We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Pharmacogenetics of neural injury recovery

    Kristin M Pearson-Fuhrhop

    Department of Anatomy & Neurobiology, University of California, Irvine, 200 S Manchester Avenue, Suite 206, Orange, CA 92868, USA

    &
    Steven C Cramer

    * Author for correspondence

    Department of Neurology, University of California, Irvine, 200 S Manchester Ave, Suite 206, Orange, CA 92868, USA.

    Published Online:https://doi.org/10.2217/pgs.13.152

    Relatively few pharmacological agents are part of routine care for neural injury, although several are used or under consideration in acute stroke, chronic stroke, traumatic brain injury and secondary stroke prevention. Tissue plasminogen activator is approved for the treatment of acute ischemic stroke, and genetic variants may impact the efficacy and safety of this drug. In the chronic phase of stroke, several drugs such as L-dopa, fluoxetine and donepezil are under investigation for enhancing rehabilitation therapy, with varying levels of evidence. One potential reason for the mixed efficacy displayed by these drugs may be the influence of genetic factors that were not considered in prior studies. An understanding of the genetics impacting the efficacy of dopaminergic, serotonergic and cholinergic drugs may allow clinicians to target these potential therapies to those patients most likely to benefit. In the setting of stroke prevention, which is directly linked to neural injury recovery, the most highly studied pharmacogenomic interactions pertain to clopidogrel and warfarin. Incorporating pharmacogenomics into neural injury recovery has the potential to maximize the benefit of several current and potential pharmacological therapies and to refine the choice of pharmacological agent that may be used to enhance benefits from rehabilitation therapy.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • del Rio-Espinola A, Fernandez-Cadenas I, Giralt D et al. A predictive clinical–genetic model of tissue plasminogen activator response in acute ischemic stroke. Ann. Neurol.72(5),716–729 (2012).▪ Thorough and important study examining and replicating genetic associations and then building a predictive model for tissue plasminogen activator safety using both clinical and genetic variables. These are important steps toward utilizing pharmacogenetic information.
    • Fernandez-Cadenas I, Del Rio-Espinola A, Giralt D et al.IL1B and VWF variants are associated with fibrinolytic early recanalization in patients with ischemic stroke. Stroke43(10),2659–2665 (2012).
    • Engelter ST, Urscheler N, Baronti F et al. Frequency and determinants of using pharmacological enhancement in the clinical practice of in-hospital stroke rehabilitation. Eur. Neurol.68(1),28–33 (2012).
    • Scheidtmann K, Fries W, Muller F, Koenig E. Effect of levodopa in combination with physiotherapy on functional motor recovery after stroke: a prospective, randomised, double-blind study. Lancet358,787–790 (2001).
    • Floel A, Hummel F, Breitenstein C, Knecht S, Cohen LG. Dopaminergic effects on encoding of a motor memory in chronic stroke. Neurology65(3),472–474 (2005).
    • Rosser N, Heuschmann P, Wersching H, Breitenstein C, Knecht S, Floel A. Levodopa improves procedural motor learning in chronic stroke patients. Arch. Phys. Med. Rehabil.89(9),1633–1641 (2008).
    • Chollet F, Tardy J, Albucher JF et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol.10(2),123–130 (2011).▪▪ Landmark study showing that focused manipulation of a neurochemical axis can improve outcome after stroke, in this case arm motor function at day 90. Few drugs are frequently prescribed to enhance rehabilitation following stroke, and this randomized, placebo-controlled trial illustrates that fluoxetine may aid in both functional recovery and the prevention of post-stroke depression. These findings have major implications for pharmacogenetics.
    • Jorge RE, Robinson RG, Arndt S, Starkstein S. Mortality and poststroke depression: a placebo-controlled trial of antidepressants. Am. J. Psychiatry160(10),1823–1829 (2003).
    • Markus HS. Stroke genetics: prospects for personalized medicine. BMC Med.10,113 (2012).
    • 10  Ezekowitz MD, Aikens TH, Brown A, Ellis Z. The evolving field of stroke prevention in patients with atrial fibrillation. Stroke41(10 Suppl.),S17–S20 (2010).
    • 11  Mega JL, Close SL, Wiviott SD et al. Cytochrome P-450 polymorphisms and response to clopidogrel. N. Engl. J. Med.360(4),354–362 (2009).▪▪ Comprehensive study examining the effect of cytochrome P450 polymorphisms on several aspects of clopidogrel function in both healthy adults and patients with stroke.
    • 12  Anderson JL, Horne BD, Stevens SM et al. Randomized trial of genotype-guided versus standard warfarin dosing in patients initiating oral anticoagulation. Circulation116(22),2563–2570 (2007).
    • 13  Mandava P, Murthy SB, Munoz M et al. Explicit consideration of baseline factors to assess recombinant tissue-type plasminogen activator response with respect to race and sex. Stroke44(6),1525–1531 (2013).
    • 14  Engelter ST. Safety in pharmacological enhancement of stroke rehabilitation. Eur. J. Phys. Rehabil. Med.49(2),261–267 (2013).▪▪ Presents a very thorough description of the pharmacological agents that have been explored as enhancements to post-stroke rehabilitation, including the adverse events reported by these studies.
    • 15  Dam M, Tonin P, De Boni A et al. Effects of fluoxetine and maprotiline on functional recovery in poststroke hemiplegic patients undergoing rehabilitation therapy. Stroke27(7),1211–1214 (1996).
    • 16  Pariente J, Loubinoux I, Carel C et al. Fluoxetine modulates motor performance and cerebral activation of patients recovering from stroke. Ann. Neurol.50(6),718–729 (2001).
    • 17  Zittel S, Weiller C, Liepert J. Citalopram improves dexterity in chronic stroke patients. Neurorehabil. Neural Repair22(3),311–314 (2008).
    • 18  Zittel S, Weiller C, Liepert J. Reboxetine improves motor function in chronic stroke. A pilot study. J. Neurol.254(2),197–201 (2007).
    • 19  Walker-Batson D, Smith P, Curtis S, Unwin H, Greenlee R. Amphetamine paired with physical therapy accelerates motor recovery after stroke. Further evidence. Stroke26(12),2254–2259 (1995).
    • 20  Feeney DM, Gonzalez A, Law WA. Amphetamine, haloperidol, and experience interact to affect rate of recovery after motor cortex injury. Science217(4562),855–857 (1982).
    • 21  Martinsson L, Hardemark H, Eksborg S. Amphetamines for improving recovery after stroke. Cochrane Database Syst. Rev.1,CD002090 (2007).
    • 22  Schuster C, Maunz G, Lutz K, Kischka U, Sturzenegger R, Ettlin T. Dexamphetamine improves upper extremity outcome during rehabilitation after stroke: a pilot randomized controlled trial. Neurorehabil. Neural Repair25(8),749–755 (2011).
    • 23  Grade C, Redford B, Chrostowski J, Toussaint L, Blackwell B. Methylphenidate in early poststroke recovery: a double-blind, placebo-controlled study. Arch. Phys. Med. Rehabil.79(9),1047–1050 (1998).
    • 24  Lokk J, Salman Roghani R, Delbari A. Effect of methylphenidate and/or levodopa coupled with physiotherapy on functional and motor recovery after stroke – a randomized, double-blind, placebo-controlled trial. Acta Neurol. Scand.123(4),266–273 (2011).
    • 25  Delbari A, Salman-Roghani R, Lokk J. Effect of methylphenidate and/or levodopa combined with physiotherapy on mood and cognition after stroke: a randomized, double-blind, placebo-controlled trial. Eur. Neurol.66(1),7–13 (2011).
    • 26  Berthier ML, Green C, Higueras C, Fernandez I, Hinojosa J, Martin MC. A randomized, placebo-controlled study of donepezil in poststroke aphasia. Neurology67(9),1687–1689 (2006).
    • 27  Whyte EM, Lenze EJ, Butters M et al. An open-label pilot study of acetylcholinesterase inhibitors to promote functional recovery in elderly cognitively impaired stroke patients. Cerebrovasc. Dis.26(3),317–321 (2008).
    • 28  Sonde L, Lokk J. Effects of amphetamine and/or L-dopa and physiotherapy after stroke – a blinded randomized study. Acta Neurol. Scand.115(1),55–59 (2007).
    • 29  Pearson-Fuhrhop KM, Minton B, Acevedo D, Shahbaba B, Cramer SC. Genetic variation in the human brain dopamine system influences motor learning and its modulation by L-dopa. PLoS ONE8(4),e61197 (2013).▪▪ Found that a dopamine gene score was associated with skilled motor learning and modulated the effect of L-dopa on motor learning. These findings can explain some of the variability in studies of L-dopa efficacy and have the potential to guide the assignment of L-dopa with rehabilitation therapy.
    • 30  Becker ML, Visser LE, van Schaik RH, Hofman A, Uitterlinden AG, Stricker BH. OCT1 polymorphism is associated with response and survival time in anti-Parkinsonian drug users. Neurogenetics12(1),79–82 (2011).
    • 31  Kato M, Serretti A. Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol. Psychiatry15(5),473–500 (2010).
    • 32  Lewis G, Mulligan J, Wiles N et al. Polymorphism of the 5-HT transporter and response to antidepressants: randomised controlled trial. Br. J. Psychiatry198(6),464–471 (2011).
    • 33  McGuffin P, Alsabban S, Uher R. The truth about genetic variation in the serotonin transporter gene and response to stress and medication. Br. J. Psychiatry198(6),424–427 (2011).
    • 34  Hadidi N, Treat-Jacobson DJ, Lindquist R. Poststroke depression and functional outcome: a critical review of literature. Heart Lung38(2),151–162 (2009).
    • 35  Smits KM, Smits LJ, Schouten JS, Stelma FF, Nelemans P, Prins MH. Influence of SERTPR and STin2 in the serotonin transporter gene on the effect of selective serotonin reuptake inhibitors in depression: a systematic review. Mol. Psychiatry9(5),433–441 (2004).
    • 36  Kohen R, Cain KC, Buzaitis A et al. Response to psychosocial treatment in poststroke depression is associated with serotonin transporter polymorphisms. Stroke42(7),2068–2070 (2011).▪ Studied the relationship between serotonin transporter protein polymorphisms and response to psychotherapy intervention in survivors of stroke; although these polymorphisms are associated with poorer response to pharmacological antidepressant therapy, they were found to predict greater response to psychotherapy in this population.
    • 37  Pinto N, Dolan ME. Clinically relevant genetic variations in drug metabolizing enzymes. Curr. Drug Metab.12(5),487–497 (2011).
    • 38  Zhong Y, Zheng X, Miao Y, Wan L, Yan H, Wang B. Effect of CYP2D6*10 and APOE polymorphisms on the efficacy of donepezil in patients with Alzheimer‘s disease. Am. J. Med. Sci.345(3),222–226 (2013).
    • 39  Albani D, Martinelli Boneschi F, Biella G et al. Replication study to confirm the role of CYP2D6 polymorphism rs1080985 on donepezil efficacy in Alzheimer‘s disease patients. J. Alzheimers Dis.30(4),745–749 (2012).
    • 40  Pilotto A, Franceschi M, D‘Onofrio G et al. Effect of a CYP2D6 polymorphism on the efficacy of donepezil in patients with Alzheimer disease. Neurology73(10),761–767 (2009).
    • 41  Seripa D, Bizzarro A, Pilotto A et al. Role of cytochrome P4502D6 functional polymorphisms in the efficacy of donepezil in patients with Alzheimer‘s disease. Pharmacogenet. Genomics21(4),225–230 (2011).
    • 42  Samson AL, Medcalf RL. Tissue-type plasminogen activator: a multifaceted modulator of neurotransmission and synaptic plasticity. Neuron50(5),673–678 (2006).
    • 43  Tjarnlund-Wolf A, Hultman K, Curtis MA, Faull RL, Medcalf RL, Jern C. Allelic imbalance of tissue-type plasminogen activator (t-PA) gene expression in human brain tissue. Thromb. Haemost.105(6),945–953 (2011).
    • 44  Ladenvall P, Johansson L, Jansson JH et al. Tissue-type plasminogen activator -7,351C/T enhancer polymorphism is associated with a first myocardial infarction. Thromb. Haemost.87(1),105–109 (2002).
    • 45  Jannes J, Hamilton-Bruce MA, Pilotto L et al. Tissue plasminogen activator -7351C/T enhancer polymorphism is a risk factor for lacunar stroke. Stroke35(5),1090–1094 (2004).
    • 46  Maguire J, Thakkinstian A, Levi C et al. Impact of COX-2 rs5275 and rs20417 and GPIIIa rs5918 polymorphisms on 90-day ischemic stroke functional outcome: a novel finding. J. Stroke Cerebrovasc. Dis.20(2),134–144 (2011).
    • 47  Depondt C, Godard P, Espel RS, Da Cruz AL, Lienard P, Pandolfo M. A candidate gene study of antiepileptic drug tolerability and efficacy identifies an association of CYP2C9 variants with phenytoin toxicity. Eur. J. Neurol.18(9),1159–1164 (2011).
    • 48  Tarasova L, Kalnina I, Geldnere K et al. Association of genetic variation in the organic cation transporters OCT1, OCT2 and multidrug and toxin extrusion 1 transporter protein genes with the gastrointestinal side effects and lower BMI in metformin-treated Type 2 diabetes patients. Pharmacogenet. Genomics22(9),659–666 (2012).
    • 49  Jekic B, Lukovic L, Bunjevacki V et al. Association of the TYMS 3G/3G genotype with poor response and GGH 354GG genotype with the bone marrow toxicity of the methotrexate in RA patients. Eur. J. Clin. Pharmacol.69(3),377–383 (2012).
    • 50  Leandro-Garcia LJ, Leskela S, Jara C et al. Regulatory polymorphisms in β-tubulin iia are associated with paclitaxel-induced peripheral neuropathy. Clin Cancer Res.18(16),4441–4448 (2012).
    • 51  Kawashima S, Ueki Y, Kato T et al. Changes in striatal dopamine release associated with human motor-skill acquisition. PLoS ONE7(2),e31728 (2012).
    • 52  Kalinderi K, Fidani L, Katsarou Z, Bostantjopoulou S. Pharmacological treatment and the prospect of pharmacogenetics in Parkinson‘s disease. Int. J. Clin. Pract.65(12),1289–1294 (2011).
    • 53  Fox SH, Lang AE. Levodopa-related motor complications – phenomenology. Mov. Disord.23(Suppl. 3),S509–S514 (2008).
    • 54  Shamy MC, Zai C, Basile VS, Kennedy JL, Muller DJ, Masellis M. Ethical and policy considerations in the application of pharmacogenomic testing for tardive dyskinesia: case study of the dopamine D3 receptor. Curr. Pharmacogenomics Person. Med.9(2),94–101 (2011).▪ Valuable review of the ethical, social and policy issues surrounding the use of pharmacogenetics in clinical practice. It is particularly relevant as the field of pharmacogenetics is becoming increasingly clinically relevant, yet genetic information is sensitive and could easily be mistreated.
    • 55  Pearson-Fuhrhop KM, Kleim JA, Cramer SC. Brain plasticity and genetic factors. Top. Stroke Rehabil.16(4),282–299 (2009).
    • 56  Pearson-Fuhrhop KM, Burke E, Cramer SC. The influence of genetic factors on brain plasticity and recovery after neural injury. Curr. Opin. Neurol.25(6),682–688 (2012).
    • 57  Simon RP, Meller R, Zhou A, Henshall D. Can genes modify stroke outcome and by what mechanisms?. Stroke43(1),286–291 (2012).
    • 58  Egan MF, Kojima M, Callicott JH et al. The BDNF Val66Met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell112(2),257–269 (2003).
    • 59  Chen ZY, Patel PD, Sant G et al. Variant brain-derived neurotrophic factor (BDNF) (Met66) alters the intracellular trafficking and activity-dependent secretion of wild-type BDNF in neurosecretory cells and cortical neurons. J. Neurosci.24(18),4401–4411 (2004).
    • 60  Kleim JA, Chan S, Pringle E et al.BDNF Val66Met polymorphism is associated with modified experience-dependent plasticity in human motor cortex. Nat. Neurosci.9(6),735–737 (2006).
    • 61  Cheeran B, Talelli P, Mori F et al. A common polymorphism in the brain-derived neurotrophic factor gene (BDNF) modulates human cortical plasticity and the response to rTMS. J. Physiol.586(Pt 23),5717–5725 (2008).
    • 62  McHughen SA, Rodriguez PF, Kleim JA et al.BDNF Val66Met polymorphism influences motor system function in the human brain. Cereb. Cortex20(5),1254–1262 (2010).
    • 63  McHughen SA, Pearson-Fuhrhop K, Ngo VK, Cramer SC. Intense training overcomes effects of the Val66Met BDNF polymorphism on short-term plasticity. Exp. Brain Res.213(4),415–422 (2011).
    • 64  Siironen J, Juvela S, Kanarek K, Vilkki J, Hernesniemi J, Lappalainen J. The Met allele of the BDNF Val66Met polymorphism predicts poor outcome among survivors of aneurysmal subarachnoid hemorrhage. Stroke38(10),2858–2860 (2007).
    • 65  Cramer SC, Procaccio V. Correlation between genetic polymorphisms and stroke recovery: analysis of the GAIN Americas and GAIN International Studies. Eur. J. Neurol.19(5),718–724 (2012).
    • 66  Shimizu E, Hashimoto K, Iyo M. Ethnic difference of the BDNF 196G/A (Val66Met) polymorphism frequencies: the possibility to explain ethnic mental traits. Am. J. Med. Genet. B Neuropsychiatr. Genet.126(1),122–123 (2004).
    • 67  Mahley RW, Rall SC Jr. Apolipoprotein E: far more than a lipid transport protein. Annu. Rev. Genomics Hum. Genet.1,507–537 (2000).
    • 68  Cedazo-Minguez A. Apolipoprotein E and Alzheimer‘s disease: molecular mechanisms and therapeutic opportunities. J. Cell. Mol. Med.11(6),1227–1238 (2007).
    • 69  Corder EH, Saunders AM, Strittmatter WJ et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer‘s disease in late onset families. Science261(5123),921–923 (1993).
    • 70  Hyman BT, Gomez-Isla T, Rebeck GW et al. Epidemiological, clinical, and neuropathological study of apolipoprotein E genotype in Alzheimer‘s disease. Ann. NY Acad. Sci.802,1–5 (1996).
    • 71  Ponsford J, McLaren A, Schonberger M et al. The association between apolipoprotein E and traumatic brain injury severity and functional outcome in a rehabilitation sample. J. Neurotrauma28(9),1683–1692 (2011).
    • 72  Zhou W, Xu D, Peng X, Zhang Q, Jia J, Crutcher KA. Meta-analysis of APOE4 allele and outcome after traumatic brain injury. J. Neurotrauma25(4),279–290 (2008).
    • 73  Marousi S, Ellul J, Antonacopoulou A, Gogos C, Papathanasopoulos P, Karakantza M. Functional polymorphisms of interleukin 4 and interleukin 10 may predict evolution and functional outcome of an ischaemic stroke. Eur. J. Neurol.18(4),637–643 (2011).
    • 74  Weaver SM, Chau A, Portelli JN, Grafman J. Genetic polymorphisms influence recovery from traumatic brain injury. Neuroscientist18(6),631–644 (2012).
    • 75  Floresco SB, Jentsch JD. Pharmacological enhancement of memory and executive functioning in laboratory animals. Neuropsychopharmacology36(1),227–250 (2011).
    • 76  Husain M, Mehta MA. Cognitive enhancement by drugs in health and disease. Trends Cogn. Sci.15(1),28–36 (2011).
    • 77  Meschia JF. Pharmacogenetics and stroke. Stroke40(11),3641–3645 (2009).
    • 78  Chan A, Pirmohamed M, Comabella M. Pharmacogenomics in neurology: current state and future steps. Ann. Neurol.70(5),684–697 (2011).
    • 79  Shuldiner AR, O‘Connell JR, Bliden KP et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA302(8),849–857 (2009).
    • 80  Li Y, Tang HL, Hu YF, Xie HG. The gain-of-function variant allele CYP2C19*17: a double-edged sword between thrombosis and bleeding in clopidogrel-treated patients. J. Thromb. Haemost.10(2),199–206 (2012).
    • 81  Holmes DR Jr, Dehmer GJ, Kaul S, Leifer D, O‘Gara PT, Stein CM. ACCF/AHA clopidogrel clinical alert: approaches to the FDA ‘boxed warning‘: a report of the American College of Cardiology Foundation Task Force on clinical expert consensus documents and the American Heart Association endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. J. Am. Coll. Cardiol.56(4),321–341 (2010).
    • 82  Sibbing D, Bernlochner I, Kastrati A, Pare G, Eikelboom JW. Current evidence for genetic testing in clopidogrel-treated patients undergoing coronary stenting. Circ. Cardiovasc. Interv.4(5),505–513; discussion 513 (2011).
    • 83  Pare G, Eikelboom JW, Sibbing D, Bernlochner I, Kastrati A. Testing should not be done in all patients treated with clopidogrel who are undergoing percutaneous coronary intervention. Circ. Cardiovasc. Interv.4(5),514–521; discussion 521 (2011).
    • 84  Wadelius M, Pirmohamed M. Pharmacogenetics of warfarin: current status and future challenges. Pharmacogenomics J.7(2),99–111 (2007).
    • 85  Cooper GM, Johnson JA, Langaee TY et al. A genome-wide scan for common genetic variants with a large influence on warfarin maintenance dose. Blood112(4),1022–1027 (2008).
    • 86  Takeuchi F, McGinnis R, Bourgeois S et al. A genome-wide association study confirms VKORC1, CYP2C9, and CYP4F2 as principal genetic determinants of warfarin dose. PLoS Genet.5(3),e1000433 (2009).
    • 87  Daneshjou R, Tatonetti NP, Karczewski KJ et al. Pathway analysis of genome-wide data improves warfarin dose prediction. BMC Genomics14(Suppl. 3),S11 (2013).
    • 88  Perera MA, Cavallari LH, Limdi NA et al. Genetic variants associated with warfarin dose in African–American individuals: a genome-wide association study. Lancet doi:10.1016/S0140-6736(13)60681-9 (2013) (Epub ahead of print).
    • 89  Lewis JP, Ryan K, O‘Connell JR, et al. Genetic variation in PEAR1 is associated with platelet aggregation and cardiovascular outcomes. Circ. Cardiovasc. Genet.6(2),184–192 (2013).
    • 90  Al-Azzam SI, Alzoubi KH, Khabour OF, Tawalbeh D, Al-Azzeh O. The contribution of platelet glycoproteins (GPIa C807T and GPIba C-5T) and cyclooxygenase 2 (COX-2G-765C) polymorphisms to platelet response in patients treated with aspirin. Gene526(2),118–121 (2013).
    • 91  Giacino JT, Whyte J, Bagiella E et al. Placebo-controlled trial of amantadine for severe traumatic brain injury. N. Engl. J. Med.366(9),819–826 (2012).