Introduction
Migraine is common in children and adolescents and can have long-term consequences on educational performance, self-esteem, and cognitive well-being1,2. However, effective, well-tolerated, evidence-based therapies for migraine prevention in children and adolescents remain limited3.
In adults, monoclonal antibodies (mAbs) to calcitonin gene-related peptide (CGRP), or its receptor, have been shown to be efficacious for migraine prevention 4–11, and in episodic cluster headache 12. Although CGRP is expressed in multiple body systems13, trials to date have demonstrated minimal adverse events. The most commonly seen adverse events in adult trials included: injection site reactions, upper respiratory symptoms and constipation. These treatments avoid the need for daily pill taking, and thus may improve adherence, which is known to be low with oral migraine preventives14.
Pediatric and adolescent migraine prevention trials will follow; however, outcome data from these will likely not be available for several years. In the interim, the decision regarding whether to treat pediatric patients with anti-CGRP mAbs will require providers to balance the potential for unknown risks against the potential benefits of the therapy. To bridge the gap until empiric data become available, this manuscript, written by members of the Pediatric & Adolescent Headache special interest group of the American Headache Society, is meant to serve as expert opinion, i.e. class IV evidence, on the use of these antibodies in children and adolescents. This topic was discussed at the Pediatric & Adolescent Headache special interest group (SIG) meeting at the 2018 Annual Scientific Meeting of the American Headache Society, and via email communication to SIG members. Interested members volunteered to collaborate as co-authors on this document. Authors divided into subgroups to draft the various sections of the manuscript. Recommendations were reached by consensus first within the subgroups and then all co-authors had the opportunity to read the entire manuscript so that final recommendations could be reached by consensus. The opinions expressed in this manuscript represent those of the co-authors and do not represent the opinion of the American Headache Society.
What we know about the use of monoclonal antibodies in children and adolescents
Monoclonal antibodies (mAbs) have been used to treat a range of pediatric disorders for two decades. These disorders include multiple sclerosis, autoimmune encephalitis, inflammatory bowel disease, systemic lupus erythematosus, juvenile idiopathic arthritis, oncologic disease, and others15. Monoclonal antibodies require parenteral administration, usually intravenous, subcutaneous, or intramuscular. They are degraded via three major routes: extracellular proteolysis mainly via proteases, target-mediated elimination, and intracellularly15. They do not interact with the cytochrome P450 system16, thus dose adjustments for hepatic and renal impairment are not required.
Pediatric dosing for mAbs is often extrapolated from adult doing based on body surface area (BSA) or weight, and age. When dosed based on weight and age, younger children tend to have faster plasma clearance and thus lower plasma concentrations and peak concentrations15. However, when dosing is based on BSA, younger children may have higher plasma exposures than adults as they have a higher surface area to volume ratio. Therefore, approved pediatric mAb dosing approaches include tiered fixed dosing which are staggered based on weight, weight-adjusted and hybrid dosing17. Pediatric pharmacokinetic studies of anti-CGRP mAbs are planned or ongoing. For estimation, 1mg/kg of IgG is only about 0.1–0.2% of the body’s IgG content (at least in adults), so mAbs used for migraine represent only a small fraction of the body’s total antibodies18. In adult subjects the half-lives of the anti-CGRP mAb range from 21 to 50 days19.
Safety and potential immunogenicity of mAbs relate to antibody type and the proportion of non-human or foreign sequences they contain, whether chimeric (-xi-), chimeric/humanized (-xizu-), humanized (-zu-), or fully human (-u-)16. All 4 of the anti-CGRP mAbs which have been studied for migraine prevention are Immunoglobulin G (IgG) based20, and are not expected to cross the blood brain barrier19. Erenumab is fully human, whereas galcanezumab, fremanezumab, and eptinezumab are humanized antibodies. Fully human and humanized antibodies may reduce the risk of marked immunogenicity, though immune reactions can still occur21. Serious infections are rare even with immunosuppressive mAbs22.
CGRP Physiology and corresponding clinical recommendations regarding monitoring of children and adolescents receiving anti-CGRP mAbs:
CGRP is found primarily in unmyelinated aδ and C sensory nerve fibers, but also has a wide distribution in the central nervous system and in non-neuronal tissues throughout the body19,23. The known physiological roles of CGRP are numerous23,24 and extend far beyond its established role in the pathophysiology of migraine. Given that children and adolescents with migraine appear to have elevated CGRP levels as compared to controls, at least during attacks25, it is unclear whether the anti-CGRP mAbs would reduce CGRP levels to a subnormal level that could impact various physiological processes or whether the magnitude of the reduction would simply restore levels to within the normal physiologic range. If the latter is true, the impact on tissues could be minimal.
Central & Peripheral Nervous System:
CGRP and its receptors are expressed in significant concentrations during postnatal animal brain development26,27 and are likely involved in central nervous system maturation27,28. CGRP may also play a role in nervous system recovery after injury 29,30. Anti-CGRP mAbs may not have a significant impact on central nervous system development (CNS) given that they are unlikely to penetrate the blood-brain-barrier in significant concentrations under physiologic conditions19,31. However, it might be possible that by siphoning off CGRP in the periphery the anti-CGRP mAbs could lead to a central reduction in CGRP through a “sink phenomenon”, as has been postulated with other peripherally-acting mAbs” 32,33. If peripheral binding of CGRP by monoclonal antibodies is capable of reducing central CGRP levels, there could potentially be an effect on brain development.
Clinical Recommendation: Until more data are available we recommend against use in children or adolescents with a potentially compromised blood-brain barrier (e.g. those with recent meningitis, neurosurgery, or central nervous system injury such as stroke) or recent peripheral nerve injury.
Maintenance of Pregnancy and Fetal Development:
CGRP appears to play a key role in utero-placental functions34,35, and vascular adaptations to pregnancy34–36. In human pregnancies complicated by pre-eclampsia, the normal CGRP-induced vascular relaxation is impaired37. IgG type antibodies cross the human placenta38. Maternal-fetal IgG transfer is limited before the 16th week of gestation, and rises rapidly after week 2239, hence anti-CGRP antibodies could impact the developing fetus particularly in the second and third trimester. Pregnant rats exposed to a CGRP antagonist had pups with restricted growth or fetal demise34. CGRP receptor component knockout mice experience hydrops fetalis and die in utero36. Given that few pregnant women were exposed to anti-CGRP mAbs during the adult trials, data on human pregnancies are minimal.
Clinical Recommendation: Based on the available data at this time, anti-CGRP mAbs should not be prescribed to adolescent or adult females known to be pregnant, breastfeeding, or planning to become pregnant within the next 6 months. Clinicians should counsel female patients who have reached menarche about the possible teratogenic effects and other possible negative pregnancy outcomes that could result from blocking the CGRP pathway, and continue to discuss pregnancy avoidance during the course of care. Clinicians should consider pregnancy testing if it is their usual practice to do so prior to prescribing migraine preventives which are known to be teratogenic40,41.
Bone:
In animal models, CGRP has been implicated in stimulating bone formation and ossification42–45. Mouse models in which the α-CGRP gene is knocked out have yielded animals with decreased bone mass43. CGRP monoclonal antibodies could theoretically impact linear growth and/or bone mass if CGRP does play a role in bone development in humans. However, at the present time we are not aware of human data on this topic. This may be relevant throughout adolescence, as 40 to 60% of adult bone mass is acquired during that time. Approximately 25% of adult bone mass is acquired during peak height velocity growth46, which typically occurs at 12.5 years for girls and 14 years for boys47. By age 18, 90% of peak bone mass has been attained48.
Clinical Recommendation: Based on the available data at this time, anti-CGRP mAbs should be used with caution in children and adolescents with known bone diseases or significant osteopenia. Clinicians should carefully monitor height and linear growth in children and adolescents receiving anti-CGRP mAbs, and consider waiting to prescribe until the child is post-pubertal. Vitamin D level optimization would be a relatively non-invasive way to support healthy bone growth.
Metabolism:
There is evidence from animal models that CGRP modulates glucose-stimulated insulin release. As compared to wild-type mice, α-CGRP knockout mice fed a high-fat diet for several weeks had more favorable glucose handling and insulin sensitivity and decreased body weight49. Both animal50 and human51 data suggest that obesity may be associated with elevated CGRP levels. If CGRP is involved in human weight regulation, then anti-CGRP monoclonal antibodies may lead to decreased weight and thus baseline body mass index (BMI) of the child or adolescent might be a consideration. However, a significant effect on body weight has not been reported in adult migraine trials to date. Another consideration is CGRP expression in the anterior pituitary. The pituitary gland is outside the blood-brain-barrier52,53, and anti-CGRP mAbs could theoretically affect pituitary function13, such as by reducing growth hormone secretion54.
Clinical Recommendations: Monitor weight and BMI in children and adolescents receiving anti-CGRP mAbs. While there is no evidence that pituitary hormones need to be monitored in all children and adolescents receiving these antibodies, if suggestive symptoms arise, consider laboratory testing to evaluate pituitary function.
Immune System:
CGRP plays a role in the neuro-immune axis, with CGRP-containing nerve fibers located in the thymus, bone marrow, spleen, skin, lymph nodes, lungs and gastrointestinal tract55. CGRP receptors are present on lymphocytes, macrophages, mast cells and dendritic cells, and, in turn, these cells can synthesize CGRP55. CGRP exerts a variety of pro- and anti-inflammatory actions, and is hypothesized to exert nervous system control over immune function55. Reassuringly, adult participants in trials to date have not developed serious infections.
Clinical Recommendation: Consider avoiding anti-CGRP mAbs in children and adolescents with known immunodeficiency or who are receiving immunosuppressive medications, and cease therapy if serious or recurrent infections develop.
Cardiovascular system:
CGRP is a potent vasodilator, and has a significant presence in the cardiovascular system and in perivascular nerves23. Adult studies have not found any significant effect of anti-CGRP mAbs on exercise in patients with stable angina56, nor on blood pressure, even with concomitant sumatriptan use57. However, open questions remain about the cardiovascular effects of blocking CGRP58.
Clinical Recommendations: Consider avoiding use in those with structural heart defects, cardiomyopathy, pulmonary hypertension, coronary artery disease (from conditions such as Kawasaki’s), and history of or significant risk factors for stroke.
Recommendations on when to consider clinical use of anti-CGRP mAbs in children and adolescents with migraine:
Given the relative unknowns, until studies in children and adolescents are completed, anti-CGRP mAbs should be considered primarily for post-pubertal adolescents experiencing relatively frequent migraine (i.e. ≥8 headache days per month) with moderate or severe migraine-related disability (as measured by PedMIDAS or other validated instrument). In the event that a younger child has severe chronic migraine that has proven refractory to multiple migraine preventive trials, it would be reasonable to consider anti-CGRP mAb therapy with careful monitoring as described above.
In order to justify taking on the possible risks of the anti-CGRP mAbs in this population, patients should have had adequate trials of, or contraindications to, established migraine preventive therapies. How many preventives should be tried first is a matter of clinical judgement and the authors of this manuscript had varied recommendations between two and three prior preventives. Some insurance companies may require patients to have trialed at least two prescription preventive medications that are deemed to have Level A evidence for migraine prevention in adults59. Given that the Childhood and Adolescent Migraine Prevention (CHAMP) trial demonstrated similar rates of improvement in children and adolescents treated with placebo as with prescription preventives60, we recommend that in these age groups the options for previous preventive treatment trials be expanded beyond prescription preventive medications to include, though not require, trial of cognitive behavioral therapy, which is well-supported by evidence61–63, as well as treatments that have fewer side effects such as neuromodulation devices64 and supplements (riboflavin, Coenzyme Q10, magnesium, and melatonin)65–73.
Treatment trials of anti-CGRP mAbs in adults have demonstrated efficacy within the first two months of treatment, with therapeutic gains remaining stable over 6 months74,75. Open label extension trials have demonstrated some gains at the one-year mark74. Previous pediatric migraine studies have demonstrated that patients who initially improve with treatment are likely to maintain this improvement76,77. The natural course of migraine may be more easily improved in the developing brain78, potentially allowing for shorter treatment duration. Given the above, and the unknowns of long-term use of the anti-CGRP mAbs particularly in the developing patient, we recommend an initial two-month trial at the lowest available dose. If no improvement is seen, a higher dose may then be trialed for another two months. If there is still no benefit with the higher dose, or it is not tolerated, the medication should be stopped. If improvement is seen, we recommend continuing the medication in an attempt to reach a patient-centered overall treatment goal of fewer than one headache per week79. If this goal is attained, the patient can be continued on the medication for another few months and then efforts should be made to wean off the medication80.
Recommendations regarding participant selection for anti-CGRP mAb trials in pediatric and adolescent migraine
In the past 15 years, migraine preventive trials in children and adolescents have demonstrated that a substantial proportion of children and adolescents assigned to the placebo arm have a clinically meaningful improvement in headache frequency3,81–83. Placebo response has a number of potential biological and psychological mechanisms and is an important area of scientific investigation in its own right84–87, and knowledge of its potential magnitude is important in designing clinical efficacy trials. In the case of pediatric and adolescent trials of anti-CGRP mAbs it is possible that placebo response rates could be even higher than what has been seen in oral preventive trials as work in adults has demonstrated that placebo response rates to injections are higher than to oral medications88–90. In addition, as at least one anti-CGRP mAb has already received US FDA approval for adults, the placebo response may be increased by participants’ high expectation of benefit91.
Novel approaches to trial design for pediatric and adolescent migraine prevention are needed in order to allow separation of placebo from active treatment, if indeed there is biologic efficacy of anti-CGRP monoclonal antibodies in these age groups above that which is provided by placebo. Trial design options include:
Single-blind placebo run-in: i.e. all participants initially receive placebo and then only those who do not respond are randomized. In pediatric and adolescent triptan trials, those in which subjects were given a single-blind placebo run-in treatment and only non-responders were randomized, were successful in demonstrating efficacy92–94. This design has been used in adult migraine research95, and is being implemented in at least one pediatric migraine preventive trial96. It enriches for participants who are either more refractory or those who have a lower “expectation” of response. Though the optimal duration of placebo run-in for an injection-based therapy is not known, data showing trajectory of improvement from recent pediatric trials would suggest that 8-weeks would be adequate at least for oral therapies 77,97.
Crossover design: i.e. the same participants are alternately treated with both placebo and the experimental treatment. This has the advantage of using participants as their own controls. However, data from CHAMP demonstrated that benefits from preventive therapy can persist for over 12 months 97, making crossover designs impractical for pediatric and adolescent migraine prevention studies.
Limit enrollment to participants who have not responded to previous trials of migraine preventive therapies: Adult studies of anti-CGRP antibodies have demonstrated a larger therapeutic gain (i.e. difference between active and placebo) among participants who had been refractory to ≤4 prior preventive medications98,99. This was driven predominantly by a reduced placebo response, which may be due to a lower expectation of response in this group. From a practical standpoint, previous preventive trials are likely to be required by insurance companies before approval is given for more expensive agents. However, results would not necessarily generalize to all children and adolescents with migraine and may therefore limit FDA-approval to patients who have previously failed other preventives.
Regardless of which study design is chosen, we recommend that children and adolescents with continuous headache not be excluded, as these are the patients most in need of novel therapeutics100. Primary and secondary endpoints should reflect those recommended in the International Headache Society’s guideline for controlled trials101, and should be collected using the NIH’s recommended Common Data Elements102. Monitoring should mirror what was done in adult trials, with the addition of monitoring growth. Given that this is a novel therapeutic class in humans, we recommend consideration of long-term outcome registries for those who are exposed to these treatments as children and adolescents103.
Use of anti-CGRP mAbs in children and adolescents with other headache disorders
Cluster Headache
Cluster headache (CH) 104 is a severe, relatively rare primary headache 105 disorder106. CH clearly occurs in children and adolescents 107–109; though clinically it seems less common than in adults, although this has not been systematically tested.
The rationale for the use of anti-CGRP pathway monoclonal antibodies in CH comes from supportive basic science and three lines of experimental medicine. CGRP is found in the trigeminovascular system 110,111, which is part of the basis for its role in migraine 112. CGRP is released into the cranial circulation during acute spontaneous 113 and nitroglycerin-induced 114 CH. Moreover, CGRP can trigger CH when patients are in bout 115, although not when they are out of bout 116, indicating that both peripheral and central mechanisms may be involved in CH pathophysiology. Most recently results of a randomized placebo-controlled trial in adults with episodic cluster headache demonstrated that galcanezumab was superior to placebo in reducing mean weekly cluster attacks. Some three-quarters of patients treated with galcanezumab had a ≥ 50% reduction in attacks 12.
The relative rarity of CH in adolescents will make a well-powered placebo controlled trial difficult to execute. However, the inherent pain severity described in this population argues for its use in this age group based on the adult study. If anti-CGRP mAbs are approved for CH in adults, one could argue for allowance of use in children and adolescents with CH followed by a registry.
New Daily Persistent Headache (NDPH)
New daily persistent headache (NDPH) 104 seems to be more common in children and adolescents as compared to adults117. There is a paucity of evidence to guide treatment of NDPH in adults, children and adolescents118, thus treatments are typically geared toward the phenotype (migraine versus tension-type)118,119. Given the known efficacy of anti-CGRP monoclonal antibodies in chronic migraine9–11, it would be reasonable to treat NDPH of a migrainous phenotype with these agents as well.
Persistent headache attributed to traumatic injury to the head
Chronic post-traumatic headache (PTH) 104 is a common secondary headache seen in the pediatric population120. CGRP has been reported to be released in PTH121, and found to be elevated in the trigeminal nucleus caudalis of those who have suffered repeated head injuries122. While PTH is a secondary headache, its phenotype often resembles that of migraine, and there may be overlapping pathophysiology. As with NDPH, there are no studies to date looking at anti-CGRP monoclonal antibodies for the treatment of persistent PTH. However, for those patients with persistent PTH of a migrainous phenotype who have not responded to conventional therapies, it may be reasonable to try anti-CGRP monoclonal antibody treatment.
Access to anti-CGRP monoclonal antibodies
The prevalence of migraine, and chronic migraine, is higher among children from lower socioeconomic backgrounds compared to their more affluent peers123. Similarly, adolescents with a household income of < $22,500 per year have a migraine prevalence nearly double that of adolescents with a household income > $90,000 per year 124. Yet children and adolescents from lower socioeconomic backgrounds are also more likely to be uninsured or under-insured. In one study, 22% of individuals in working poor families were uninsured compared to only 5% in moderate to affluent families125. Access and quality of healthcare is often suboptimal for uninsured or underinsured pediatric patients126. Many of these children rely on community clinics and county or city hospitals for their migraine management; these facilities may not have child neurologists or pediatric headache specialists127.
Given that migraine is a treatable cause of missed school and impaired performance at school123,128,129, it is imperative that clinicians advocate for equality of access to migraine therapeutics for children and adolescents, whether the intervention is a prescribed medication, a device, or non-pharmacologic therapy. The passage of the Best Pharmaceuticals Act for Children (BPCA) and the Pediatric Research Equity ACT (PREA) has resulted in increased evaluation and FDA labeling of drugs in children through the pharmaceutical industry and through government sponsored trials130. However, pediatric labeling still exists for fewer than 50% of products131. This results in “off-label” drug use being common practice in the care of pediatric patients. The absence of FDA labeling for a specific age group or for a specific disorder does not imply that the drug’s use is improper for that age or disorder, but that the evidence required by law to allow inclusion of that group in the label has not yet been obtained130. Well-designed trials of the anti-CGRP monoclonal antibodies for migraine prevention in children and adolescents are needed not only to allow practitioners to make informed therapeutic decisions, but also to allow better access to, and insurance coverage of, these medications in this age group. In the meantime, clinicians will need to advocate for patients who need access to these therapies today, despite the high price-tag.
Conclusions
Anti-CGRP mAbs have been shown to be effective for migraine prevention in adults and have not raised major safety issues, though long-term safety data are not yet available. Pediatric and adolescent trials of anti-CGRP mAbs should be designed to maximize the chances of determining efficacy in these age groups, and should focus on those who have not been successful with current multidisciplinary care. In the interim, the use of anti-CGRP mAbs for the treatment of headache disorders in children and adolescents may be considered in appropriate cases, but should be done with close follow-up and attention to patient characteristics such as age, pubertal state, and medical comorbidities.
Table 1.
Suggested indications, contraindications and monitoring for use of CGRP monoclonal antibodies in children and adolescents with migraine
| Indications | Contraindications | Monitoring |
|---|---|---|
|
|
|
BMI=body-mass index
Acknowledgments
Financial disclosures:
CS: Has received grant support from Pfizer and Amgen.
JVP: reports speaking fees for Amgen and Novartis, and consulting for Healint.
SLO: Receives royalties from Cambridge University Press
CO: None
WQ: None
IP: none
AMLB: Is on the advisory board for Teva, Novartis and Allergan and the speakers’ bureau for Aralez.
CM: None
JG: Speaker’s bureau for Supernus and has received travel funds from the American Academy of Neurology.
MCV: Has received travel funds from the American Academy of Neurology and honoraria for authorship from Merck.
SH: none
MC: none
CC: none
RR: none
RHF: Receives funding from the National Institutes of Health.
AB: none
MY: Has received consulting fees from Amgen, Impax, Upsher-Smith, and Avanir
ADH: Alder, Amgen, Curelator, Depomed, Electrocore, Impax, Lilly, Teva and Upsher-Smith
SWP: Receives funding from the NIH
PJG: PJG reports grants and personal fees from Amgen and Eli-Lilly and Company, and personal fees from Alder Biopharmaceuticals, Allergan, Autonomic Technologies Inc., Dr. Reddy’s Laboratories, Electrocore LLC, eNeura, Novartis, Scion, Teva Pharmaceuticals, and Trigemina Inc., and personal fees from MedicoLegal work, Journal Watch, Up-to-Date, Oxford University Press, Massachusetts Medical Society, and Wolters Kluwer; and a patent Magnetic stimulation for headache assigned to eNeura without fee.
AAG: Has received consulting fees from Zosano, Eli Lilly, Impax and Biohaven. She has received honoraria from UpToDate (for authorship) and JAMA Neurology (as an associate editor). eNeura provides consulting payments for work done by Dr. Gelfand to the UCSF Pediatric Headache program. She receives personal compensation for medical-legal consulting. Her spouse receives consulting fees from Genentech, research support from Genentech, Quest Diagnostics and MedDay, and personal compensation for medical-legal consulting.
Funding:
Dr. Szperka receives research funding from the National Institute of Health (K23NS102521). There was no specific funding for this project.
Abbreviations:
- CGRP
calcitonin gene-related peptide
- mAb
monoclonal antibody, anti-CGRP
- mAb
a monoclonal antibody that targets CGRP or its receptor
- CNS
central nervous system
- BSA
body surface area
- BMI
body mass index
- NDPH
New Daily Persistent Headache
- CH
Cluster Headache
References:
- 1.Fuh JL, Wang SJ, Lu SR, Liao YC, Chen SP, Yang CY. Headache disability among adolescents: a student population-based study. Headache. 2010;50(2):210–218. [DOI] [PubMed] [Google Scholar]
- 2.Rocha-Filho PA, Santos PV. Headaches, quality of life, and academic performance in schoolchildren and adolescents. Headache. 2014;54(7):1194–1202. [DOI] [PubMed] [Google Scholar]
- 3.El-Chammas K, Keyes J, Thompson N, Vijayakumar J, Becher D, Jackson JL. Pharmacologic treatment of pediatric headaches: a meta-analysis. JAMA pediatrics. 2013;167(3):250–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Dodick DW, Ashina M, Brandes JL, et al. ARISE: A Phase 3 randomized trial of erenumab for episodic migraine. Cephalalgia : an international journal of headache. 2018;38(6):1026–1037. [DOI] [PubMed] [Google Scholar]
- 5.Goadsby PJ, Reuter U, Hallstrom Y, et al. A Controlled Trial of Erenumab for Episodic Migraine. The New England journal of medicine. 2017;377(22):2123–2132. [DOI] [PubMed] [Google Scholar]
- 6.Stauffer VL, Dodick DW, Zhang Q, Carter JN, Ailani J, Conley RR. Evaluation of Galcanezumab for the Prevention of Episodic Migraine: The EVOLVE-1 Randomized Clinical Trial. JAMA Neurol. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dodick DW, Silberstein SD, Bigal ME, et al. Effect of Fremanezumab Compared With Placebo for Prevention of Episodic Migraine: A Randomized Clinical Trial. Jama. 2018;319(19):1999–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Skljarevski V, Matharu M, Millen BA, Ossipov MH, Kim BK, Yang JY. Efficacy and safety of galcanezumab for the prevention of episodic migraine: Results of the EVOLVE-2 Phase 3 randomized controlled clinical trial. Cephalalgia : an international journal of headache. 2018:333102418779543. [DOI] [PubMed] [Google Scholar]
- 9.Silberstein SD, Dodick DW, Bigal ME, et al. Fremanezumab for the Preventive Treatment of Chronic Migraine. The New England journal of medicine. 2017;377(22):2113–2122. [DOI] [PubMed] [Google Scholar]
- 10.Tepper S, Ashina M, Reuter U, et al. Safety and efficacy of erenumab for preventive treatment of chronic migraine: a randomised, double-blind, placebo-controlled phase 2 trial. The Lancet Neurology. 2017;16(6):425–434. [DOI] [PubMed] [Google Scholar]
- 11.Bigal ME, Edvinsson L, Rapoport AM, et al. Safety, tolerability, and efficacy of TEV-48125 for preventive treatment of chronic migraine: a multicentre, randomised, double-blind, placebo-controlled, phase 2b study. The Lancet Neurology. 2015;14(11):1091–1100. [DOI] [PubMed] [Google Scholar]
- 12.Martinez JM, Goadsby PJ, Dodick DW, et al. A Placebo-Controlled Study of Galcanezumab in Patients with Episodic Cluster Headache: from the 8-Week Double-Blind Treatment Phase. Headache. 2018;58:Late-Breaking Abstract. [Google Scholar]
- 13.Deen M, Correnti E, Kamm K, et al. Blocking CGRP in migraine patients - a review of pros and cons. The journal of headache and pain. 2017;18(1):96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Berger A, Bloudek LM, Varon SF, Oster G. Adherence with migraine prophylaxis in clinical practice. Pain practice : the official journal of World Institute of Pain. 2012;12(7):541–549. [DOI] [PubMed] [Google Scholar]
- 15.Malik P, Edginton A. Pediatric physiology in relation to the pharmacokinetics of monoclonal antibodies. Expert opinion on drug metabolism & toxicology. 2018;14(6):585–599. [DOI] [PubMed] [Google Scholar]
- 16.Catapano AL, Papadopoulos N. The safety of therapeutic monoclonal antibodies: implications for cardiovascular disease and targeting the PCSK9 pathway. Atherosclerosis. 2013;228(1):18–28. [DOI] [PubMed] [Google Scholar]
- 17.Zheng S, Gaitonde P, Andrew MA, Gibbs MA, Lesko LJ, Schmidt S. Model-based assessment of dosing strategies in children for monoclonal antibodies exhibiting target-mediated drug disposition. CPT: pharmacometrics & systems pharmacology. 2014;3:e138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ryman JT, Meibohm B. Pharmacokinetics of Monoclonal Antibodies. CPT: pharmacometrics & systems pharmacology. 2017;6(9):576–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Raffaelli B, Reuter U. The Biology of Monoclonal Antibodies: Focus on Calcitonin Gene-Related Peptide for Prophylactic Migraine Therapy. Neurotherapeutics. 2018;15(2):324–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Pellesi L, Guerzoni S, Pini LA. Spotlight on Anti-CGRP Monoclonal Antibodies in Migraine: The Clinical Evidence to Date. Clinical pharmacology in drug development. 2017;6(6):534–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hwang WY, Foote J. Immunogenicity of engineered antibodies. Methods (San Diego, Calif). 2005;36(1):3–10. [DOI] [PubMed] [Google Scholar]
- 22.Williamson EM, Berger JR. Central Nervous System Infections With Immunomodulatory Therapies. Continuum (Minneapolis, Minn). 2015;21(6 Neuroinfectious Disease):1577–1598. [DOI] [PubMed] [Google Scholar]
- 23.Russell FA, King R, Smillie SJ, Kodji X, Brain SD. Calcitonin gene-related peptide: physiology and pathophysiology. Physiological reviews. 2014;94(4):1099–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Poyner DR. Calcitonin gene-related peptide: multiple actions, multiple receptors. Pharmacol Ther. 1992;56(1):23–51. [DOI] [PubMed] [Google Scholar]
- 25.Fan PC, Kuo PH, Chang SH, Lee WT, Wu RM, Chiou LC. Plasma calcitonin gene-related peptide in diagnosing and predicting paediatric migraine. Cephalalgia : an international journal of headache. 2009;29(8):883–890. [DOI] [PubMed] [Google Scholar]
- 26.Terrado J, Gerrikagoitia I, Martinez-Millan L, et al. Expression of the genes for alpha-type and beta-type calcitonin gene-related peptide during postnatal rat brain development. Neuroscience. 1997;80(3):951–970. [DOI] [PubMed] [Google Scholar]
- 27.Dickerson IM, Bussey-Gaborski R, Holt JC, Jordan PM, Luebke AE. Maturation of suprathreshold auditory nerve activity involves cochlear CGRP-receptor complex formation. Physiol Rep. 2016;4(14). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.D’Antoni S, Zambusi L, Codazzi F, et al. Calcitonin gene-related peptide (CGRP) stimulates purkinje cell dendrite growth in culture. Neurochemical research. 2010;35(12):2135–2143. [DOI] [PubMed] [Google Scholar]
- 29.Zhai L, Sakurai T, Kamiyoshi A, et al. Endogenous calcitonin gene-related peptide suppresses ischemic brain injuries and progression of cognitive decline. J Hypertens. 2018;36(4):876–891. [DOI] [PubMed] [Google Scholar]
- 30.Chung AM. Calcitonin gene-related peptide (CGRP): role in peripheral nerve regeneration. Rev Neurosci. 2018;29(4):369–376. [DOI] [PubMed] [Google Scholar]
- 31.Edvinsson L, Warfvinge K. Recognizing the role of CGRP and CGRP receptors in migraine and its treatment. Cephalalgia : an international journal of headache. 2017:333102417736900. [DOI] [PubMed] [Google Scholar]
- 32.DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(15):8850–8855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Henderson SJ, Andersson C, Narwal R, et al. Sustained peripheral depletion of amyloid-beta with a novel form of neprilysin does not affect central levels of amyloid-beta. Brain : a journal of neurology. 2014;137(Pt 2):553–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yallampalli C, Chauhan M, Endsley J, Sathishkumar K. Calcitonin gene related family peptides: importance in normal placental and fetal development. Adv Exp Med Biol. 2014;814:229–240. [DOI] [PubMed] [Google Scholar]
- 35.Yallampalli C, Chauhan M, Thota CS, Kondapaka S, Wimalawansa SJ. Calcitonin gene-related peptide in pregnancy and its emerging receptor heterogeneity. Trends Endocrinol Metab. 2002;13(6):263–269. [DOI] [PubMed] [Google Scholar]
- 36.Dong YL, Reddy DM, Green KE, et al. Calcitonin gene-related peptide (CALCA) is a proangiogenic growth factor in the human placental development. Biol Reprod. 2007;76(5):892–899. [DOI] [PubMed] [Google Scholar]
- 37.Dong YL, Green KE, Vegiragu S, et al. Evidence for decreased calcitonin gene-related peptide (CGRP) receptors and compromised responsiveness to CGRP of fetoplacental vessels in preeclamptic pregnancies. The Journal of clinical endocrinology and metabolism. 2005;90(4):2336–2343. [DOI] [PubMed] [Google Scholar]
- 38.Malek A Role of IgG antibodies in association with placental function and immunologic diseases in human pregnancy. Expert Rev Clin Immunol. 2013;9(3):235–249. [DOI] [PubMed] [Google Scholar]
- 39.Saji F, Samejima Y, Kamiura S, Koyama M. Dynamics of immunoglobulins at the feto-maternal interface. Reviews of reproduction. 1999;4(2):81–89. [DOI] [PubMed] [Google Scholar]
- 40.Alsaad AM, Chaudhry SA, Koren G. First trimester exposure to topiramate and the risk of oral clefts in the offspring: A systematic review and meta-analysis. Reproductive toxicology (Elmsford, NY). 2015;53:45–50. [DOI] [PubMed] [Google Scholar]
- 41.Vajda FJ, O’Brien TJ, Graham J, Lander CM, Eadie MJ. Associations between particular types of fetal malformation and antiepileptic drug exposure in utero. Acta neurologica Scandinavica. 2013;128(4):228–234. [DOI] [PubMed] [Google Scholar]
- 42.Lerner UH, Persson E. Osteotropic effects by the neuropeptides calcitonin gene-related peptide, substance P and vasoactive intestinal peptide. J Musculoskelet Neuronal Interact. 2008;8(2):154–165. [PubMed] [Google Scholar]
- 43.Lerner UH. Deletions of genes encoding calcitonin/alpha-CGRP, amylin and calcitonin receptor have given new and unexpected insights into the function of calcitonin receptors and calcitonin receptor-like receptors in bone. J Musculoskelet Neuronal Interact. 2006;6(1):87–95. [PubMed] [Google Scholar]
- 44.Huebner AK, Keller J, Catala-Lehnen P, et al. The role of calcitonin and alpha-calcitonin gene-related peptide in bone formation. Arch Biochem Biophys. 2008;473(2):210–217. [DOI] [PubMed] [Google Scholar]
- 45.Grassel SG. The role of peripheral nerve fibers and their neurotransmitters in cartilage and bone physiology and pathophysiology. Arthritis Res Ther. 2014;16(6):485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Golden NH, Abrams SA. Optimizing bone health in children and adolescents. Pediatrics. 2014;134(4):e1229–1243. [DOI] [PubMed] [Google Scholar]
- 47.Bailey DA, Martin AD, McKay HA, Whiting S, Mirwald R. Calcium accretion in girls and boys during puberty: a longitudinal analysis. Journal of bone and mineral research : the official journal of the American Society for Bone and Mineral Research. 2000;15(11):2245–2250. [DOI] [PubMed] [Google Scholar]
- 48.Bachrach LK. Acquisition of optimal bone mass in childhood and adolescence. Trends in endocrinology and metabolism: TEM. 2001;12(1):22–28. [DOI] [PubMed] [Google Scholar]
- 49.Walker CS, Li X, Whiting L, et al. Mice lacking the neuropeptide alpha-calcitonin gene-related peptide are protected against diet-induced obesity. Endocrinology. 2010;151(9):4257–4269. [DOI] [PubMed] [Google Scholar]
- 50.Gram DX, Hansen AJ, Wilken M, et al. Plasma calcitonin gene-related peptide is increased prior to obesity, and sensory nerve desensitization by capsaicin improves oral glucose tolerance in obese Zucker rats. Eur J Endocrinol. 2005;153(6):963–969. [DOI] [PubMed] [Google Scholar]
- 51.Zelissen PM, Koppeschaar HP, Lips CJ, Hackeng WH. Calcitonin gene-related peptide in human obesity. Peptides. 1991;12(4):861–863. [DOI] [PubMed] [Google Scholar]
- 52.Eftekhari S, Salvatore CA, Johansson S, Chen TB, Zeng Z, Edvinsson L. Localization of CGRP, CGRP receptor, PACAP and glutamate in trigeminal ganglion. Relation to the blood-brain barrier. Brain research. 2015;1600:93–109. [DOI] [PubMed] [Google Scholar]
- 53.Wimalawansa SJ, el-Kholy AA. Comparative study of distribution and biochemical characterization of brain calcitonin gene-related peptide receptors in five different species. Neuroscience. 1993;54(2):513–519. [DOI] [PubMed] [Google Scholar]
- 54.Nakamura Y, Shimatsu A, Murabe H, Mizuta H, Ihara C, Nakao K. Calcitonin gene-related peptide as a GH secretagogue in human and rat pituitary somatotrophs. Brain research. 1998;807(1–2):203–207. [DOI] [PubMed] [Google Scholar]
- 55.Assas BM, Pennock JI, Miyan JA. Calcitonin gene-related peptide is a key neurotransmitter in the neuro-immune axis. Front Neurosci. 2014;8:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Depre C, Antalik L, Starling A, et al. A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Effect of Erenumab on Exercise Time During a Treadmill Test in Patients With Stable Angina. Headache. 2018;58(5):715–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.de Hoon J, Van Hecken A, Vandermeulen C, et al. Phase 1, randomized, parallel-group, double-blind, placebo-controlled trial to evaluate the effects of erenumab (AMG 334) and concomitant sumatriptan on blood pressure in healthy volunteers. Cephalalgia : an international journal of headache. 2018:333102418776017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.MaassenVanDenBrink A, Meijer J, Villalon CM, Ferrari MD. Wiping Out CGRP: Potential Cardiovascular Risks. Trends in pharmacological sciences. 2016;37(9):779–788. [DOI] [PubMed] [Google Scholar]
- 59.Silberstein SD, Holland S, Freitag F, et al. Evidence-based guideline update: pharmacologic treatment for episodic migraine prevention in adults: report of the Quality Standards Subcommittee of the American Academy of Neurology and the American Headache Society. Neurology. 2012;78(17):1337–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Powers SW, Coffey CS, Chamberlin LA, et al. Trial of Amitriptyline, Topiramate, and Placebo for Pediatric Migraine. The New England journal of medicine. 2017;376(2):115–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Powers SW, Kashikar-Zuck SM, Allen JR, et al. Cognitive behavioral therapy plus amitriptyline for chronic migraine in children and adolescents: a randomized clinical trial. Jama. 2013;310(24):2622–2630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ng QX, Venkatanarayanan N, Kumar L. A Systematic Review and Meta-Analysis of the Efficacy of Cognitive Behavioral Therapy for the Management of Pediatric Migraine. Headache. 2017;57(3):349–362. [DOI] [PubMed] [Google Scholar]
- 63.Eccleston C, Palermo TM, Williams AC, et al. Psychological therapies for the management of chronic and recurrent pain in children and adolescents. The Cochrane database of systematic reviews. 2014(5):Cd003968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Starling AJ, Tepper SJ, Marmura MJ, et al. A multicenter, prospective, single arm, open label, observational study of sTMS for migraine prevention (ESPOUSE Study). Cephalalgia : an international journal of headache. 2018;38(6):1038–1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Schoenen J, Jacquy J, Lenaerts M. Effectiveness of high-dose riboflavin in migraine prophylaxis. A randomized controlled trial. Neurology. 1998;50(2):466–470. [DOI] [PubMed] [Google Scholar]
- 66.Goncalves AL, Martini Ferreira A, Ribeiro RT, Zukerman E, Cipolla-Neto J, Peres MF. Randomised clinical trial comparing melatonin 3 mg, amitriptyline 25 mg and placebo for migraine prevention. Journal of neurology, neurosurgery, and psychiatry. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Peikert A, Wilimzig C, Kohne-Volland R. Prophylaxis of migraine with oral magnesium: results from a prospective, multi-center, placebo-controlled and double-blind randomized study. Cephalalgia : an international journal of headache. 1996;16(4):257–263. [DOI] [PubMed] [Google Scholar]
- 68.Sandor PS, Di Clemente L, Coppola G, et al. Efficacy of coenzyme Q10 in migraine prophylaxis: a randomized controlled trial. Neurology. 2005;64(4):713–715. [DOI] [PubMed] [Google Scholar]
- 69.Irwin SL, Qubty W, Allen IE, Patniyot I, Goadsby PJ, Gelfand AA. Transcranial Magnetic Stimulation for Migraine Prevention in Adolescents: A Pilot Open-Label Study. Headache. 2018;58(5):724–731. [DOI] [PubMed] [Google Scholar]
- 70.Hershey AD, Powers SW, Vockell AL, et al. Coenzyme Q10 deficiency and response to supplementation in pediatric and adolescent migraine. Headache. 2007;47(1):73–80. [DOI] [PubMed] [Google Scholar]
- 71.Slater SK, Nelson TD, Kabbouche MA, et al. A randomized, double-blinded, placebo-controlled, crossover, add-on study of CoEnzyme Q10 in the prevention of pediatric and adolescent migraine. Cephalalgia : an international journal of headache. 2011;31(8):897–905. [DOI] [PubMed] [Google Scholar]
- 72.Wang F, Van Den Eeden SK, Ackerson LM, Salk SE, Reince RH, Elin RJ. Oral magnesium oxide prophylaxis of frequent migrainous headache in children: a randomized, double-blind, placebo-controlled trial. Headache. 2003;43(6):601–610. [DOI] [PubMed] [Google Scholar]
- 73.Orr SL. The Evidence for the Role of Nutraceuticals in the Management of Pediatric Migraine: a Review. Current pain and headache reports. 2018;22(5):37. [DOI] [PubMed] [Google Scholar]
- 74.Ashina M, Dodick D, Goadsby PJ, et al. Erenumab (AMG 334) in episodic migraine: Interim analysis of an ongoing open-label study. Neurology. 2017;89(12):1237–1243. [DOI] [PubMed] [Google Scholar]
- 75.Skljarevski V, Oakes TM, Zhang Q, et al. Effect of Different Doses of Galcanezumab vs Placebo for Episodic Migraine Prevention: A Randomized Clinical Trial. JAMA neurology. 2018;75(2):187–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kabbouche MA, Powers SW, Vockell AL, et al. Outcome of a multidisciplinary approach to pediatric migraine at 1, 2, and 5 years. Headache. 2005;45(10):1298–1303. [DOI] [PubMed] [Google Scholar]
- 77.Kroner JW, Peugh J, Kashikar-Zuck SM, et al. Trajectory of Improvement in Children and Adolescents With Chronic Migraine: Results From the Cognitive-Behavioral Therapy and Amitriptyline Trial. The journal of pain : official journal of the American Pain Society. 2017;18(6):637–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Charles JA, Peterlin BL, Rapoport AM, Linder SL, Kabbouche MA, Sheftell FD. Favorable outcome of early treatment of new onset child and adolescent migraine-implications for disease modification. The journal of headache and pain. 2009;10(4):227–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Szperka CL, Zerafati G, Oakley C, et al. Pediatric Patient and Parent Goals and Preferences for Preventive Headache Treatment. Paper presented at: American Headache Society 59th Annual Scientific Meeting2017; Boston, MA. [Google Scholar]
- 80.Kacperski J Prophylaxis of migraine in children and adolescents. Paediatric drugs. 2015;17(3):217–226. [DOI] [PubMed] [Google Scholar]
- 81.Lewis DW, Winner P, Wasiewski W. The placebo responder rate in children and adolescents. Headache. 2005;45(3):232–239. [DOI] [PubMed] [Google Scholar]
- 82.Powers SW, Coffey CS, Chamberlin LA, et al. Trial of Amitriptyline, Topiramate, and Placebo for Pediatric Migraine. N Engl J Med. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Allergan. A Study Using Botulinum Toxin Type A as Headache Prophylaxis in Adolescents With Chronic Migraine. 2018; https://clinicaltrials.gov/ct2/show/study/NCT01662492. Accessed 7/16/18.
- 84.Kroon Van Diest AM, Ernst MM, Slater S, Powers SW. Similarities and Differences Between Migraine in Children and Adults: Presentation, Disability, and Response to Treatment. Current pain and headache reports. 2017;21(12):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Faria V, Linnman C, Lebel A, Borsook D. Harnessing the placebo effect in pediatric migraine clinic. The Journal of pediatrics. 2014;165(4):659–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Hoekman DR, Zeevenhooven J, van Etten-Jamaludin FS, et al. The Placebo Response in Pediatric Abdominal Pain-Related Functional Gastrointestinal Disorders: A Systematic Review and Meta-Analysis. The Journal of pediatrics. 2017;182:155–163. e157. [DOI] [PubMed] [Google Scholar]
- 87.Kam-Hansen S, Jakubowski M, Kelley JM, et al. Altered placebo and drug labeling changes the outcome of episodic migraine attacks. Science translational medicine. 2014;6(218):218ra215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Macedo A, Farre M, Banos JE. A meta-analysis of the placebo response in acute migraine and how this response may be influenced by some of the characteristics of clinical trials. Eur J Clin Pharmacol. 2006;62(3):161–172. [DOI] [PubMed] [Google Scholar]
- 89.Kaptchuk TJ, Stason WB, Davis RB, et al. Sham device v inert pill: randomised controlled trial of two placebo treatments. BMJ. 2006;332(7538):391–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Meissner K, Fassler M, Rucker G, et al. Differential effectiveness of placebo treatments: a systematic review of migraine prophylaxis. JAMA internal medicine. 2013;173(21):1941–1951. [DOI] [PubMed] [Google Scholar]
- 91.Crum AJ, Leibowitz KA, Verghese A. Making mindset matter. BMJ (Clinical research ed). 2017;356:j674. [DOI] [PubMed] [Google Scholar]
- 92.Ho TW, Pearlman E, Lewis D, et al. Efficacy and tolerability of rizatriptan in pediatric migraineurs: results from a randomized, double-blind, placebo-controlled trial using a novel adaptive enrichment design. Cephalalgia : an international journal of headache. 2012;32(10):750–765. [DOI] [PubMed] [Google Scholar]
- 93.Lewis DW, Winner P, Hershey AD, Wasiewski WW, Adolescent Migraine Steering C. Efficacy of zolmitriptan nasal spray in adolescent migraine. Pediatrics. 2007;120(2):390–396. [DOI] [PubMed] [Google Scholar]
- 94.Derosier FJ, Lewis D, Hershey AD, et al. Randomized trial of sumatriptan and naproxen sodium combination in adolescent migraine. Pediatrics. 2012;129(6):e1411–1420. [DOI] [PubMed] [Google Scholar]
- 95.Mathew NT, Frishberg BM, Gawel M, et al. Botulinum toxin type A (BOTOX) for the prophylactic treatment of chronic daily headache: a randomized, double-blind, placebo-controlled trial. Headache. 2005;45(4):293–307. [DOI] [PubMed] [Google Scholar]
- 96.Gelfand A Melatonin for Adolescent Migraine Prevention Study (MAP). 2018; https://clinicaltrials.gov/ct2/show/NCT03150797?term=children+melatonin&cond=Migraine+Disorders&rank=1. Accessed 7/16/18.
- 97.Hershey A, Powers S, Peugh J, Coffey CS. Discussion of Trajectory of Improvement in CHAMP Trial. In:2018.
- 98.Goadsby PJ, Lanteri-Minet M, Ferrari M, Wen S, Klatt J. Efficacy and Safety of Erenumab in Episodic Migraine Patients with 2–4 Prior Preventive Treatment Failures: Results from the Phase 3b LIBERTY Study. Paper presented at: American Headache Society 60th Annual Scientific Meeting2018; San Francisco, CA. [Google Scholar]
- 99.Aurora SK, Ruff DD, Pearlman EM, Govindan S, Zhang Q. Efficacy of Galcanezumab in Patients Who Failed to Respond to Preventives Previously: Results from EVOLVE-1, EVOLVE-2, and REGAIN Studies. Paper presented at: American Academy of Neurology; April 21–27, 2018, 2018; Los Angeles, CA. [Google Scholar]
- 100.Gelfand AA, Qubty W, Goadsby PJ. Pediatric Migraine Prevention-First, Do No Harm. JAMA neurology. 2017;74(8):893–894. [DOI] [PubMed] [Google Scholar]
- 101.Tfelt-Hansen P, Pascual J, Ramadan N, et al. Guidelines for controlled trials of drugs in migraine: third edition. A guide for investigators. Cephalalgia : an international journal of headache. 2012;32(1):6–38. [DOI] [PubMed] [Google Scholar]
- 102.Oshinsky ML, Tanveer S. Improving research through NINDS Headache Common Data Elements. Cephalalgia : an international journal of headache. 2018:333102418773076. [DOI] [PubMed] [Google Scholar]
- 103.Services USDoHaH, Administration FaD. Guidance for Industry: Premarketing Risk Assessment. In: (CDER) CfDEaR, (CBER) CfBEaR, eds 2005.
- 104.Headache Classification Committee of the International Headache Society (IHS). The International Classification of Headache Disorders, 3rd Edition. Cepahalalgia. 2018;38:1–211. [DOI] [PubMed] [Google Scholar]
- 105.Nesbitt AD, Goadsby PJ . Cluster Headache. British Medical Journal. 2012;344:e2407. [DOI] [PubMed] [Google Scholar]
- 106.Schor LI. Cluster Headache: Investigating severity of pain, suicidality, personal burden, access to effective treatment, and demographics among a large International survey sample. Cephalalgia. 2017;37(1S):172.28880582 [Google Scholar]
- 107.McNabb S, Whitehouse W. Cluster headache-like disorder in childhood. Archives of Disease in Childhood. 1999;81(6):511–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lampl C Childhood-onset cluster headache. Pediatr Neurol. 2002;27:138–140. [DOI] [PubMed] [Google Scholar]
- 109.Mack KJ, Goadsby PJ. Trigeminal Autonomic Cephalalgias in Children and Adolescents- Cluster Headache and Related Conditions. Seminars in Pediatric Neurology. 2016;23:23–26. [DOI] [PubMed] [Google Scholar]
- 110.Edvinsson L, Haanes KA, Warfvinge K, Krause DN. CGRP as the target of new migraine therapies - successful translation from bench to clinic. Nat Rev Neurol. 2018;14(6):338–350. [DOI] [PubMed] [Google Scholar]
- 111.Hoffmann J, May A. Diagnosis, pathophysiology, and management of cluster headache. The Lancet Neurology. 2018;17(1):75–83. [DOI] [PubMed] [Google Scholar]
- 112.Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S. Pathophysiology of Migraine- A disorder of sensory processing. Physiological Reviews. 2017;97:553–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Goadsby PJ, Edvinsson L. Human in vivo evidence for trigeminovascular activation in cluster headache. Brain. 1994;117:427–434. [DOI] [PubMed] [Google Scholar]
- 114.Fanciullacci M, Alessandri M, Figini M, Geppetti P, Michelacci S. Increase in plasma calcitonin gene-related peptide from extracerebral circulation during nitroglycerin-induced cluster headache attack. Pain. 1995;60:119–123. [DOI] [PubMed] [Google Scholar]
- 115.Vollesen ALH, Snoer A, Beske RP, et al. Effect of Infusion of Calcitonin Gene-Related Peptide on Cluster Headache Attacks: A Randomized Clinical Trial. JAMA Neurol. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Gelfand AA, Goadsby PJ. Cluster Headache and Calcitonin Gene-Related Peptide-More on Quantum Therapeutics in Headache Medicine. JAMA Neurol. 2018. [DOI] [PubMed] [Google Scholar]
- 117.Grengs LR, Mack KJ. New Daily Persistent Headache Is Most Likely to Begin at the Start of School. Journal of child neurology. 2016;31(7):864–868. [DOI] [PubMed] [Google Scholar]
- 118.Nierenburg H, Newman LC. Update on New Daily Persistent Headache. Current treatment options in neurology. 2016;18(6):25. [DOI] [PubMed] [Google Scholar]
- 119.Joshi SG, Mathew PG, Markley HG. New daily persistent headache and potential new therapeutic agents. Current neurology and neuroscience reports. 2014;14(2):425. [DOI] [PubMed] [Google Scholar]
- 120.Kacperski J Pharmacotherapy for Persistent Posttraumatic Headaches in Children and Adolescents: A Brief Review of the Literature. Paediatric drugs. 2018. [DOI] [PubMed] [Google Scholar]
- 121.Dodick DW, Goadsby PJ, Silberstein SD, et al. Safety and efficacy of ALD403, an antibody to calcitonin gene-related peptide, for the prevention of frequent episodic migraine: a randomised, double-blind, placebo-controlled, exploratory phase 2 trial. The Lancet Neurology. 2014;13(11):1100–1107. [DOI] [PubMed] [Google Scholar]
- 122.Tyburski AL, Cheng L, Assari S, Darvish K, Elliott MB. Frequent mild head injury promotes trigeminal sensitivity concomitant with microglial proliferation, astrocytosis, and increased neuropeptide levels in the trigeminal pain system. The journal of headache and pain. 2017;18(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Arruda MA, Bigal ME. Migraine and migraine subtypes in preadolescent children: association with school performance. Neurology. 2012;79(18):1881–1888. [DOI] [PubMed] [Google Scholar]
- 124.Bigal ME, Lipton RB, Winner P, et al. Migraine in adolescents: association with socioeconomic status and family history. Neurology. 2007;69(1):16–25. [DOI] [PubMed] [Google Scholar]
- 125.Guendelman S, Pearl M . Access to care for children of the working poor. Arch Pediatr Adolesc Med. 2001;155(6):651–658. [DOI] [PubMed] [Google Scholar]
- 126.Kogan MD, Newacheck PW, Blumberg SJ, et al. Underinsurance among children in the United States. The New England journal of medicine. 2010;363(9):841–851. [DOI] [PubMed] [Google Scholar]
- 127.St Peter RF, Newacheck PW, Halfon N. Access to care for poor children. Separate and unequal? JAMA : the journal of the American Medical Association. 1992;267(20):2760–2764. [PubMed] [Google Scholar]
- 128.Powers SW, Coffey CS, Chamberlin LA, et al. Trial of Amitriptyline, Topiramate, and Placebo for Pediatric Migraine. The New England journal of medicine. 2017;376(2):115–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Powers SW, Hershey AD, Coffey CS, et al. The Childhood and Adolescent Migraine Prevention (CHAMP) Study: A Report on Baseline Characteristics of Participants. Headache. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Frattarelli DA, Galinkin JL, Green TP, et al. Off-label use of drugs in children. Pediatrics. 2014;133(3):563–567. [DOI] [PubMed] [Google Scholar]
- 131.Sachs AN, Avant D, Lee CS, Rodriguez W, Murphy MD. Pediatric information in drug product labeling. JAMA : the journal of the American Medical Association. 2012;307(18):1914–1915. [DOI] [PubMed] [Google Scholar]
