2014;72(Suppl 1):9–11.

Koppel BS et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders.

Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology.

2014;82:1556–1563.

Patten SB et al. Major depression in multiple sclerosis: a population-based perspective. Neurology.

2003;61:1524–1527.

Patten SB et al. Psychotic disorders in MS: population-based evidence of an association. Neurology.

2005;65:1123–1125.

Schiffer RB et al. Association between bipolar affective disorder and multiple sclerosis. Am J Psychiatry.

1986;143:94–95.

Minden SL et al. Evidence-based guideline: assessment and management of psychiatric disorders in individuals

with MS. Report of the Guideline Development Subcommittee of the American Academy of Neurology.

Neurology. 2014;82:174–181.

Panitch HS et al. Randomized, controlled trial of dextromethorphan/quinidine for pseudobulbar affect in multiple

sclerosis. Ann Neurol. 2006;59:780–787.

Miller A, Panitch H. Therapeutic use of dextromethorphan: key learnings from treatment of pseudobulbar affect.

J Neurol Sci. 2007;259:67–73.

Pioro EP et al. Dextromethorphan plus ultra low-dose quinidine reduces pseudobulbar affect. Ann Neurol.

2010;68:693–702.

Bhargava P et al. The vitamin D to ameliorate multiple sclerosis (VIDAMS) trial:study design for a multicenter,

randomized, double-blind controlled trial of vitamin D in multiple sclerosis. Contemp Clin Trials. 2014;39:288–

293.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163.

164.

165.

166.

167.

Burton JM et al. A phase I/II dose-escalation trial of vitamin D3 and calcium in multiple sclerosis [published

corrections appear in Neurology. 2010;75:1029, Dosage error in article text; Neurology. 2010;75:480].

Neurology. 2010;74:1852–1859.

Soilu-Hanninen M et al. A randomised, double blind, placebo controlled trial with vitamin D 3

as an add on

treatment to interferon β-1b in patients with multiple sclerosis. J Neurol Neurosurg Psychiatry. 2012;83:565–

571.

Stein MS et al. A randomized trial of high-dose vitamin D2 in relapsing-remitting multiple sclerosis. Neurology.

2011;77:1611–1618.

Institute of Medicine, Food and Nutrition Board. Dietary Reference Intakes for Calcium and Vitamin D.

Washington, DC: National Academy of Sciences; 2011.

Jacobs LD et al. Intramuscular interferon beta-1a therapy initiated during a first demyelinating event in multiple

sclerosis: CHAMPS Study Group. N EnglJ Med. 2000;343:898–904.

Comi G et al. Effect of early interferon treatment on conversion to definite multiple sclerosis: a randomised

study. Lancet. 2001;357:1579–1582.

Kappos L et al. Treatment with interferon beta-1b delays conversion to clinically definite and McDonald MS in

patients with clinically isolate syndromes. Neurology. 2006;67:1242–1249.

Comi G et al. Effect of glatiramer acetate on conversion to clinically definite multiple sclerosis in patients with

clinically isolated syndrome (PreCISe study): a randomised, double-blind, placebo-controlled trial [published

correction appears in Lancet. 2010;375:1436]. Lancet. 2009;374:1503–1511.

Miller AE et al. Oral teriflunomide for patients with a first clinical episode suggestive of multiple sclerosis

(TOPIC): a randomized, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014;13:977–986.

Copaxone (Glatiramer Acetate Injection) [package insert]. North Wales, PA: Teva Neuroscience; 2014.

Carter NJ, Keating GM. Glatiramer acetate: a review of its use in relapsing-remitting multiple sclerosis and in

delaying the onset of clinically definite multiple sclerosis. Drugs. 2010;70:1545–1577.

Edgar CM et al. Lipoatrophy in patients with multiple sclerosis on glatiramer acetate. Can J Neurol Sci.

2004;31:58–63.

Jolly H et al. Impact of warm compresses on local injection site reactions with self-administered glatiramer

acetate. J Neurosci Nurs. 2008;40:232–239.

Galetta SL, Markowitz C. US FDA-approved disease modifying treatments for multiple sclerosis: review of

adverse effect profiles. CNS Drugs. 2005;19:239–252.

Walther EU, Hohlfeld R. Multiple sclerosis: side effects of interferon beta therapy and their management.

Neurology. 1999;53:1622–1627.

Nikfar S et al. A meta-analysis of the efficacy and tolerability of interferon-β in multiple sclerosis, overall and by

drug and disease type. Clin Ther. 2010;32:1871–1888.

Patten SB et al. Anti-depressant use in association with interferon and glatiramer acetate treatment in multiple

sclerosis. Mult Scler. 2008;14:406–411.

Porcel J et al. Long-term emotional state of multiple sclerosis patients treated with interferon beta. Mult Scler.

2005;12:802–807.

Brandes DW et al. Alleviating flu-like symptoms with dose titration and analgesics in MS patients on

intramuscular interferon beta-1a therapy: a pilot study. Curr Med Res Opin. 2007;23:1667–1672.

Rio J et al. Corticosteroids, ibuprofen, and acetaminophen for IFNbeta-1a flu symptoms in MS: a randomized

trial. Neurology. 2004;63:525–528.

Bayas A, Rieckmann P. Managing the adverse effects of interferon-beta therapy in multiple sclerosis. Drug Saf.

2000;22:149–159.

Mikol D et al; Rebiject Study Group. A randomized, multicentre, open-label, parallel-group trial of the tolerability

of interferon beta-1a (Rebif) administered by autoinjection or manual injection in relapsing-remitting multiple

sclerosis. Mult Scler. 2005;11:585–591.

Lanzillo R et al. Vitamin K cream reduces reaction at the injection site in patients with relapsing-remitting

multiple sclerosis treated with subcutaneous interferon beta—VIKING study. Mult Scler. 2015;21:1215–1216.

pii: 1352458514562989.

Costello K et al. Recognizing nonadherence inpatients with multiple sclerosis and maintaining treatment

adherence in the long term. Medscape J Med. 2008;10:225.

Saunders C et al. Factors that influence adherence and strategies to maintain adherence to injected therapies for

patients with multiple sclerosis. J Neurosci Nurs. 2010;42(5, Suppl):S10–S18.

Patti F. Optimizing the benefit of multiple sclerosis therapy: the importance of treatment adherence. Patient

168.

169.

170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193.

194.

Prefer Adherence. 2010;4:1–9.

Kleinman NL et al. Medication adherence with disease modifying treatments for multiple sclerosis among US

employees. J Med Econ. 2010;13:633–640.

Caon C et al. Injectable disease-modifying therapy for relapsing-remitting multiple sclerosis: a review of

adherence data. J Neurosci Nurs. 2010;42(5, Suppl):S5–S9.

Steinberg SC et al. Impact of adherence to interferons in the treatment of multiple sclerosis: a non-experimental,

retrospective, cohort study. Clin Drug Investig. 2010;30:89–100.

Prosser LA et al. Patient and community preferences for treatments and health states in multiple sclerosis. Mult

Scler. 2003;9:311–319.

Durelli L et al. Every-other-day interferon beta-1b versus once-weekly interferon beta-1a for multiple sclerosis:

results of a 2-year prospective randomised multicentre study (INCOMIN). Lancet. 2002;359:1453–1460.

Panitch H et al. Randomized, comparative study of interferon beta-1a treatment regimens in MS: the

EVIDENCE trial. Neurology. 2002;59:1496–1506.

Mikol DD et al. Comparison of subcutaneous interferon beta-1a with glatiramer acetate in patients with

relapsing multiple sclerosis (the REbif vs Glatiramer Acetate in Relapsing MS Disease [REGARD] study): a

multicentre, randomised, parallel, open-label trial. Lancet Neurol. 2008;7:903–914.

O’Connor P et al. 250 microg or 500 microg interferon beta-1b versus 20 mg glatiramer acetate in relapsingremitting multiple sclerosis: a prospective, randomised, multicentre study [published corrections appear in Lancet

Neurol. 2009;8:981; Lancet Neurol. 2011;10:115]. Lancet Neurol. 2009;8:889–897.

Vermersch P et al. Teriflunomide versus subcutaneous interferon beta-1a in patients with relapsing multiple

sclerosis: a randomised, controlled phase 3 trial. Mult Scler. 2014;20:705–716.

Coles AJ et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a

randomised controlled phase 3 trial. Lancet. 2012;380:1829–1839.

Sorenson PS. New management algorithms in multiple sclerosis. Curr Opin Neurol. 2014;27:246–259.

Gold R et al. Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclerosis. New Engl J Med.

2012;367:1098–1107.

O’Gorman J et al. Effect of aspirin pre-treatment or slow dose titration on flushing and gastrointestinal events in

healthy volunteers receiving delayed-release dimethyl fumarate. Clin Ther. 2015;37:1402.e5–1419.e5. pii:

S0149-2918(15)00188-5.

Phillips JT et al. Managing flushing and gastrointestinal events associated with delayed-release dimethyl

fumarate: experiences of an international panel. Mult Scler Relat Disord. 2014;3:513–519.

Tecfidera (Dimethyl Fumarate Delayed-Release Capsules) [package insert]. Cambridge, MA: Biogen; 2015.

Thrower BW. Relapse management in multiple sclerosis. Neurologist. 2009;15:1–5.

Alam SM et al. Methylprednisolone in multiple sclerosis: a comparison of oral with intravenous therapy at

equivalent high dose. J Neurol Neurosurg Psychiatry. 1993;56:1219–1220.

Ravnborg M et al. Methylprednisolone in combination with interferon beta-1a for relapsing-remitting multiple

sclerosis (MECOMBIN study): a multicentre, double-blind, randomised, placebo-controlled, parallel-group trial

[published correction appears in Lancet Neurol. 2010;9:759]. Lancet Neurol. 2010;9:672–680.

Imitola J, Racke MK. Is no evidence of disease activity a realistic goal for patients with multiple sclerosis?

JAMA Neurol. 2015;72:145–147.

Stangel M et al. Towards the implementation of ‘no evidence of disease activity’ in multiple sclerosis treatment:

the multiple sclerosis decision model. Ther Adv Neurol Disord. 2015;8:3–13.

Rotstein DL et al. Evaluation of no evidence of disease activity in a 7-year longitudinal multiple sclerosis cohort.

JAMA Neurol. 2015;72:152–158.

Kalincik T et al. Switch to natalizumab versus fingolimod in active relapsing-remitting multiple sclerosis. Ann

Neurol. 2015;77:425–435.

Laroni A et al. Safety of the first dose of fingolimod for multiple sclerosis: results of an open-label clinical trial.

BMC Neurol. 2014;14:65.

Multiple sclerosis patient taking Novartis’ Gilenya contracts PML. http://www.healthline.com/healthnews/multiple-sclerosis-patient-taking-novartis-gilenya-contracts-pml-030915. Accessed June 18,

2015.

Miravalle A, Corboy JR. Therapeutic options in multiple sclerosis: five new things. Neurology. 2010;75(18, Suppl

1):S22–S27.

Focosi D et al. Progressive multifocal leukoencephalopathy: what’s new? Neuroscientist. 2010;16:308–323.

Foley J. Recommendations for the selection, treatment, and management of patients utilizing natalizumab therapy

195.

196.

197.

198.

199.

200.

201.

202.

203.

204.

205.

206.

207.

208.

209.

210.

211.

212.

213.

214.

215.

216.

217.

218.

219.

220.

for multiple sclerosis. Am J Manag Care. 2010;16(6, Suppl):S178–S183.

Clifford DB et al. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple

sclerosis: lessons from 28 cases. Lancet Neurol. 2010;9:438–446.

Gilenya (fingolimod) [package insert]. Stein, Switzerland: Novartis Pharma Stein AG; 2015.

Arvin AM et al. Varicella-zoster virus infections in patients treated with fingolimod: risk assessment and

consensus recommendations for management. JAMA Neurol. 2015;72:31–39.

Confavreux C et al. Rate of pregnancy-related relapse in multiple sclerosis: pregnancy in Multiple Sclerosis

Group. N EnglJ Med. 1998;339:285–291.

Amato MP et al. Pregnancy and fetal outcomes after interferon-β exposure in multiple sclerosis. Neurology.

2010;75:1794–1802.

Sandberg-Wollheim M et al. Pregnancy outcomes in multiple sclerosis following subcutaneous interferon beta-1a

therapy. Mult Scler. 2011;17:423–430.

Fragoso YD et al. Long-term use of glatiramer acetate by 11 pregnant women with multiple sclerosis: a

retrospective, multicentre case series. CNS Drugs. 2010;24:969–976.

Karlsson G et al. Pregnancy outcomes in the clinical development program of fingolimod in multiple sclerosis.

Neurology. 2014;82:1–7.

Aubagio (teriflunomide) [package insert]. Cambridge, MA: Genzyme Corporation; 2014.

Kieseier BC, Benamor M. Pregnancy outcomes following maternal and paternal exposure to teriflunomide

during treatment for relapsing-remitting multiple sclerosis. Neurol Ther. 2014;3:133–138.

Lemtrada (alemtuzumab) [package insert]. Cambridge, MA: Genzyme Corporation; 2014.

Goodin DS et al. Neutralizing antibodies to interferon beta: assessment of their clinical and radiographic impact:

an evidence report: report of the Therapeutics and Technology Assessment Subcommittee of the American

Academy of Neurology [published correction appears in Neurology. 2007;69:712]. Neurology. 2007;68:977–

984.

Polman CH et al. Recommendations for clinical use of data on neutralizing antibodies to interferon-beta therapy

in multiple sclerosis. Lancet Neurol. 2010;9:740–750.

Karussis D et al; AC001 Multi-center Israeli Study Group. Long-term treatment of multiple sclerosis with

glatiramer acetate: natural history of the subtypes of anti-glatiramer acetate antibodies and their correlation with

clinical efficacy. J Neuroimmunol. 2010;220:125–130.

Vennegoor A et al. Clinical relevance of serum natalizumab concentration and anti-natalizumab antibodies in

multiple sclerosis. Mult Scler. 2012;19:593–600.

Goodin DS et al. The use of mitoxantrone (Novantrone) for the treatment of multiple sclerosis: report of the

Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology.

2003;61:1332–1338.

Novantrone (mitoxantrone injection) [package insert]. Rockland, MA: EMD Serono; 2010.

Marriott JJ et al. Evidence report: the efficacy and safety of mitoxantrone (Novantrone) in the treatment of

multiple sclerosis: report of the Therapeutics and Technology Assessment Subcommittee of the American

Academy of Neurology. Neurology. 2010;74:1463–1470.

Ellis R et al. Therapy-related acute leukaemia with mitoxantrone: four years on, what is the risk and can it be

limited? Mult Scler. 2015;21:642–645.

Funch D et al. Adherence to recommended dosing and monitoring for mitoxantrone in patients with multiple

sclerosis: a healthcare claims database study supplemented with medical records—the RETRO study.

Pharmacoepidemiol Drug Saf. 2010;19:448–456.

Tysabri (natalizumab) [package insert]. Cambridge, MA: Biogen Idec; 2015.

de la Hera B et al. Natalizumab-related anaphylactoid reactions in MS patients are associated with HLA class II

alleles. Neurol Neuroimmunol Neuroinflamm. 2014;1e47.

Berger JR, Koralnik IJ. Progressive multifocal leukoencephalopathy and natalizumab—unforeseen

consequences. N EnglJ Med. 2005;353:414–416.

Daniels GH et al. Altemtuzumab-related thyroid dysfunction in a phase 2 trial of patients with relapsing-remitting

multiple sclerosis. J Clin Endocrinol Metab. 2014;99:80–89.

Havla J et al. Bridging, switching or drug holidays—how to treat a patient who stops natalizumab? Ther Clin

Risk Manag. 2013;9:361–369.

Nuedexta (Dextromethorphan and Quinidine) [package insert]. Aliso Viejo, CA: Avanir Pharmaceuticals; 2015.

p. 1232

Migraine, tension-type, and cluster headache are classified as primary

headache disorders. Subtle differences in symptomatology and the

presence of auxiliary symptoms assist in defining headache type.

Case 58-1 (Questions 1, 2, 3,

6),

Case 58-2 (Questions 1, 3),

Case 58-4 (Question 1)

Virtually all patients who suffer from migraine, tension-type, and cluster

headaches are candidates to receive abortive (or symptomatic)

medications during an acute attack. Prophylactic treatment may be

considered for all primary headache types and is driven by various

factors including headache frequency, the level of disability imparted by

the headache, and response to acute treatment modalities.

Case 58-1 (Questions 4,

7–10, 11, 12),

Case 58-2 (Questions 2, 4,

5),

Case 58-4 (Questions 2, 3),

Case 58-5 (Questions 1, 2)

Triptans are the drugs of choice as migraine-specific treatments for

acute episodes. Triptans are effective in managing associated symptoms

of migraine (e.g., nausea, photo- and phonophobia). Differences within

the class are observed on the basis of pharmacokinetics.

Case 58-1 (Questions 5, 7,

8),

Case 58-2 (Question 4, Table

58-1)

Evidence supports the use of propranolol, amitriptyline, or topiramate as

first-line agents for migraine prophylaxis. The decision to initiate

prophylaxis takes into account headache frequency, the impact of

attacks on patient functioning, and response to acute treatments.

Medical history and adverse effect profile are relevant considerations

for specific drug selection.

Case 58-1 (Question 12),

Case 58-2 (Question 5)

Overuse of symptomatic or abortive agents (including triptans and overthe-counter, combination, and opioid analgesics) can increase the

intensity and chronicity of all headache types. To prevent medication

overuse headache, use of these agents should be limited to fewer than

10 days/month.

Case 58-3 (Question 1)

Cluster headaches are severely painful and short-duration attacks that

tend to occur nightly during susceptible periods and then enter remission

for a period of months or years. Treatments of choice for abortive

treatment must be fast-acting and include subcutaneous sumatriptan,

intranasal zolmitriptan, and oxygen inhalation.

Case 58-4 (Questions 1–3)

Tension-type headaches (previously known as tension or muscle

contraction headaches) usually cause mild-to-moderate discomfort.

Conventional analgesics are often appropriate for acute management.

Case 58-5 (Question 1, 2)

Some patients with tension-type headache may require prophylactic

treatment, for which amitriptyline, mirtazapine, and venlafaxine have

demonstrated efficacy.

EPIDEMIOLOGY AND DESCRIPTION

Prevalence

Previous investigations have determined that migraine prevalence approximates

12%.

1

,

2 Survey data obtained from the American Migraine Prevalence and

Prevention Study (2004) support the same percentage, as applied to meeting

International Classification of Headache Disorders-2 criteria.

2 Across ambulatory

care settings, migraine accounts for 0.5% of all encounters, with a majority occurring

in primary care settings whereas emergency department (ED) visits approximate

3%,

3 being further stratified within the top 10 causes for ED encounters in the U.S.

4

There is demonstrated variability across both gender and age, with a higher

incidence in females,

1–3 and a higher prevalence between 30 and 39 years.

1 Such ageand gender-associated prevalence has been further substantiated in surveillance

reviews.

4

p. 1233

p. 1234

Classification

There is inherent difficulty in evaluating and classifying headache, because this

symptom may be attributed to either a physiologic, psychological, or pathologic

process, or may be an adverse outcome associated with medications. Because the

processes contributing to headache are vast, it is imperative to conduct a

comprehensive patient assessment. Broadly categorized, headache is defined as

primary or secondary, with the latter typically being attributable to an organic cause.

Primary headache is inclusive of migraine, tension headache, and trigeminal

cephalalgias, of which cluster headache would be one category. Further, headache

associated with physical activity, exposure to exogenous stimuli, or those evolving

during the sleep cycle would be categorized as primary.

5 Secondary headache may be

attributable to, but not limited to, traumatic injury, vascular or nonvascular cranial

pathology, tumor formation (neurofibroma), or an infectious process.

5

The appropriate classification ultimately speaks to the larger issue of optimal

management, via either medical procedures or the initiation of medication. Because

secondary headache is associated with a causative component, targeting that cause

becomes a principle intervention strategy.

PRIMARY HEADACHE DISORDERS

Migraine

The timeline for the evolution of migraine can often be expressed in terms of minutes.

The clinical assessment process for differentiating headache type is dependent upon

recognizing general characteristics of migraine and further distinguishing such

characteristics from other primary headache forms. An assessment of location can be

useful but should not be used as a prime differentiator between headache types

because migraine location can be unilateral or bilateral in nature. Patient assessment

should include an evaluation of both pain intensity and pain quality, because

migraines are commonly described as moderate-to-severe pulsating pain.

5 Migraine

duration typically ranges from 4 to 72 hours and may be accompanied by auditory

(phonophobia) and light (photophobia) sensitivity, as well as nausea and vomiting.

5

,

6

These auxiliary symptoms are mediated via the hypothalamus and the chemoreceptor

trigger zone (CTZ), indicating diffuse neuroanatomic involvement. The

aforementioned features have demonstrated reliable predictive value in migraine

diagnosis, as defined by the POUND mnemonic (pulsating, 4–72 hour duration,

unilateral nature, nausea or vomiting, disabling effect).

7 The presence of 4 out of the

5 criteria indicates high probability of migraine.

7

Migraine may be preceded by a constellation of optical or sensory features,

defined as an aura. The presence or absence of an aura highlights an important

differentiator in migraine diagnosis. International Classification of Headache

Disorders criteria indicate a disparity in the number of episodes consistent with a

diagnosis of migraine without aura (minimum of five episodes) versus migraine with

aura (minimum of two episodes).

5 This criterion is not the only parameter

considered, because additional criteria pertaining to headache characteristics also

need to be met for an appropriate migraine diagnosis. Auras most often affect vision,

with either positive (visual feature) or negative (localized blindness) character.

5

Sensory effects can be characterized similarly as positive (pins and needles

description) or negative (loss of sensation or numbness).

5 Aura character that extends

beyond the visual and sensory, such as motor or speech irregularity, is linked more

toward specialized migraine diagnoses.

5 Patients with suspected migraine should be

advised to document aura timing and description, because this information can be

valuable for accurate diagnosis.

Patient report of pain location in migraine may be widespread due to the branching

of the trigeminal nerve, which facilitates sensory effects across facial regions.

Anatomically, the trigeminal nerve features branches of V1, V2, and V3.

8 Branch V1

innervates the scalp, orbital region, and meninges, V2 is directed to the top of the jaw

and sinus region, and V3 innervates lower mandibular regions.

8 Additionally,

patients may report pain referral to the suboccipital region due to the close proximity

between occipital nerves and the spinal nucleus.

8

Cluster Headache

Cluster headache is characterized within the domain of primary headache disorders.

Whereas migraine can be unilateral or bilateral, cluster headache is primarily

unilateral involving orbital and/or temporal pain, which can persist for up to 3

hours.

5

Independent of duration, such episodes may occur multiple times during the

day.

5 Associated symptoms that occur with high prevalence during cluster headache

include lacrimation, rhinorrhea, miosis, ptosis, diaphoresis, and flushing.

5

Preferential incidence of cluster headache occurs in males, but there is similarity to

migraine on the basis of age prevalence.

Tension-Type Headache

Tension-type headache is of bilateral character and by comparison to migraine, is

often associated with less intense pain without a pulsating quality. An individual’s

subjective description of pain quality and intensity can be useful in differentiation.

Patients may use such terminology as “tightening” to characterize symptoms. The

combination of symptoms of photophobia, phonophobia, and nausea/vomiting is not

considered to be consistent with the diagnosis of tension headache; however, the

individual symptoms of photo- or phonophobia may accompany tension-type

headache.

5

To complete the discussion pertaining to primary headache pathology, the

International Headache Society (IHS) defines headache in association with cough,

exercise, sexual activity, cold stimuli, and external compression under the umbrella

of primary headache disorders.

5

SECONDARY HEADACHE

Causation is the key principle underlying secondary headache. Such attributable

causes include head or neck trauma, cervical vascular dysfunction, nonvascular

cranial dysfunction, infection, psychiatric pathology, or substance withdrawal

effects.

5 Causation becomes a critically important principle to consider in patient

assessment, particularly when headache onset deviates substantially from existing

prevalence and epidemiologic data.

Acute Headaches

Acute headaches may be linked to subarachnoid hemorrhage, stroke, meningitis, or

intracranial mass (e.g., neurofibroma, abscess, etc.). It is within this clinical context

that diagnostic measures of neuroimaging and lumbar puncture are more commonly

implemented. Commonalities of headaches associated with such pathology are of

severe quality and rapid evolution, perhaps described as the “worst headache”

experienced. An increase in intracranial pressure is of particular importance, and

recognition of factors that can impact intracranial pressure becomes critical in patient

assessment.

Subacute Headaches

Subacute headaches may be a sign of increased intracranial pressure, intracranial

mass lesion, temporal arteritis, sinusitis, or trigeminal neuralgia. Trigeminal

neuralgia usually occurs after the age of 40 and is more common in women than in

men. The pain usually occurs along the second or third divisions of the trigeminal

(facial) nerve and lasts only moments. Trigeminal neuralgia is characterized by

sudden, intense pain that recurs paroxysmally, often in response to triggers such as

talking, chewing, or shaving.

p. 1234

p. 1235

The evaluation of headache pathology should incorporate the consideration of

reported prevalence data, patient age, the presence of triggers or aggravating factors,

the contribution of past medical history to headache origin, the patient’s description

and characterization of symptoms and frequency, and any record of self-care

interventions employed to manage headache. Evaluating the impact of headache on

daily activity and function is a similarly important consideration. A diagnosis may be

confirmed on the basis of this information without the need for further laboratory or

procedural measures. If objective or subjective information becomes suggestive of a

clinical anomaly (e.g., late-age onset of headache, suspected structural anomalies or

neural tissue pathology, etc.), a more comprehensive evaluation, inclusive of

additional diagnostic measures (e.g., imaging, lumbar puncture), is indicated.

Pathologic Basis

The pathologic picture of headache requires an understanding of vascular physiology

and the functions of the trigeminovascular circuit and neuropeptides. Headache

involves both peripheral and central pain processing, afferent neuronal excitability,

and pain sensitivity of cerebral arteries and venous sinuses.

5

,

9 The pathology has

often been described within the context of interactions between vascular and

neurologic systems.

6

,

10 Muscular tenderness similarly contributes to headache

pathology.

9

,

11 Such an effect is more commonly considered in association with

tension headache.

Recent evidence has conferred support for the interplay between vascular and

neurologic function. Afferent and efferent fibers of the trigeminovascular circuitry,

vasoactive neuropeptides stored within this system, and serotonin (5-HT) maintain

significant pathologic importance.

6

,

12

,

13 Neuropeptides, such as substance P,

neurokinin A, nitric oxide, calcitonin gene-related peptide, and adenylate cyclaseactivating peptide, are promoters of neuroinflammation and vessel dilation.

6

,

13

Recent evidence further suggests an elevation in pro-inflammatory C-reactive protein

in migraine sufferers compared to controls.

14 Consequently, therapeutic interventions

have focused on modulating the effects of 5-HT with recent efforts directed toward

the modulation of neuropeptide function.

Drug Therapy

The primary goal of therapy is the resolution of headache symptoms, most

prominently pain, and attenuating disability associated with the headache.

Nonpharmacologic recommendations should similarly be paired with therapeutic

options. Pharmacologic treatment options can be divided into abortive and

prophylactic, the former resolving acute episodes and the latter being administered

chronically to reduce the incidence of recurrent headaches. Abortive treatment may

sufficiently manage tension-type headache, often negating the need for prophylactic

treatment; however, this may be considered when acute treatment is ineffective,

overused, or when the headache is chronic or occurring in frequent episodes.

Conversely, patients with migraine and cluster headache, while requiring abortive

treatment, may commonly also require long-term prophylactic treatment.

Success in managing acute episodes can often be achieved with the use of

conventional analgesics or headache-specific medications (e.g., dihydroergotamine,

triptans). Medication selection should be based on the patient’s prior treatment

experience and the level of disability imparted by the headache episode. With regard

to prophylactic therapy, evidence supports the use of select antidepressants,

anticonvulsants, and antihypertensives. The selection of such agents should be

considered within the context of adverse effect profiles and the potential for utility in

managing other medical conditions (e.g., prescribing a β-antagonist for migraine

prophylaxis in a patient with a history of hypertension). Rational prescribing limits

both the potential for polypharmacy and the risk of adverse effects.

MIGRAINE HEADACHE

Migraine is characterized by pain of a throbbing quality with moderate-to-severe

intensity and the presence of associated symptoms, including nausea, vomiting, light,

and/or auditory sensitivity.

5

,

6 Migraine can be unilateral or bilateral and can be

further classified on the presence of an aura.

5 The occurrence of an aura has

diagnostic relevance based upon the number of episodes and can encompass a range

of sensory disturbance, not solely limited to visual field effects.

5

Pathophysiology

Vascular constriction was a prior central focus of migraine pathology, resulting in

hypoperfusion and being resolved through compensatory vasodilation.

6

,

15 Such

alterations in blood flow have been observed in neuroimaging of migraine

patients.

6

,

16–18 There are further alterations in neuronal depolarization, termed

cortical spreading depression.

6

,

19

,

20 There has been further demonstration of causality

between cortical spreading depression and the activation of trigeminovascular

circuitry.

6

,

21

Implications of cortical spreading depression include changes in pH,

nitric oxide concentration, and the glutamatergic system.

21 The trigeminovascular

system consists of afferent fibers with a foundation in trigeminal ganglia, projecting

peripherally to intracranial blood vessels and venous sinuses and to extracranial

tissues.

6

,

22 Trigeminal branches innervate the scalp, orbit, meninges, and mandibular

regions.

8 Activation of the trigeminovascular system facilitates the release of

vasodilatory-promoting neuropeptides (substance P, neurokinin A, calcitonin generelated peptide, nitric oxide), and subsequent release results in vessel dilation and

inflammation.

6

,

13

,

23

,

24 Further effects include the activation of both the superior

salivary nucleus and parasympathetic efferent fibers.

20

Evidence from positron emission tomography scanning (a technique to measure

regional cerebral blood flow as an index of neuronal activity) suggests that episodic

dysfunction of the brainstem, with corresponding effects on the trigeminal system, is

involved.

25 Weiller et al

25 confirmed brainstem activation (periaqueductal gray,

dorsal raphe nucleus, and locus coeruleus) at the onset of migraine headaches in a

small sample of patients, and this area may represent an endogenous “migraine

generator.” Whereas the brainstem may be regarded as an anatomic point of

initiation, sensory information in migraine is propagated via higher regions of the

hypothalamus, thalamus, and cortex. Sporadic dysfunction of the nociceptive system

(periaqueductal gray and dorsal raphe nucleus) and the neural control of cerebral

blood flow (dorsal raphe nucleus and locus coeruleus) are hypothesized to trigger

migraine headache via effects of these brain structures on the trigeminovascular

system.

The therapeutic effects of drugs that stimulate 5-HT1

receptors (e.g.,

dihydroergotamine, sumatriptan), antagonize 5-HT2

receptors (e.g., cyproheptadine),

prevent 5-HT reuptake (e.g., amitriptyline) or release (e.g., calcium-channel

blockers), or inhibit brainstem serotonergic raphe neurons (e.g., valproate) all lend

support to the hypothesis that 5-HT is an important mediator of migraine.

Furthermore, brainstem nuclei activated during migraine have high densities of

serotonergic neurons. Specific 5-HT receptor subtypes, 5-HT1B and 5-HT1D, are

largely distributed in blood

26 and nerves,

27

respectively. These same 5-HT receptor

subtypes

p. 1235

p. 1236

are the targets of antimigraine drugs such as the triptans and ergot alkaloids.

GENETICS

The genetic basis of migraine is perhaps most effectively understood within the

context of familial hemiplegic migraine. This subtype has been further categorized

into multiple mutation-induced forms with associated genes CACNA1A, ATP1A2,

SLC1A3, SLC4A4, and SCN1A.

6

,

28 Such genes encode α1 subunits (CACNA1A,

SCN1A) and α2 subunits (ATP1A2), or are linked with glutamate-associated

(SLC1A3) and sodium-bicarbonate-associated (SLC4A4) transporters.

6

,

28 The

former α-subunit encoders regulate ion passage through calcium and sodium

channels. A further genetic linkage with migraine pathology has been identified in the

potassium channel, TRESK.

28

Implications of mutations at this level impact neuronal

resting membrane potential, because an identified frameshift mutation negates

TRESK action.

28

,

29 There is further support for the influence of genetics on migraine

pathology when considering the disproportionate prevalence across gender. Because

prevalence is higher for females, polymorphisms of the estrogen receptor-1 (ESR-1)

gene have been associated with increases in migraine risk.

1

,

30

,

31 Advances in the

understanding of the genetic bases for pathology maintain importance, as such

developments impart promise for novel interventional strategies.

Migraine pathology is multifaceted and neuroanatomically diffuse, involving the

brainstem trigeminal nuclei, trigeminovascular afferents, thalamus, hypothalamus,

CTZ, and cortical regions. Therapeutic strategies target the dysfunction in these

regions, the mediation of associated symptomatology, and the modulation of

neuropeptides and neurotransmission.

Quantitative Assessment Instruments

Various patient assessment instruments used to evaluate headache impact include the

Headache Impact Test (HIT-6), Migraine Disability Assessment (MIDAS), and the

Migraine-Specific Quality of Life Questionnaire (MSQ). The HIT-6 utilizes a Likert

scale and evaluates pain, social and cognitive function, and psychological impact.

32

,

33

A higher score on this assessment confers a greater headache impact. Rendas-Baum

et al.

33 confirmed validation of this instrument in the context of chronic migraine. The

Migraine Disability Assessment (MIDAS) is a self-administered, 7-item

questionnaire, assessing headache-induced absenteeism, activity limitations, and

headache frequency and intensity. This instrument has demonstrated reliability

34 and

validity.

34–36 A regression analysis confirmed a significant, independent association

between MIDAS score and pain intensity, headache frequency, and patient age.

34 The

MSQ addresses migraine impact across patient social activity, emotional activity,

and the capacity of migraine to prevent activity.

33 With established validity and

reliability,

37–39 strong correlations between the MSQ and HIT-6 have been confirmed

in the context of chronic migraine.

39

MIGRAINE WITHOUT AURA

CASE 58-1

QUESTION 1: L.P. is a 27-year-old female presenting to her primary care physician (PCP) with a chief

complaint of a “severe, throbbing headache.” L.P. reports that headaches began 18 months ago, but during the

past 3 months, headaches have been occurring with greater frequency, approximating one episode every 2 to 3

weeks. She further describes bilateral headache pain, occurring “without warning,” and coinciding symptoms of

nausea and vomiting, photophobia, and suboccipital pain. Prior episodes have been self-treated with

acetaminophen 500 mg (4–8 tablets/episode as needed) with variable efficacy. Her past medical history is

significant for generalized anxiety disorder, gastroesophageal reflux, and seasonal allergies. Current medication

usage includes levonorgestrel/ethinyl estradiol 0.10/0.02 mg by mouth daily, venlafaxine XR 75 mg by mouth

daily for generalized anxiety disorder (GAD), and omeprazole 20 mg by mouth daily × 2 months for recentonset gastroesophageal reflux disease symptoms. Social history is negative for both tobacco use and illicit

substances. L.P. reports the “occasional” consumption of alcohol, stated as “1 to 2 cocktails after work on

Fridays.” As headaches have occurred with greater frequency, absenteeism from work has resulted and this

issue has prompted a visit with the PCP. Physical and neurologic examinations are unremarkable.

What subjective and objective information is consistent with a migraine diagnosis, and how do these

signs/symptoms differ from tension-type headache?

Symptoms reported by L.P. are consistent with migraine. A study by Kelman

addressed migraine location, reporting unilateral character in 67% of study subjects

and bilateral character in nearly 24%.

40 For L.P., migraine location would not be a

differentiating characteristic between migraine and tension headache, because tension

headache is more commonly associated with bilateral character.

9 Pain quality,

described as “severe” and “throbbing,” is consistent with migraine and precipitates

interruptions with daily activity (e.g., absenteeism from work) contrasted with

tension headache which is associated with sensations of tightness.

9 Similarly, the

occurrence of nausea, vomiting, and photophobia more appropriately aligns with

migraine.

5

Migraine prevalence is higher in female patients than in male patients,

approximating 17%2 with a cumulative incidence in females of 43%.

41 Migraine

prevalence within the range of 18 to 29 years has been reported as nearly 21%.

2

L.P.’s sex and age would be compatible with prevalence data although it should be

noted that a 28% prevalence statistic for females has been reported in the age range

of 30 to 39.

2

CASE 58-1, QUESTION 2: Which characteristics may be classified as precipitating factors, or “triggers,” for

migraine pathology? What are possible precipitating factors in L.P.?

Factors involved in precipitating migraine should be routinely assessed for all

patients. Assessment should address factors within a patient’s medical history,

medication usage, social history, and environment. L.P.’s medical history is

significant for GAD, she denies the usage of tobacco and illicit substances, she does

report the occasional use of alcohol. Alterations in neurotrophic factors and an

increase in anxiety symptoms have been demonstrated in migraine patients.

42 Such

evidence imparts value to assess this aspect of L.P.’s medical history. While the

consumption of alcohol should be addressed, the linkage between alcohol

consumption and precipitating migraines is less clear.

43

,

44 The potential provocation

of cortical spreading depression, an electrophysiologic factor underlying migraine

pathology, in association with alcohol has been suggested.

45 Although not reported by

L.P., an evaluation of diet may provide useful information in determining migraine

triggers. The evaluation of dietary patterns and quality has yielded disparities,

reporting lower-quality dietary intake in migraine patients.

46

CASE 58-1, QUESTION 3: Are further diagnostic and laboratory tests indicated in the case of L.P.?

The most important diagnostic evaluations of patients presenting with headache

should be based on a thorough medical history and physical examination. Imaging

(e.g., CT scans, MRI) is generally not required in the context of uncomplicated

migraine.

47

p. 1236

p. 1237

The report of symptoms by L.P. is consistent with migraine and is sufficient for

diagnosis. Her symptoms are not associated with a secondary cause, such as trauma

or other injury to the head or neck, and her neurologic examination is unremarkable.

Similarly, without reason to suspect subarachnoid hemorrhage or symptoms

secondary to meningitis, lumbar puncture would not be indicated. Clinical

manifestations that may warrant neurologic imaging to identify artifacts or structural

anomalies include coordination deficits, localized neurologic pathology, sensory

manifestations, an abnormal neurologic examination, or the presence of atypical

headache characteristics.

7

,

47 Further, a change in headache character and headache

onset with advanced age warrants neuroimaging.

7

CASE 58-1, QUESTION 4: What are the goals of therapy for the management of headache in L.P.?

The approach to management and the treatment objectives for L.P. include

resolving the acute symptoms of pain, nausea, and photosensitivity, educating L.P. on

both prescription and self-care treatment, addressing the contribution of past medical

history and concurrent medication usage to headache episodes, and identifying

potential triggers.

CASE 58-1, QUESTION 5: What is the most appropriate recommendation with regard to the use of

acetaminophen to treat headache episodes in L.P.?

L.P. has confirmed self-treatment of headaches with acetaminophen, and in such

instances, it is critical to not only evaluate each patient’s potential for self-care but

also determine whether self-care treatment options can effectively and safely manage

the diagnosis. Reported efficacy of acetaminophen has been mixed. Evidence does

support acetaminophen efficacy in the migraine patient,

48

,

49 and comparisons of

acetaminophen with placebo yielded a nearly 20% difference in headache

resolution.

48

,

49 However, a study evaluating the parenteral administration of

acetaminophen for acute management found no significant difference in efficacy

versus placebo.

48

,

50 Considering L.P.’s report of “variable efficacy” and increase in

headache frequency, she is not an appropriate self-care candidate and continued,

routine use of acetaminophen to manage acute episodes is not recommended.

48

CASE 58-1, QUESTION 6: What drug-related problems associated with the precipitation or worsening of

migraine should be addressed in L.P.?

Oral contraceptives may either worsen or precipitate migraine attacks in women

without a previous history of this problem.

51

Incidence may be related to estrogen

dose

52 and/or duration of oral contraceptive use.

52

,

53 Alternatively, migraine has been

implicated as an effect of estrogen withdrawal.

52

,

54 Such patients may benefit from

use of a continuous combined oral contraceptive product (e.g., one with no hormonefree interval), use of low-dose estrogen supplementation after 21 days of active

contraceptives, or the use of a progestin-only oral contraceptive.

51

,

55

,

56

The American Congress of Obstetricians and Gynecologists supports oral

contraceptive use in migraine patients, but recommend avoidance in patients with

tobacco use, in those 35 years or older, and/or in those with the presence of

neurologic signs.

57 None of these factors are present in L.P.; therefore, oral

contraceptive use may be continued. However, the implications of oral contraceptive

usage and headache should be discussed with L.P. to allow for informed decisionmaking.

Further, consultation with L.P. should address whether GAD is being optimally

managed. The innervation of trigeminovascular circuitry by neurotransmitters (e.g.,

norepinephrine, 5-HT) linked to affective pathology (e.g., anxiety, depression) has

been established

58 and is known to coexist with such patients reporting increased

headache frequency.

59

It is prudent to address anxiety management with L.P. and to

further assess her response to venlafaxine to determine whether dose adjustments or

medication changes are warranted.

CASE 58-1, QUESTION 7: Identify treatment strategies and recommend a medication as a first-line

treatment option for the management of migraine in L.P.

Treatment should be aimed at rapidly resolving symptoms and the restoration of

the patient’s routine activity.

47

,

60 Migraines may be managed via a “step” or

“stratified” approach.

60

,

61 With a “step” care model, conventional analgesics are

initiated first with the opportunity to progress to using more targeted therapies as

necessary.

60

,

61 A “stratified” care model is based on the character of the migraine and

will further incorporate validated assessment measures into the treatment

decision.

60

,

61

In this model, a more targeted, migraine-specific treatment may be

considered first line. A comparison of treatment approaches has favored the

“stratified” model.

61 The current treatment approach for L.P. has mimicked a “step”

care model, using acetaminophen as a first-line medication.

Triptans (5-HT1B/1D Receptor Agonists)

Triptans, categorized as first (sumatriptan) and second generation (zolmitriptan,

naratriptan, rizatriptan, almotriptan, frovatriptan, and eletriptan), are effective and

well tolerated in acute migraine management and are considered appropriate for

those not responsive to conventional analgesics.

Triptans can be compared on the basis of efficacy, onset, and recurrence potential.

Naratriptan and frovatriptan demonstrate lower recurrence potential whereas the

remaining agents within the class (sumatriptan, rizatriptan, almotriptan, eletriptan,

and zolmitriptan) yield higher efficacy and faster onset.

60 Compared with ergot

derivatives, triptans exhibit greater receptor subtype specificity, targeting 5-HT1B

and 5-HT1D receptors.

60

,

62

It is important to note that the specific nature of the effect

differs according to receptor subtype. Constriction of the vasculature is mediated via

5-HT1B interaction

60

,

62

,

63 whereas interaction via 5-HT1D antagonizes release of

neuropeptides.

60

,

63 Triptans further diminish neuronal excitability within the

trigeminovascular circuitry,

64 and there is evidence of impact on nitric oxide-based

signal transduction.

63

Table 58-1 depicts dosing and pharmacokinetic parameters for the triptans.

Sumatriptan

Sumatriptan is the prototype of the triptan class and is available in several

formulations, including an oral tablet, nasal spray, and subcutaneous injection (pen

devices and a needle-free solution). A recent analysis confirmed efficacy relative to

placebo across sumatriptan doses and formulations; however, subcutaneous delivery

yielded a more robust analgesic response.

65 Migraine intensity and the character of

associated symptoms (e.g., severity of nausea and vomiting, early-onset nausea)

impact formulation selection. For patients who are nauseated and prone to vomit

during an acute attack, the subcutaneous and intranasal dosage forms are preferred.

Both formulations have a rapid onset of effect. Reduction in headache intensity is

reported within 10 minutes after subcutaneous injection and within 15 minutes after

administration of the nasal spray.

41 Comparisons of orally administered sumatriptan

(doses of 25, 50, and 100 mg) have yielded higher efficacy rates relative to placebo

in achieving headache relief at 2 hours.

66 Higher sumatriptan doses of 50 and 100 mg

were superior to placebo in achieving headache relief at 4 hours.

66

Comparative efficacy studies with sumatriptan have yielded mixed results.

Subcutaneous sumatriptan is more effective and

p. 1237

p. 1238

faster acting than dihydroergotamine (DHE) nasal spray.

67 When compared with

subcutaneous DHE, subcutaneous sumatriptan is more effective at 1 and 2 hours, but

the two treatments are equally effective at 3 and 4 hours.

68

In this trial, headache

recurrence rates at 24 hours favored subcutaneous DHE.

68

Intranasal administration

of these agents supports efficacy of sumatriptan for both headache relief and the relief

of associated symptoms (e.g., nausea) 60 minutes after dosing.

69 Similar efficacy has

been reported for comparison with almotriptan

70 and zolmitriptan,

65 with diminished

efficacy being reported versus eletriptan

71 and rizatriptan.

65

Adverse Effects

Although sumatriptan is well tolerated, there is some variability in the adverse effect

profile across different formulations. In general, both oral and intranasal sumatriptan

formulations are associated with a low incidence of systemic adverse events. Oral

administration of sumatriptan has been associated with adverse effects including

dizziness, fatigue, nausea, and vomiting,

66

,

72 whereas intranasal administration may

cause abnormal taste and nasal discomfort.

73

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more