnicity: relevance to precision addiction medicine. Curr Neuropharmacol. 2020;18(7):578–595. doi:10.2174/1570159X17666191118125702
  • Wu X, Hudmon K, Detry M, et al. D2 dopamine receptor gene polymorphisms among African-Americans and Mexican-Americans: a lung cancer case-control study. Cancer Epidemiol Biomarkers Prev. 2000;9(10):1021–1026.
  • Blum K, Chen ALC, Thanos PK, et al. Genetic addiction risk score (GARS) ™, a predictor of vulnerability to opioid dependence. Front Biosci. 2018;10:175–196. doi:10.2741/e816
  • Blum K, Noble EP, Sheridan PJ, et al. Allelic association of human dopamine D2 receptor gene in alcoholism. JAMA. 1990;263(15):2055–2060. doi:10.1001/jama.1990.03440150063027
  • Noble EP, Blum K, Khalsa ME, et al. Allelic association of the D2 dopamine receptor gene with cocaine dependence [published correction appears in drug alcohol depend 1993 Dec;34(1):83–4]. Drug Alcohol Depend. 1993;33(3):271–285. doi:10.1016/0376-8716(93)90113-5
  • Camplain R, Camplain C, Trotter R, et al. Racial/ethnic differences in drug and alcohol-related arrest outcomes in a Southwest county from 2009 to 2018. Am J Public Health. 2020;110(S1):S85–S92. doi:10.2105/AJPH.2019.305409
  • Green KM, Doherty EE, Ensminger ME. What’s drugs got to do with it? Addict Beh. 2020;110:106539. doi:10.1016/j.addbeh.2020.106539
  • Downs BW, Blum K, Baron D, et al. Death by opioids: are there non-addictive scientific solutions? J Syst Integr Neurosci. 2019;5. doi:10.15761/JSIN.1000211
  • Blum K, Gondré-Lewis MC, Modestino EJ, et al. Understanding the scientific basis of Post-traumatic Stress Disorder (PTSD): precision behavioral management overrides stigmatization. Mol Neurobiol. 2019;56(11):7836–7850. doi:10.1007/s12035-019-1600-8
  • Blum K, Modestino EJ, Neary J, et al. Promoting Precision Addiction Management (PAM) to combat the global opioid crisis. Biomed J Sci Tech Res. 2018;2(2):1–4. doi:10.26717/BJSTR.2018.02.000738
  • Blum K, Gold M, Modestino EJ, et al. Would induction of dopamine homeostasis via coupling genetic addiction risk score (GARS®) and pro-dopamine regulation benefit benzodiazepine use disorder (BUD)? J Syst Integr Neurosci. 2018;4. doi:10.15761/JSIN.1000196
  • Blum K, Gondré-Lewis MC, Baron D, et al. Introducing precision addiction management of reward deficiency syndrome, the construct that underpins all addictive behaviors. Front Psychiatry. 2018;9:548. doi:10.3389/fpsyt.2018.00548
  • Blum K, Modestino EJ, Lott L, et al. Introducing “Precision Addiction Management (PAM®)” as an adjunctive genetic guided therapy for abusable drugs in America. Open Access J Behav Sci Psychol. 2018;1(2):1–4.
  • Blum K, Modestino EJ, Gondré-Lewis MC, et al. Global opioid epidemic: doomed to fail without genetically based precision addiction medicine (PAM™): lessons learned from America. Precis Med. 2017;2(1):17–22.
  • Blum K, Bowirrat A, Baron D, et al. Biotechnical development of genetic addiction risk score (GARS) and selective evidence for inclusion of polymorphic allelic risk in substance use disorder (SUD). J Syst Integr Neurosci. 2019;6. doi:10.15761/JSIN.1000221
  • Fried L, Modestino EJ, Siwicki D, et al. Hypodopaminergia and “Precision Behavioral Management” (PBM): it is a generational family affair. Curr Pharm Biotechnol. 2020;21(6):528–541. doi:10.2174/1389201021666191210112108
  • Kotyuk E, Magi A, Eisinger A, et al. Co-occurrences of substance use and other potentially addictive behaviors: epidemiological results from the psychological and genetic factors of the addictive behaviors (PGA) study. J Behav Addict. 2020;9(2):272–288. doi:10.1556/2006.2020.00033
  • Blum K, Han D, Modestino EJ, et al. A systematic, intensive statistical investigation of data from the comprehensive analysis of reported drugs (CARD) for compliance and illicit opioid abstinence in substance addiction treatment with buprenorphine/naloxone. Subst Use Misuse. 2018;53(2):220–229. doi:10.1080/10826084.2017.1400064
  • Blum K, Han D, Femino J, et al. Systematic evaluation of “compliance” to prescribed treatment medications and “abstinence” from psychoactive drug abuse in chemical dependence programs: data from the comprehensive analysis of reported drugs. PLoS One. 2014;9(9):e104275. doi:10.1371/journal.pone.0104275
  • Miller M, Chen AL, Stokes SD, et al. Early intervention of intravenous KB220IV–neuroadaptagen amino-acid therapy (NAAT) improves behavioral outcomes in a residential addiction treatment program: a pilot study. J Psychoactive Drugs. 2012;44(5):398–409. doi:10.1080/02791072.2012.737727
  • Febo M, Blum K, Badgaiyan RD, et al. Dopamine homeostasis: brain functional connectivity in reward deficiency syndrome. Front Biosci. 2017;22:669–691. doi:10.2741/4509
  • Nikoshkov A, Drakenberg K, Wang X, et al. Opioid neuropeptide genotypes in relation to heroin abuse: dopamine tone contributes to reversed mesolimbic proenkephalin expression. Proc Natl Acad Sci U S A. 2008;105(2):786–791. doi:10.1073/pnas.0710902105
  • Blum K, Hauser M, Fratantonio J, Badgaiyan RD. Molecular genetic testing in pain and addiction: facts, fiction and clinical utility. Addict Genet. 2015;2(1):1–5. doi:10.1515/addge-2015-0001
  • Chen AL, Chen TJ, Waite RL, et al. Hypothesizing that brain reward circuitry genes are genetic antecedents of pain sensitivity and critical diagnostic and pharmacogenomic treatment targets for chronic pain conditions. Med Hypotheses. 2009;72(1):14–22. doi:10.1016/j.mehy.2008.07.059
  • Jung Y, Montel RA, Shen PH, et al. Assessment of the association of D2 dopamine receptor gene and reported allele frequencies with alcohol use disorders: a systematic review and meta-analysis. JAMA Netw Open. 2019;2(11):e1914940. doi:10.1001/jamanetworkopen.2019.14940
  • Blum K, Chen AL, Oscar-Berman M, et al. Generational association studies of dopaminergic genes in reward deficiency syndrome (RDS) subjects: Selecting appropriate phenotypes for reward dependence behaviors. Int J Environ Res Public Health. 2011;8(12):4425–4459. doi:10.3390/ijerph8124425o
  • Blum K, Baron D, Lott L, et al. In search of reward deficiency syndrome (RDS)-free controls: the “holy grail” in genetic addiction risk testing. Curr Psychopharmacol. 2020;9(1):7–21. doi:10.2174/2211556008666191111103152
  • Chen TJ, Blum K, Mathews D, et al. Are dopaminergic genes involved in a predisposition to pathological aggression? Hypothesizing the importance of “super normal controls” in psychiatricgenetic research of complex behavioral disorders. Med Hypotheses. 2005;65(4):703–707. doi:10.1016/j.mehy.2005.04.037
  • Barr CL, Kidd KK. Population frequencies of the A1 allele at the dopamine D2 receptor locus. Biol Psychiatry. 1993;34(4):204–209. doi:10.1016/0006-3223(93)90073-
  • Hill SY, Zezza N, Wipprecht G, et al. Linkage studies of D2 and D4 receptor genes and alcoholism. Am J Med Genet. 1999;88(6):676–685. doi:10.1002/(sici)1096-8628(19991215)88:6<676::aid-ajmg18>3.0.co;2-s
  • Ponce G, Jimenez-Arriero MA, Rubio G, et al. The A1 allele of the DRD2 gene (TaqI A polymorphisms) is associated with antisocial personality in a sample of alcohol-dependent patients. Eur Psychiatry. 2003;18(7):356–360. doi:10.1016/j.eurpsy.2003.06.006
  • Blum K, Oscar-Berman M, Badgaiyan RD, et al. Hypothesizing dopaminergic genetic antecedents in schizophrenia and substance seeking behavior. Med Hypotheses. 2014;82(5):606–614. doi:10.1016/j.mehy.2014.02.019
  • Green AI, Zimmet SV, Strous RD, et al. Clozapine for comorbid substance use disorder and schizophrenia: Do patients with schizophrenia have a reward-deficiency syndrome that can be ameliorated by clozapine? Harv Rev Psychiatry. 1999;6(6):287–296. doi:10.3109/10673229909017206
  • van Ree JM, De Wied D. Neuroleptic-like profile of γ-type endorphins as related to schizophrenia. Trends Pharmacol Sci. 1982;3:358–361. doi:10.1016/0165-6147(82)91183-X
  • Gondré-Lewis MC, Bassey R, Blum K. Pre-clinical models of reward deficiency syndrome: a behavioral octopus. Neurosci Biobehav Rev. 2020;115:164–188. PMID: 32360413; PMCID: PMC7594013. doi:10.1016/j.neubiorev.2020.04.021
  • Li CY, Mao X, Wei L. Genes and (common) pathways underlying drug addiction. PLoS Comput Biol. 2008;4(1):e2. doi:10.1371/journal.pcbi.0040002
  • Dang LC, Samanez-Larkin GR, Castrellon JJ, et al. Associations between dopamine D2 receptor availability and BMI depend on age. Neuroimage. 2016;138:176–183. doi:10.1016/j.neuroimage.2016.05.044
  • Fetsko LA, Xu R, Wang Y. Effects of age and dopamine D2L receptor-deficiency on motor and learning functions. Neurobiol Aging. 2005;26(4):521–530. doi:10.1016/j.neurobiolaging.2004.04.005
  • Karrer TM, Josef AK, Mata R, et al. Reduced dopamine receptors and transporters but not synthesis capacity in normal aging adults: A meta-analysis. Neurobiol Aging. 2017;57:36–46. doi:10.1016/j.neurobiolaging.2017.05.006
  • Volkow ND, Wang GJ, Fowler JS, et al. Measuring age-related changes in dopamine D2 receptors with 11C-raclopride and 18F-N-methylspiroperidol. Psychiatry Res. 1996;67(1):11–16. doi:10.1016/0925-4927(96)02809-0
  • Cadet JL. Epigenetics of stress, addiction, and resilience: therapeutic implications. Mol Neurobiol. 2016;53(1):545–560. doi:10.1007/s12035-014-9040-y
  • Kreek MJ, Levran O, Reed B, et al. Opiate addiction and cocaine addiction: Underlying molecular neurobiology and genetics. J Clin Invest. 2012;122(10):3387–3393. doi:10.1172/JCI60390
  • Wang SC, Chen YC, Lee CH, et al. Opioid addiction, genetic susceptibility, and medical treatments: a review. Int J Mol Sci. 2019;20(17):4294. doi:10.3390/ijms20174294
  • Le Foll B, Gallo A, Le Strat Y, et al. Genetics of dopamine receptors and drug addiction: A comprehensive review. Behav Pharmacol. 2009;20(1):1–17. doi:10.1097/FBP.0b013e3283242f05
  • De Sa Nogueira D, Merienne K, Befort K. Neuroepigenetics and addictive behaviors: where do we stand? Neurosci Biobehav Rev. 2019;106:58–72. doi:10.1016/j.neubiorev.2018.08.018
  • Duncan JR. Current perspectives on the neurobiology of drug addiction: A focus on genetics and factors regulating gene expression. ISRN Neurol. 2012;2012:972607. doi:10.5402/2012/972607
  • Onaivi ES, Ali SF, Chirwa SS, et al. Ibogaine signals addiction genes and methamphetamine alteration of long-term potentiation. Ann N Y Acad Sci. 2002;965:28–46. doi:10.1111/j.1749-6632.2002.tb04149.x
  • Li ZH, Liu YF, Li KN, et al. Analysis of functional and pathway association of differential co-expressed genes: A case study in drug addiction. J Biomed Inform. 2012;45(1):30–36. doi:10.1016/j.jbi.2011.08.014
  • Blum K, Madigan MA, Fried L, et al. Coupling Genetic Addiction Risk Score (GARS) and pro dopamine regulation (KB220) to combat substance use disorder (SUD). Glob J Addict Rehabilit Med. 2017;1(2):25–34. doi:10.19080/GJARM.2017.01.55556
  • Krebs EE, Gravely A, Nugent S, et al. Effect of opioid vs nonopioid medications on pain-related function in patients with chronic back pain or hip or knee osteoarthritis pain: The SPACE randomized clinical trial. JAMA. 2018;319(9):872–882. PMID: 29509867; PMCID: PMC5885909. doi:10.1001/jama.2018.0899
  • Blum K, Oscar-Berman M, Blum SH, et al. Can genetic testing coupled with enhanced dopaminergic activation reduce recidivism rates in the workers compensation legacy cases? J Alcohol Drug Depend. 2014;2(3):161. doi:10.4172/2329-6488.1000161
  • Blum K, Jacobs W, Modestino EJ, et al. Insurance companies fighting the peer review empire without any validity: The case for addiction and pain Modalities in the face of an American drug epidemic. SEJ Surg Pain. 2018;1(1):1–11.
  • Blum K, Oscar-Berman M, Dinubile N, et al. Coupling Genetic Addiction Risk Score (GARS) with electrotherapy: fighting iatrogenic opioid dependence. J Addict Res Ther. 2013;4(163):1000163. doi:10.4172/2155-6105.1000163
  • Chen TJ, Blum K, Payte JT, et al. Narcotic antagonists in drug dependence: Pilot study showing enhancement of compliance with SYN-10, amino-acid precursors and enkephalinase inhibition therapy. Med Hypotheses. 2004;63(3):538–548. doi:10.1016/j.mehy.2004.02.051
  • Blum K, Modestino EJ, Badgaiyan RD, et al. Analysis of evidence for the combination of pro-dopamine regulator (KB220PAM) and naltrexone to prevent opioid use disorder relapse. EC Psychol Psychiatr. 2018;7(8):564–579.
  • Hill E, Han D, Dumouchel P, et al. Long term Suboxone™ emotional reactivity as measured by automatic detection in speech [published correction appears in PLoS One. 2013;8(8):doi/10.1371/annotation/be0b3a26-c1bc-4d92-98c1-c516acc8dcf2.Barh, Debmalya [added]]. PLoS One. 2013;8(7):e69043. doi:10.1371/journal.pone.0069043
  • Foley KF, DeSanty KP, Kast RE. Bupropion: pharmacology and therapeutic applications. Expert Rev Neurother. 2006;6(9):1249–1265. doi:10.1586/14737175.6.9.1249
  • Cano-Cebrián MJ, Zornoza-Sabina T, Guerri C, et al. Local Acamprosate modulates dopamine release in the rat nucleus accumbens through NMDA receptors: an in vivo microdialysis study. Naunyn Schmiedebergs Arch Pharmacol. 2003;367(2):119–125. doi:10.1007/s00210-002-0674-3
  • Quintero GC. Role of nucleus accumbens glutamatergic plasticity in drug addiction. Neuropsychiatr Dis Treat. 2013;9:1499–1512. doi:10.2147/NDT.S45963
  • Gueorguieva R, Wu R, Donovan D, et al. Baseline trajectories of drinking moderate Acamprosate and naltrexone effects in the COMBINE study. Alcohol Clin Exp Res. 2011;35(3):523–531. doi:10.1111/j.1530-0277.2010.01369.x
  • Blum K, Chen TJ, Meshkin B, et al. Manipulation of catechol-O-methyl-transferase (COMT) activity to influence the attenuation of substance seeking behavior, a subtype of Reward Deficiency Syndrome (RDS), is dependent upon gene polymorphisms: a hypothesis. Med Hypotheses. 2007;69(5):1054–1060. doi:10.1016/j.mehy.2006.12.062
  • Karkhanis AN, Rose JH, Weiner JL, et al. Early-life social isolation stress increases kappa opioid receptor responsiveness and downregulates the dopamine system. Neuropsychopharmacology. 2016;41(9):2263–2274. doi:10.1038/npp.2016.21
  • Blum K, Briggs AH, Elston SF, et al. Reduced leucine-enkephalin–like immunoreactive substance in hamster basal ganglia after long-term ethanol exposure. Science. 1982;216(4553):1425–1427. doi:10.1126/science.7089531
  • Volkow ND, Fowler JS, Wang GJ, et al. Imaging dopamine’s role in drug abuse and addiction. Neuropharmacology. 2009;56(Suppl1):3–8. doi:10.1016/j.neuropharm.2008.05.022
  • Volkow ND, Fowler JS, Wang GJ, et al. Dopamine in drug abuse and addiction: results of imaging studies and treatment implications. Arch Neurol. 2007;64(11):1575–1579. doi:10.1001/archneur.64.11.1575
  • London ED. Human brain imaging links dopaminergic systems to impulsivity. Curr Topics Behav Neurosci. 2020;47:53–71. doi:10.1007/7854_2019_125
  • Bogomolova EV, Rauschenbach IY, Adonyeva NV, et al. Dopamine down-regulates activity of alkaline phosphatase in drosophila: the role of D2-like receptors. J Insect Physiol. 2010;56(9):1155–1159. doi:10.1016/j.jinsphys.2010.03.014
  • Jeziorski M, White FJ. Dopamine agonists at repeated “autoreceptor-selective” doses: Effects upon the sensitivity of A10 dopamine autoreceptors. Synapse. 1989;4(4):267–280. doi:10.1002/syn.890040403
  • Burris KD, Pacheco Ma, Filtz TM, et al. Lack of discrimination by agonists for D2 and D3 dopamine receptors. Neuropsychopharmacology. 1995;12(4):335–345. doi:10.1016/0893-133X(94)00099-L
  • Luedtke RR, Artymyshyn RP, Monks BR, et al. Comparison of the expression, transcription and genomic organization of D2 dopamine receptors in outbred and inbred strains of rat. Brain Res. 1992;584(1–2):45–54. doi:10.1016/0006-8993(92)90876-b
  • Ivins KJ, Luedtke RR, Artymyshyn RP, et al. Regulation of dopamine D2 receptors in a novel cell line (SUP1). Mol Pharmacol. 1991;39(4):531–539.
  • McGonigle P, Boyson SJ, Reuter S, et al. Effects of chronic treatment with selective and nonselective antagonists on the subtypes of dopamine receptors. Synapse. 1989;3(1):74–82. doi:10.1002/syn.890030111
  • Filtz TM, Artymyshyn RP, Guan W, et al. Paradoxical regulation of dopamine receptors in transfected 293 cells. Mol Pharmacol. 1993;44(2):371–379.
  • Filtz TM, Guan W, Artymyshyn RP, et al. Mechanisms of up-regulation of D2L dopamine receptors by agonists and antagonists in transfected HEK-293 cells. J Pharmacol Exp Ther. 1994;271(3):1574–1582.
  • Burris KD, Fausing SM, Molinoff PB. Regulation of D2 and D3 receptors in transfected cells by agonists and antagonists. Adv Pharmacol. 1998;42:443–446. doi:10.1016/s1054-3589(08)60783-8
  • Boundy VA, Pacheco MA, Guan W, et al. Agonists and antagonists differentially regulate the high affinity state of the D2L receptor in human embryonic kidney 293 cells. Mol Pharmacol. 1995;48(5):956–964.
  • Blum K, Chen AL, Chen TJ, et al. Activation instead of blocking mesolimbic dopaminergic reward circuitry is a preferred modality in the long term treatment of reward deficiency syndrome (RDS): A commentary. Theor Biol Med Model. 2008;5:24. doi:10.1186/1742-4682-5-24
  • Hietala J, West C, Syvalahti E, et al. Striatal D2 dopamine receptor binding characteristics in vivo in patients with alcohol dependence. Psychopharmacology. 1994;116:285–290. doi:10.1007/BF02245330
  • Volkow ND, Wang GJ, Fowler JS, et al. Decreases in dopamine receptors but not in dopamine transporters in alcoholics. Alcohol Clin Exp Res. 1996;20:1594–1598. doi:10.1111/j.1530-0277.1996.tb05936.x
  • Feltmann K, Borroto-Escuela DO, Rüegg J, et al. Effects of Long-term alcohol drinking on the dopamine D2 receptor: gene expression and heteroreceptor complexes in the striatum in rats. Alcohol Clin Exp Res. 2018;42(2):338–351. doi:10.1111/acer.13568
  • Gardner EL. Addiction and brain reward and antireward pathways. Adv Psychosom Med. 2011;30:22–60. doi:10.1159/000324065
  • Blum K, Chen TJ, Downs BW, et al. Neurogenetics of dopaminergic receptor supersensitivity in activation of brain reward circuitry and relapse: Proposing “deprivation-amplification relapse therapy” (DART). Postgrad Med. 2009;121(6):176–196. doi:10.3810/pgm.2009.11.2087
  • Blum K, Badgaiyan RD, Braverman ER, et al. Hypothesizing that, a pro-dopamine regulator (KB220Z) should optimize, but not hyper-activate the activity of trace amine-associated receptor 1 (TAAR-1) and induce anti-craving of psychostimulants in the long-term. J Reward Defic Syndr Addict Sci. 2016;2(1):14–21. doi:10.17756/jrdsas.2016-023
  • Kenneth B, Edward MJ, Marjorie GLC, et al. Pro-Dopamine regulator (KB220) a fifty year sojourn to combat reward deficiency syndrome (RDS): evidence based bibliography (Annotated). CPQ Neurol Psychol. 2018;1(2). Available from: https://www.cientperiodique.com/journal/fulltext/CPQNP/1/2/13.
  • Blum K, Thanos PK, Wang GJ, et al. The food and drug addiction epidemic: targeting dopamine homeostasis. Curr Pharm Des. 2018;23(39):6050–6061. doi:10.2174/1381612823666170823101713
  • McLaughlin T, Blum K, Steinberg B, et al. Pro-dopamine regulator, KB220Z, attenuates hoarding and shopping behavior in a female, diagnosed with SUD and ADHD. J Behav Addict. 2018;7(1):192–203. doi:10.1556/2006.6.2017.081
  • Blum K, Febo M, Fried L, et al. Pro-dopamine regulator - (KB220) to balance brain reward circuitry in reward deficiency syndrome (RDS). J Reward Defic Syndr Addict Sci. 2017;3(1):3–13. doi:10.17756/jrdsas.2017-034
  • McLaughlin T, Han D, Nicholson J, et al. Improvement of long-term memory access with a pro-dopamine regulator in an elderly male: are we targeting dopamine tone? J Syst Integr Neurosci. 2017;3(3). doi:10.15761/JSIN.1000165
  • Blum K, Briggs AH, Trachtenberg MC, et al. Enkephalinase inhibition: regulation of ethanol intake in genetically predisposed mice. Alcohol. 1987;4(6):449–456. doi:10.1016/0741-8329(87)90084-X
  • McLaughlin T, Oscar-Berman M, Simpatico T, et al. Hypothesizing repetitive paraphilia behavior of a medication refractive Tourette’s syndrome patient having rapid clinical attenuation with KB220Z-nutrigenomic amino-acid therapy (NAAT). J Behav Addict. 2013;2(2):117–124. doi:10.1556/JBA.2.2013.2.8
  • McLaughlin T, Blum K, Oscar-Berman M, et al. Putative dopamine agonist (KB220Z) attenuates lucid nightmares in PTSD patients: Role of enhanced brain reward functional connectivity and homeostasis redeeming joy. J Behav Addict. 2015;4(2):106–115. doi:10.1556/2006.4.2015.008
  • Blum K, Chen TJ, Morse S, et al. Overcoming qEEG abnormalities and reward gene deficits during protracted abstinence in male psychostimulant and polydrug abusers utilizing putative dopamine D2 agonist therapy: part 2. Postgrad Med. 2010;122(6):214–226. doi:10.3810/pgm.2010.11.2237
  • Blum K, Liu Y, Wang W, et al. rsfMRI effects of KB220Z™ on neural pathways in reward circuitry of abstinent genotyped heroin addicts. Postgrad Med. 2015;127(2):232–241. doi:10.1080/00325481.2015.994879
  • Blum K, Chen AL, Chen TJ, et al. LG839: anti-obesity effects and polymorphic gene correlates of reward deficiency syndrome. Adv Ther. 2008;25(9):894–913. doi:10.1007/s12325-008-0093-z
  • Brown RJ, Blum K, Trachtenberg MC. Neurodynamics of relapse prevention: a neuronutrient approach to outpatient DUI offenders. J Psychoactive Drugs. 1990;22(2):173–187. doi:10.1080/02791072.1990.10472542
  • Blum K, Trachtenberg MC, Elliott CE, et al. Enkephalinase inhibition and precursor amino acid loading improves inpatient treatment of alcohol and polydrug abusers: double-blind placebo-controlled study of the nutritional adjunct SAAVE. Alcohol. 1988;5(6):481–493. doi:10.1016/0741-8329(88)90087-0
  • Trachtenberg MC, Blum K. Improvement of cocaine-induced neuromodulator deficits by the neuronutrient tropamine. J Psychoactive Drugs. 1988;20(3):315–331. doi:10.1080/02791072.1988.10472501
  • Blum K, Chen ALC, Chen TJH, et al. Putative targeting of Dopamine D2 receptor function in Reward Deficiency Syndrome (RDS) by Synaptamine Complex™ Variant (KB220): clinical trial showing anti-anxiety effects. Gene Ther Mol Bio. 2009;13:214–230.
  • Solanki N, Abijo T, Galvao C, et al. Administration of a putative pro-dopamine regulator, a neuronutrient, mitigates alcohol intake in alcohol-preferring rats. Behav Brain Res. 2020;385:112563. PMID: 32070691. doi:10.1016/j.bbr.2020.112563
  • Thanos PK, Michaelides M, Umegaki H, et al. D2R DNA transfer into the nucleus accumbens attenuates cocaine self-administration in rats. Synapse. 2008;62(7):481–486. doi:10.1002/syn.20523
  • Thanos PK, Volkow ND, Freimuth P, et al. Overexpression of dopamine D2 receptors reduces alcohol self-administration. J Neurochem. 2001;78(5):1094–1103. doi:10.1046/j.1471-4159.2001.00492.x
  • Hu Y, Salmeron BJ, Krasnova IN, et al. Compulsive drug use is associated with imbalance of orbitofrontal- and prelimbic-striatal circuits in punishment-resistant individuals. Proc Natl Acad Sci U S A. 2019;116(18):9066–9071. doi:10.1073/pnas.1819978116
  • Febo M, Blum K, Badgaiyan RD, et al. Enhanced functional connectivity and volume between cognitive and reward centers of naïve rodent brain produced by pro-dopaminergic agent KB220Z. PLoS One. 2017;12(4):e0174774. doi:10.1371/journal.pone.0174774
  • Tapocik JD, Luu TV, Mayo CL, et al. Neuroplasticity, axonal guidance and micro-RNA genes are associated with morphine self-administration behavior. Addict Biol. 2013;18(3):480–495. doi:10.1111/j.1369-1600.2012.00470.x
  • Bertran-Gonzalez J, Hervé D, Girault JA, et al. What is the degree of segregation between striatonigral and striatopallidal projections? Front Neuroanat. 2010;4:136. doi:10.3389/fnana.2010.00136
  • Hikida T, Yawata S, Yamaguchi T, et al. Pathway-specific modulation of nucleus accumbens in reward and aversive behavior via selective transmitter receptors. Proc Natl Acad Sci U S A. 2013;110(1):342–347. doi:10.1073/pnas.1220358110
  • Hikida T, Kimura K, Wada N, et al. Distinct roles of synaptic transmission in direct and indirect striatal pathways to reward and aversive behavior. Neuron. 2010;66(6):896–907. doi:10.1016/j.neuron.2010.05.011
  • Noble EP, Blum K, Ritchie T, et al. Allelic association of the D2 dopamine receptor gene with receptor-binding characteristics in alcoholism. Arch Gen Psychiatry. 1991;48(7):648–654. PMID: 2069496. doi:10.1001/archpsyc.1991.01810310066012
  • Yudell M, Roberts D, DeSalle R, et al. Science and society: taking race out of human genetics. Science. 2016;351(6273):564–565. doi:10.1126/science.aac4951
  • Goldman D, Brown GL, Albaugh B, et al. DRD2 dopamine receptor genotype, linkage disequilibrium, and alcoholism in American Indians and other populations. Alcohol Clin Exp Res. 1993;17(2):199–204. doi:10.1111/j.1530-0277.1993.tb00749.x
  • Levy JE, Kunitz SJ. Indian Drinking: Navajo Practice and Anglo-American Theories. New York: Wiley; 1974.
  • Ishiguro H, Arinami T, Saito T, et al. Association study between the −141C Ins/Del and TaqI A polymorphisms of the dopamine D2 receptor gene and alcoholism. Alcohol Clin Exp Res. 1998;22(4):845–848.
  • Arinami T, Itokawa M, Komiyama T, et al. Association between severity of alcoholism and the A1 allele of the dopamine D2 receptor gene TaqI A RFLP in Japanese. Biol Psychiatry. 1993;33(2):108–114. doi:10.1016/0006-3223(93)90309-2
  • Berrettini WH, Persico AM. Dopamine D2 receptor gene polymorphisms and vulnerability to substance abuse in African Americans. Biol Psychiatry. 1996;40(2):144–147. doi:10.1016/0006-3223(96)00036-4
  • O’Hara BF, Smith SS, Bird G, et al. Dopamine D2 receptor RFLPs, haplotypes and their association with substance use in black and Caucasian research volunteers. Hum Hered. 1993;43(4):209–218. doi:10.1159/000154133
  • Blum K, Noble EP, Sheridan PJ, et al. Genetic predisposition in alcoholism: association of the D2 dopamine receptor TaqI B1 RFLP with severe alcoholics. Alcohol. 1993;10(1):59–67. doi:10.1016/0741-8329(93)90054-r
  • Ettienne EB, Chapman E, Maneno M, et al. Pharmacogenomics-guided policy in opioid use disorder (OUD) management: an ethnically-diverse case-based approach. Addict Behav Rep. 2017;6:8–14. doi:10.1016/j.abrep.2017.05.001
  • Ettienne EB, Ofoegbu A, Maneno MK, et al. Pharmacogenomics and opioid use disorder: clinical decision support in an African American cohort. J Natl Med Assoc. 2019;111(6):674–681. doi:10.1016/j.jnma.2019.09.006
  • Reuss CV, Kalava A, King KC. Prescription of Controlled Substances: benefits and Risks. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021. PMID: 30726003.
  • Accurso AJ, Rastegar DA. The effect of a payer-mandated decrease in buprenorphine dose on aberrant drug tests and treatment retention among patients with opioid dependence. J Subst Abuse Treat. 2016;61:74–79. doi:10.1016/j.jsat.2015.09.004
  • Volkow N. Toward precision medicine in addiction treatment. Am J Addict. 2018;27(1):35–36. doi:10.1111/ajad.12666
  • Blum K, Chen TJ, Meshkin B, et al. Genotrim, a DNA-customized nutrigenomic product, targets genetic factors of obesity: hypothesizing a dopamine-glucose correlation demonstrating reward deficiency syndrome (RDS). Med Hypotheses. 2007;68(4):844–852. doi:10.1016/j.mehy.2006.08.041
  • Blum K, Chen TJ, Meshkin B, et al. Reward deficiency syndrome in obesity: a preliminary cross-sectional trial with a Genotrim variant. Adv Ther. 2006;23(6):1040–1051. doi:10.1007/BF02850224
  • Delker E, Brown Q, Hasin DS. Alcohol consumption in demographic subpopulations: an epidemiologic overview. Alcohol Res. 2016;38(1):7–15.
  • Downs B, Oscar-Berman M, Waite R, et al. Have we hatched the addiction egg: reward deficiency syndrome solution system™. J Genet Syndr Gene Ther. 2013;4(136):14318. doi:10.4172/2157-7412.100013
  • Blum K, Febo M, Fried L, et al. Hypothesizing that neuropharmacological and neuroimaging studies of glutaminergic-dopaminergic optimization complex (KB220Z) are associated with “dopamine homeostasis” in reward deficiency syndrome (RDS). Subst Use Misuse. 2017;52(4):535–547. PMID: 2803347. doi:10.1080/10826084.2016.1244551
  • Nurnberger JI, Austin J, Berrettini WH, et al. What should a Psychiatrist know about genetics? Review and recommendations from the residency education committee of the International Society of Psychiatric Genetics. J Clin Psychiatry. 2018;80(1):17nr12046. doi:10.4088/JCP.17nr12046
  • Chapman EC, Ettiene E, Ofoegbu A, et al. The impact of pharmacogenetic testing on buprenorphine dosing in an African-American population. Second International Conference on Addiction Medicine and Reward Deficiency Syndrome. March 17–19; 2017; Baltimore, MD.
  • Leavey DF, Stein MB, Wendt FR, et al. Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat Neurosci. 2021;24(7):954–963. doi:10.1038/s41593-021-00860-2