Journal Home > Volume 5 , Issue 4

Migraine is a difficult disorder to identify with regard to its pathophysiological mechanisms, and its treatment has been primarily difficult owing to interindividual differences. Substantial rates of nonresponsiveness to medications are common, making migraine treatment complicated. In this review, we systematically analyzed recent studies concerning neuroimaging findings regarding the neurophysiology of migraine. We linked the current imaging research with anecdotal evidence from interindividual factors such as duration and pain intensity of migraine, age, gender, hormonal interplay, and genetics. These factors suggested the use of nonpharmacological therapies such as transcranial magnetic stimulation, transcranial direct current stimulation, and placebo therapy for the treatment of migraine. Finally, we discussed how interindividual differences are related to such nondrug treatments.


menu
Abstract
Full text
Outline
About this article

Individual differences of maladaptive brain changes in migraine and their relationship with differential effectiveness of treatments

Show Author's information Karen M. von Deneen1,2Ling Zhao3( )Jixin Liu1,2( )
Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an 710126, Shaanxi, China
Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi'an 710126, Shaanxi, China
Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China

Abstract

Migraine is a difficult disorder to identify with regard to its pathophysiological mechanisms, and its treatment has been primarily difficult owing to interindividual differences. Substantial rates of nonresponsiveness to medications are common, making migraine treatment complicated. In this review, we systematically analyzed recent studies concerning neuroimaging findings regarding the neurophysiology of migraine. We linked the current imaging research with anecdotal evidence from interindividual factors such as duration and pain intensity of migraine, age, gender, hormonal interplay, and genetics. These factors suggested the use of nonpharmacological therapies such as transcranial magnetic stimulation, transcranial direct current stimulation, and placebo therapy for the treatment of migraine. Finally, we discussed how interindividual differences are related to such nondrug treatments.

Keywords: neuroimaging, migraine, nonpharmacological treatment, individual differences

References(155)

[1]
D Borsook, N Maleki, L Becerra, et al. Understanding migraine through the lens of maladaptive stress responses: a model disease of allostatic load. Neuron. 2012, 73(2): 219-234.
[2]
RB Lipton, ME Bigal, M Diamond, et al. Migraine prevalence, disease burden, and the need for preventive therapy. Neurology. 2007, 68(5): 343-349.
[3]
YW Woldeamanuel, RP Cowan. Migraine affects 1 in 10 people worldwide featuring recent rise: a systematic review and meta-analysis of community-based studies involving 6 million participants. J Neurol Sci. 2017, 372: 307-315.
[4]
XH Hu, LE Markson, RB Lipton, et al. Burden of migraine in the United States: disability and economic costs. Arch Intern Med. 1999, 159(8): 813-818.
[5]
DI Friedman, T de Ver Dye. Migraine and the environment. Headache. 2009, 49(6): 941-952.
[6]
TA Smitherman, R Burch, H Sheikh, et al. The prevalence, impact, and treatment of migraine and severe headaches in the United States: a review of statistics from national surveillance studies. Headache. 2013, 53(3): 427-436.
[7]
TJ Schwedt, DW Dodick. Advanced neuroimaging of migraine. Lancet Neurol. 2009, 8(6): 560-568.
[8]
S Derry, RA Moore. Paracetamol (acetaminophen) with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev. 2013(4): CD008040.
[9]
C Suthisisang, N Poolsup, W Kittikulsuth, et al. Efficacy of low-dose ibuprofen in acute migraine treatment: systematic review and meta-analysis. Ann Pharmacother. 2007, 41(11): 1782-1791.
[10]
J Pascual, V Mateos, C Roig, et al. Marketed oral triptans in the acute treatment of migraine: a systematic review on efficacy and tolerability. Headache. 2007, 47(8): 1152-1168.
[11]
S Silberstein, SA McDonald, J Goldstein, et al. Sumatriptan/naproxen sodium for the acute treatment of probable migraine without aura: a randomized study. Cephalalgia. 2014, 34(4): 268-279.
[12]
RM Gallagher. Acute treatment of migraine with dihydroergotamine nasal spray. Arch Neurol. 1996, 53(12): 1285-1291.
[13]
SD Silberstein, FG Freitag, TD Rozen, et al. Tramadol/acetaminophen for the treatment of acute migraine pain: findings of a randomized, placebo- controlled trial. Headache. 2005, 45(10): 1317-1327.
[14]
EA Schulman, KF Dermott. Sumatriptan plus metoclopramide in triptan-nonresponsive migraineurs. Headache. 2003, 43(7): 729-733.
[15]
SD Silberstein. Practice parameter: evidence-based guidelines for migraine headache (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000, 55(6): 754-762.
[16]
H Hedegaard, AM Miniño, M Warner. Drug overdose deaths in the United States, 1999-2017. NCHS Data Brief. 2018, 329: 1-8.
[17]
SH Schnoll, MF Weaver. Addiction and pain. Am J Addict. 2003, 12: S27-S35.
[18]
L Scholl, P Seth, M Kariisa, et al. Drug and opioid- involved overdose deaths—United States, 2013-2017. MMWR Morb Mortal Wkly Rep. 2018, 67(5152): 1419-1427.
[19]
RE Wells, J Beuthin, L Granetzke. Complementary and integrative medicine for episodic migraine: an update of evidence from the last 3 years. Curr Pain Headache Rep. 2019, 23(2): 10.
[20]
RR Edwards, RH Dworkin, DC Turk, et al. Patient phenotyping in clinical trials of chronic pain treatments: IMMPACT recommendations. Pain. 2016, 157(9): 1851-1871.
[21]
M Kobayashi, A Pascual-Leone. Transcranial magnetic stimulation in neurology. Lancet Neurol. 2003, 2(3): 145-156.
[22]
AT Barker, R Jalinous, IL Freeston. Non-invasive magnetic stimulation of human motor cortex. Lancet. 1985, 1(8437): 1106-1107.
[23]
RB Lipton, SH Pearlman. Transcranial magnetic simulation in the treatment of migraine. Neurotherapeutics. 2010, 7(2): 204-212.
[24]
R Bhola, E Kinsella, N Giffin, et al. Single-pulse transcranial magnetic stimulation (sTMS) for the acute treatment of migraine: evaluation of outcome data for the UK post market pilot program. J Headache Pain. 2015, 16: 535.
[25]
RB Lipton, DW Dodick, SD Silberstein, et al. Single- pulse transcranial magnetic stimulation for acute treatment of migraine with aura: a randomised, double- blind, parallel-group, sham-controlled trial. Lancet Neurol. 2010, 9(4): 373-380.
[26]
S Rocha, L Melo, C Boudoux, et al. Transcranial direct current stimulation in the prophylactic treatment of migraine based on interictal visual cortex excitability abnormalities: a pilot randomized controlled trial. J Neurol Sci. 2015, 349(1/2): 33-39.
[27]
MA Nitsche, W Paulus. Sustained excitability elevations induced by transcranial DC motor cortex stimulation in humans. Neurology. 2001, 57(10): 1899-1901.
[28]
OD Creutzfeldt, GH Fromm, H Kapp. Influence of transcortical d-c currents on cortical neuronal activity. Exp Neurol. 1962, 5: 436-452.
[29]
JM Stilling, O Monchi, F Amoozegar, et al. Transcranial magnetic and direct current stimulation (TMS/tDCS) for the treatment of headache: a systematic review. Headache. 2019, 59(3): 339-357.
[30]
K Meissner, M Fässler, G Rücker, et al. Differential effectiveness of placebo treatments: a systematic review of migraine prophylaxis. JAMA Intern Med. 2013, 173(21): 1941-1951.
[31]
PC Tfelt-Hansen, A Hougaard. Migraine: Differential effects of placebos in migraine clinical trials. Nat Rev Neurol. 2014, 10(1): 10-11.
[32]
Headache Classification Committee of the International Headache Society (IHS). The international classification of headache disorders, 3rd edition (beta version). Cephalalgia. 2013, 33(9): 629-808.
[33]
DW Dodick. A phase-by-phase review of migraine pathophysiology. Headache. 2018, 58: 4-16.
[34]
A May. Understanding migraine as a cycling brain syndrome: reviewing the evidence from functional imaging. Neurol Sci. 2017, 38(Suppl 1): 125-130.
[35]
K Li, LJ Liu, Q Yin, et al. Abnormal rich club organization and impaired correlation between structural and functional connectivity in migraine sufferers. Brain Imaging Behav. 2017, 11(2): 526-540.
[36]
JX Liu, L Zhao, FF Lei, et al. Disrupted resting-state functional connectivity and its changing trend in migraine suffers. Hum Brain Mapp. 2015, 36(5): 1892-1907.
[37]
KG Vetvik, EA MacGregor. Sex differences in the epidemiology, clinical features, and pathophysiology of migraine. Lancet Neurol. 2017, 16(1): 76-87.
[38]
V Raieli, R Pitino, G Giordano, et al. Migraine in a pediatric population: a clinical study in children younger than 7 years of age. Dev Med Child Neurol. 2015, 57(6): 585-588.
[39]
N Maleki, T Kurth, AE Field. Age at menarche and risk of developing migraine or non-migraine headaches by young adulthood: a prospective cohort study. Cephalalgia. 2017, 37(13): 1257-1263.
[40]
JX Liu, L Lan, JY Mu, et al. Genetic contribution of catechol-O-methyltransferase in hippocampal structural and functional changes of female migraine sufferers. Hum Brain Mapp. 2015, 36(5): 1782-1795.
[41]
JX Liu, SH Ma, JY Mu, et al. Integration of white matter network is associated with interindividual differences in psychologically mediated placebo response in migraine patients. Hum Brain Mapp. 2017, 38(10): 5250-5259.
[42]
R Shyti, B de Vries, A van den Maagdenberg. Migraine genes and the relation to gender. Headache. 2011, 51(6): 880-890.
[43]
F Brighina, V Raieli, LM Messina, et al. Non- invasive brain stimulation in pediatric migraine: a perspective from evidence in adult migraine. Front Neurol. 2019, 10: 364.
[44]
A Moore, S Derry, C Eccleston, et al. Expect analgesic failure; pursue analgesic success. BMJ. 2013, 346: f2690.
[45]
FS Collins, H Varmus. A new initiative on precision medicine. N Engl J Med. 2015, 372(9): 793-795.
[46]
A Mailis-Gagnon, I Giannoylis, J Downar, et al. Altered central somatosensory processing in chronic pain patients with "hysterical" anesthesia. Neurology. 2003, 60(9): 1501-1507.
[47]
EA Moulton, R Burstein, S Tully, et al. Interictal dysfunction of a brainstem descending modulatory center in migraine patients. PLoS One. 2008, 3(11): e3799.
[48]
T Schmidt-Wilcke, S Gänssbauer, T Neuner, et al. Subtle grey matter changes between migraine patients and healthy controls. Cephalalgia. 2008, 28(1): 1-4.
[49]
R Burstein, M Jakubowski, E Garcia-Nicas, et al. Thalamic sensitization transforms localized pain into widespread allodynia. Ann Neurol. 2010, 68(1): 81-91.
[50]
A Russo, A Tessitore, F Esposito, et al. Pain processing in patients with migraine: an event-related fMRI study during trigeminal nociceptive stimulation. J Neurol. 2012, 259(9): 1903-1912.
[51]
A Tessitore, A Russo, F Esposito, et al. Interictal cortical reorganization in episodic migraine without aura: an event-related fMRI study during parametric trigeminal nociceptive stimulation. Neurol Sci. 2011, 32(Suppl 1): S165-S167.
[52]
N Maleki, L Becerra, J Brawn, et al. Concurrent functional and structural cortical alterations in migraine. Cephalalgia. 2012, 32(8): 607-620.
[53]
A Russo, M Silvestro, A Tessitore, et al. Advances in migraine neuroimaging and clinical utility: from the MRI to the bedside. Expert Rev Neurother. 2018, 18(7): 533-544.
[54]
NK Logothetis, J Pauls, M Augath, et al. Neurophysiological investigation of the basis of the fMRI signal. Nature. 2001, 412(6843): 150-157.
[55]
ZJ Li, L Lan, F Zeng, et al. The altered right frontoparietal network functional connectivity in migraine and the modulation effect of treatment. Cephalalgia. 2017, 37(2): 161-176.
[56]
ZJ Li, ML Liu, L Lan, et al. Altered periaqueductal gray resting state functional connectivity in migraine and the modulation effect of treatment. Sci Rep. 2016, 6: 20298.
[57]
JY Mu, T Chen, SL Quan, et al. Neuroimaging features of whole-brain functional connectivity predict attack frequency of migraine. Hum Brain Mapp. 2019, .
DOI
[58]
L Hu, GD Iannetti. Issues in pain prediction—beyond pain and gain. Trends Neurosci. 2016, 39(10): 640-642.
[59]
The American College of Radiology. ACR-ASNR-SPR practice guideline for the performance and interpretation of magnetic resonance imaging (MRI) of the brain. 2013.
[60]
JX Liu, JY Mu, QQ Liu, et al. Brain structural properties predict psychologically mediated hypoalgesia in an 8-week sham acupuncture treatment for migraine. Hum Brain Mapp. 2017, 38(9): 4386-4397.
[61]
D Le Bihan, JF Mangin, C Poupon, et al. Diffusion tensor imaging: Concepts and applications. J Magn Reson Imaging. 2001, 13(4): 534-546.
[62]
JL Zhang, YL Wu, JJ Su, et al. Assessment of gray and white matter structural alterations in migraineurs without aura. J Headache Pain. 2017, 18(1): 74.
[63]
CD Chong, TJ Schwedt. Migraine affects white-matter tract integrity: a diffusion-tensor imaging study. Cephalalgia. 2015, 35(13): 1162-1171.
[64]
TJ Schwedt, CC Chiang, CD Chong, et al. Functional MRI of migraine. Lancet Neurol. 2015, 14(1): 81-91.
[65]
LB Zhang, XJ Lu, YZ Bi, et al. Pavlov’s pain: the effect of classical conditioning on pain perception and its clinical implications. Curr Pain Headache Rep. 2019, 23(3): 19.
[66]
SE Lakhan, M Avramut, SJ Tepper. Structural and functional neuroimaging in migraine: insights from 3 decades of research. Headache. 2013, 53(1): 46-66.
[67]
T Sprenger, D Borsook. Migraine changes the brain: neuroimaging makes its mark. Curr Opin Neurol. 2012, 25(3): 252-262.
[68]
RW Evans, RC Burch, BM Frishberg, et al. Neuroimaging for migraine: the American headache society systematic review and evidence-based guideline. Headache. 2019, .
DOI
[69]
CD Chong, TJ Schwedt, DW Dodick. Migraine: what imaging reveals. Curr Neurol Neurosci Rep. 2016, 16(7): 64.
[70]
N Maleki, RL Gollub. What have we learned from brain functional connectivity studies in migraine headache? Headache. 2016, 56(3): 453-461.
[71]
GA Alexiou, MI Argyropoulou. Neuroimaging in childhood headache: a systematic review. Pediatr Radiol. 2013, 43(7): 777-784.
[72]
ZH Jia, SY Yu. Grey matter alterations in migraine: a systematic review and meta-analysis. Neuroimage Clin. 2017, 14: 130-140.
[73]
A Hougaard, FM Amin, M Ashina. Migraine and structural abnormalities in the brain. Curr Opin Neurol. 2014, 27(3): 309-314.
[74]
M Deen, CE Christensen, A Hougaard, et al. Serotonergic mechanisms in the migraine brain-a systematic review. Cephalalgia. 2017, 37(3): 251-264.
[75]
G Demarquay, F Mauguière. Central nervous system underpinnings of sensory hypersensitivity in migraine: insights from neuroimaging and electrophysiological studies. Headache. 2016, 56(9): 1418-1438.
[76]
CW Jin, K Yuan, LM Zhao, et al. Structural and functional abnormalities in migraine patients without aura. NMR Biomed. 2013, 26(1): 58-64.
[77]
XY Li, L Hu. The role of stress regulation on neural plasticity in pain chronification. Neural Plast. 2016, 2016: 6402942.
[78]
ZG Zhang, L Hu, YS Hung, et al. Gamma-band oscillations in the primary somatosensory cortex—a direct and obligatory correlate of subjective pain intensity. J Neurosci. 2012, 32(22): 7429-7438.
[79]
JX Liu, W Qin, JF Nan, et al. Gender-related differences in the dysfunctional resting networks of migraine suffers. PLoS One. 2011, 6(11): e27049.
[80]
JX Liu, L Zhao, GY Li, et al. Hierarchical alteration of brain structural and functional networks in female migraine sufferers. PLoS One. 2012, 7(12): e51250.
[81]
C Mainero, J Boshyan, N Hadjikhani. Altered functional magnetic resonance imaging resting-state connectivity in periaqueductal gray networks in migraine. Ann Neurol. 2011, 70(5): 838-845.
[82]
TJ Schwedt, L Larson-Prior, RS Coalson, et al. Allodynia and descending pain modulation in migraine: a resting state functional connectivity analysis. Pain Med. 2014, 15(1): 154-165.
[83]
A Tessitore, A Russo, A Giordano, et al. Disrupted default mode network connectivity in migraine without aura. J Headache Pain. 2013, 14: 89.
[84]
T Xue, K Yuan, P Cheng, et al. Alterations of regional spontaneous neuronal activity and corresponding brain circuit changes during resting state in migraine without aura. NMR Biomed. 2013, 26(9): 1051-1058.
[85]
T Xue, K Yuan, L Zhao, et al. Intrinsic brain network abnormalities in migraines without aura revealed in resting-state fMRI. PLoS One. 2012, 7(12): e52927.
[86]
K Yuan, W Qin, P Liu, et al. Reduced fractional anisotropy of corpus callosum modulates inter- hemispheric resting state functional connectivity in migraine patients without aura. PLoS One. 2012, 7(9): e45476.
[87]
K Yuan, L Zhao, P Cheng, et al. Altered structure and resting-state functional connectivity of the basal Ganglia in migraine patients without aura. J Pain. 2013, 14(8): 836-844.
[88]
L Zhao, JX Liu, XL Dong, et al. Alterations in regional homogeneity assessed by fMRI in patients with migraine without aura stratified by disease duration. J Headache Pain. 2013, 14: 85.
[89]
TJ Schwedt, CD Chong. Functional imaging and migraine: new connections? Curr Opin Neurol. 2015, 28(3): 265-270.
[90]
AV Apkarian, MN Baliki, PY Geha. Towards a theory of chronic pain. Prog Neurobiol. 2009, 87(2): 81-97.
[91]
MA Farmer, MN Baliki, AV Apkarian. A dynamic network perspective of chronic pain. Neurosci Lett. 2012, 520(2): 197-203.
[92]
M Maizels, S Aurora, M Heinricher. Beyond neurovascular: migraine as a dysfunctional neurolimbic pain network. Headache. 2012, 52(10): 1553-1565.
[93]
V Nagesh, M Welch, SK Aurora, et al. Is there a brainstem generator of chronic daily headache? J Headache Pain. 2000, 1(2): 67-71.
[94]
LH Schulte, A Allers, A May. Hypothalamus as a mediator of chronic migraine: Evidence from high- resolution fMRI. Neurology. 2017, 88(21): 2011-2016.
[95]
LH Schulte, A May. The migraine generator revisited: continuous scanning of the migraine cycle over 30 days and three spontaneous attacks. Brain. 2016, 139(7): 1987-1993.
[96]
LH Schulte, A May. Of generators, networks and migraine attacks. Curr Opin Neurol. 2017, 30(3): 241-245.
[97]
A May. Diagnosis and clinical features of trigemino- autonomic headaches. Headache. 2013, 53(9): 1470- 1478.
[98]
D Borsook, R Veggeberg, N Erpelding, et al. The Insula: a "hub of activity" in migraine. Neuroscientist. 2016, 22(6): 632-652.
[99]
A May. New insights into headache: an update on functional and structural imaging findings. Nat Rev Neurol. 2009, 5(4): 199-209.
[100]
L Hu, GD Iannetti. Neural indicators of perceptual variability of pain across species. Proc Natl Acad Sci USA. 2019, 116(5): 1782-1791.
[101]
DD Price. Psychological and neural mechanisms of the affective dimension of pain. Science. 2000, 288(5472): 1769-1772.
[102]
JX Liu, L Lan, GY Li, et al. Migraine-related gray matter and white matter changes at a 1-year follow-up evaluation. J Pain. 2013, 14(12): 1703-1708.
[103]
N Schmitz, F Admiraal-Behloul, EB Arkink, et al. Attack frequency and disease duration as indicators for brain damage in migraine. Headache. 2008, 48(7): 1044-1055.
[104]
DM Ferriero, SP Miller. Imaging selective vulnerability in the developing nervous system. J Anat. 2010, 217(4): 429-435.
[105]
JA Markham, JI Koenig. Prenatal stress: role in psychotic and depressive diseases. Psychopharmacology. 2011, 214(1): 89-106.
[106]
ME Bigal, MA Arruda. Migraine in the pediatric population—evolving concepts. Headache. 2010, 50(7): 1130-1143.
[107]
GJ Pandina, S Ness, E Polverejan, et al. Cognitive effects of topiramate in migraine patients aged 12 through 17 years. Pediatr Neurol. 2010, 42(3): 187-195.
[108]
WF Stewart, C Wood, ML Reed, et al. Cumulative lifetime migraine incidence in women and men. Cephalalgia. 2008, 28(11): 1170-1178.
[109]
DC Buse, AN Manack, KM Fanning, et al. Chronic migraine prevalence, disability, and sociodemographic factors: results from the American Migraine Prevalence and Prevention Study. Headache. 2012, 52(10): 1456-1470.
[110]
EA MacGregor, A Hackshaw. Prevalence of migraine on each day of the natural menstrual cycle. Neurology. 2004, 63(2): 351-353.
[111]
JM Pavlovic, D Akcali, H Bolay, et al. Sex-related influences in migraine. J Neurosci Res. 2017, 95(1-2): 587-593.
[112]
B de Vries, RR Frants, MD Ferrari, et al. Molecular genetics of migraine. Hum Genet. 2009, 126(1): 115-132.
[113]
M Wessman, M Kallela, MA Kaunisto, et al. A susceptibility locus for migraine with aura, on chromosome 4q24. Am J Hum Genet. 2002, 70(3): 652-662.
[114]
V Anttila, M Kallela, G Oswell, et al. Trait components provide tools to dissect the genetic susceptibility of migraine. Am J Hum Genet. 2006, 79(1): 85-99.
[115]
DR Nyholt, RP Curtain, LR Griffiths. Familial typical migraine: significant linkage and localization of a gene to Xq24-28. Hum Genet. 2000, 107(1): 18-23.
[116]
T Wieser, J Pascual, A Oterino, et al. A novel locus for familial migraine on Xp22. Headache. 2010, 50(6): 955-962.
[117]
B Bayerer, J Engelbergs, I Savidou, et al. Single nucleotide polymorphisms of the serotonin transporter gene in migraine—an association study. Headache. 2010, 50(2): 319-322.
[118]
AN Liu, S Menon, NJ Colson, et al. Analysis of the MTHFR C677T variant with migraine phenotypes. BMC Res Notes. 2010, 3: 213.
[119]
A Tammimäki, PT Männistö. Catechol-O- methyltransferase gene polymorphism and chronic human pain: a systematic review and meta-analysis. Pharmacogenet Genomics. 2012, 22(9): 673-691.
[120]
S Cargnin, F Magnani, M Viana, et al. An opposite- direction modulation of the COMT Val158Met polymorphism on the clinical response to intrathecal morphine and triptans. J Pain. 2013, 14(10): 1097-1106.
[121]
E Vachon-Presseau, M Roy, MO Martel, et al. The stress model of chronic pain: evidence from basal cortisol and hippocampal structure and function in humans. Brain. 2013, 136(3): 815-827.
[122]
G Cosentino, B Fierro, S Vigneri, et al. Cyclical changes of cortical excitability and metaplasticity in migraine: evidence from a repetitive transcranial magnetic stimulation study. Pain. 2014, 155(6): 1070-1078.
[123]
A Antal, N Kriener, N Lang, et al. Cathodal transcranial direct current stimulation of the visual cortex in the prophylactic treatment of migraine. Cephalalgia. 2011, 31(7): 820-828.
[124]
M Hallett. Transcranial magnetic stimulation: a primer. Neuron. 2007, 55(2): 187-199.
[125]
F Brighina, A Piazza, G Vitello, et al. rTMS of the prefrontal cortex in the treatment of chronic migraine: a pilot study. J Neurol Sci. 2004, 227(1): 67-71.
[126]
M Teepker, J Hötzel, N Timmesfeld, et al. Low- frequency rTMS of the vertex in the prophylactic treatment of migraine. Cephalalgia. 2010, 30(2): 137-144.
[127]
F Brigo, M Storti, R Nardone, et al. Transcranial magnetic stimulation of visual cortex in migraine patients: a systematic review with meta-analysis. J Headache Pain. 2012, 13(5): 339-349.
[128]
S Zaghi, N Heine, F Fregni. Brain stimulation for the treatment of pain: a review of costs, clinical effects, and mechanisms of treatment for three different central neuromodulatory approaches. J Pain Manag. 2009, 2(3): 339-352.
[129]
SM Andrade, REL de Brito Aranha, EA de Oliveira, et al. Transcranial direct current stimulation over the primary motor vs prefrontal cortex in refractory chronic migraine: a pilot randomized controlled trial. J Neurol Sci. 2017, 378: 225-232.
[130]
AF Dasilva, ME Mendonca, S Zaghi, et al. tDCS- induced analgesia and electrical fields in pain-related neural networks in chronic migraine. Headache. 2012, 52(8): 1283-1295.
[131]
A Przeklasa-Muszyńska, M Kocot-Kępska, J Dobrogowski, et al. Transcranial direct current stimulation (tDCS) and its influence on analgesics effectiveness in patients suffering from migraine headache. Pharmacol Rep. 2017, 69(4): 714-721.
[132]
A Viganò, TS D'Elia, SL Sava, et al. Transcranial Direct Current Stimulation (tDCS) of the visual cortex: a proof-of-concept study based on interictal electrophysiological abnormalities in migraine. J Headache Pain. 2013, 14: 23.
[133]
P Auvichayapat, T Janyacharoen, A Rotenberg, et al. Migraine prophylaxis by anodal transcranial direct current stimulation, a randomized, placebo-controlled trial. J Med Assoc Thai. 2012, 95(8): 1003-1012.
[134]
JG Speciali, M Peres, ME Bigal. Migraine treatment and placebo effect. Expert Rev Neurother. 2010, 10(3): 413-419.
[135]
L Colloca, F Benedetti. Placebo analgesia induced by social observational learning. Pain. 2009, 144(1/2): 28-34.
[136]
F Eippert, J Finsterbusch, U Bingel, et al. Direct evidence for spinal cord involvement in placebo analgesia. Science. 2009, 326(5951): 404.
[137]
C Büchel, S Geuter, C Sprenger, et al. Placebo analgesia: a predictive coding perspective. Neuron. 2014, 81(6): 1223-1239.
[138]
F Benedetti, M Amanzio, S Vighetti, et al. The biochemical and neuroendocrine bases of the hyperalgesic nocebo effect. J Neurosci. 2006, 26(46): 12014-12022.
[139]
JK Zubieta. Placebo effects mediated by endogenous opioid activity on -opioid receptors. J Neurosci. 2005, 25(34): 7754-7762.
[140]
DJ Scott, CS Stohler, CM Egnatuk, et al. Individual differences in reward responding explain placebo- induced expectations and effects. Neuron. 2007, 55(2): 325-336.
[141]
F Antonaci, P Chimento, HC Diener, et al. Lessons from placebo effects in migraine treatment. J Headache Pain. 2007, 8(1): 63-66.
[142]
AT Drysdale, L Grosenick, J Downar, et al. Resting- state connectivity biomarkers define neurophysiological subtypes of depression. Nat Med. 2017, 23(1): 28-38.
[143]
TD Wager, CW Woo. Imaging biomarkers and biotypes for depression. Nat Med. 2017, 23(1): 16-17.
[144]
M Barad, JA Sturgeon, S Fish, et al. Response to BotulinumtoxinA in a migraine cohort with multiple comorbidities and widespread pain. Reg Anesth Pain Med. 2019, 44(6): 660-668.
[145]
F Parrales Bravo, AA del Barrio García, MM Gallego, et al. Prediction of patient’s response to OnabotulinumtoxinA treatment for migraine. Heliyon. 2019, 5(2): e01043.
[146]
LB Kisler, I Weissman-Fogel, RC Coghill, et al. Individualization of migraine prevention. Clin J Pain. 2019, 35(9): 753-765.
[147]
JX Liu, JY Mu, T Chen, et al. White matter tract microstructure of the mPFC-amygdala predicts interindividual differences in placebo response related to treatment in migraine patients. Hum Brain Mapp. 2019, 40(1): 284-292.
[148]
AB Gago-Veiga, J Pagán, K Henares, et al. To what extent are patients with migraine able to predict attacks? J Pain Res. 2018, 11: 2083-2094.
[149]
RB Lipton, S Munjal, DC Buse, et al. Predicting inadequate response to acute migraine medication: results from the American migraine prevalence and prevention (AMPP) study. Headache. 2016, 56(10): 1635-1648.
[150]
J Pagán, MI De Orbe, A Gago, et al. Robust and accurate modeling approaches for migraine per-patient prediction from ambulatory data. Sensors. 2015, 15(7): 15419-15442.
[151]
WB Young, KC Bradley, MW Anjum, et al. Duloxetine prophylaxis for episodic migraine in persons without depression: a prospective study. Headache. 2013, 53(9): 1430-1437.
[152]
M Ishii, Y Sakairi, H Hara, et al. Negative predictors of clinical response to triptans in patients with migraine. Neurol Sci. 2012, 33(2): 453-461.
[153]
HJ Maas, N Snelder, M Danhof, et al. Prediction of attack frequency in migraine treatment. Cephalalgia. 2008, 28(8): 847-855.
[154]
AR Artemenko, AL Kurenkov, SS Nikitin, et al. Duloxetine in the treatment of chronic migraine. Zh Nevrol Psikhiatr Im S S Korsakova. 2010, 110(1): 49-54.
[155]
HJ Maas, M Danhof, OED Pasqua. Prediction of headache response in migraine treatment. Cephalalgia. 2006, 26(4): 416-422.
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 06 November 2019
Revised: 08 December 2019
Accepted: 15 December 2019
Published: 18 May 2020
Issue date: December 2019

Copyright

© The authors 2019

Acknowledgements

This work was supported by the National Key Research and Development project (No. 2019YFC1709701), the National Natural Science Foundation of China (No. 81871330, No. 81973962), and the National Natural Foundation for Excellent Youth Fund (No. 81722050).

Rights and permissions

Creative Commons Non Commercial CC BY-NC: This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (http://www.creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access pages (https://us.sagepub.com/en-us/nam/open-access-at-sage)

Return