zafonte_tbi

Embed Size (px)

Citation preview

  • 8/3/2019 zafonte_tbi

    1/12

    J Head Trauma RehabilVol. 19, No. 2, pp. 89100

    c 2004 Lippincott Williams & Wilkins, Inc.

    Acute Care Management

    of Post-TBI SpasticityRoss Zafonte, DO; Elie P. Elovic, MD; Lisa Lombard, MD

    The management of persons with traumatic brain injury (TBI) who have acute spasticity remainsa challenge. A correct stratification is essential to clinical care and determination of efficacy fromproposed interventions. The secondary severe sequlae of spasticity after TBI can result in pro-found functional impairment. These concerns are often best addressed early in the patientscourse.Limited research in the area of the acute care treatment of severe spasticity is available. The au-thors employ a review of the available data as well as recount their own clinical experience inthe acute care management of severe spasticity to assist in developing an order for the plethoraof potential treatments available to clinicians and researchers. We propose to use a case exam-

    ple to emphasize key clinic points in the management of spasticity in the acute care setting.Key words: acute care, intensive care, spasticity, TBI

    M ANAGEMENT of the person with trau-matic brain injury (TBI) who has con-siderable spasticity must begin in the acutecare setting. Throughout the rehabilitationprocess it is important to treat someone whohas sustained a TBI with the expectation that

    he/she will have significant recovery. Perhapsthe most important yet difficult goal for spas-ticity management in the acute care settingis maintaining range of motion, but despiteaggressive approaches, this may not alwaysbe possible. Confounding acute medical is-sues may limit treatment options and makespasticity management even more challeng-ing. However the authors have seen individu-als who have emerged from low-level states,only to have their functioning greatly lim-ited by loss of range of motion as a resultof ineffective spasticity intervention. There-

    From the Department of Physical Medicine andRehabilitation, University of Pittsburgh, Pittsburgh,Pa (Drs Zafonte and Lombard); the Kessler MedicalRehabilitation Research and Education Corporation,West Orange, NJ, and the University of Medicine and

    Dentistry, NJ (Dr Elovic).

    Corresponding author: Ross Zafonte, DO, Departmentof Physical Medicine and Rehabilitation, University of

    Pittsburgh, 3471 fifth ave suite 201, Kaufman Bldg,Pittsburgh, PA 15213 (e-mail: [email protected]).

    fore, an ordered approach to both physicaland pharmacological interventions is essentialto possibly prevent the secondary sequelae ofspasticity. Since the literature regarding spas-ticity therapy is sparse, this review will em-ploy a case example to emphasize key points

    in the decision-making paradigms for suchpersons.

    CASE DISCUSSION

    A 24-year-old male was admitted to the inten-sive care unit (ICU) after a motor vehicle crash.He required a prolonged extrication time and wasnoted to be mildly hypotensive at the seen of theaccident. He was transported to a trauma cen-ter where his post resuscitation Glasgow ComaScale was 3T. Computed axial tomographic (CAT)scan imaging noted multiple petechial hemor-rhages and a markedly depressed skull fracture

    in the left parietal region, as well as a large sub-dural hematoma and a midline shift of 5 mmwas appreciated. He underwent evacuation ofthe hematoma and repair of the depressed skullfracture. A 24-hour period of elevated intracranialpressure was responsive to intermittent manni-tol therapy. His course was complicated by respi-

    ratory insufficiency and pneumonia. It is now 1-week post injury and we are invited into the ICUto evaluate the patients severe tone. Our case

    89

  • 8/3/2019 zafonte_tbi

    2/12

    90 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    discussion will focus only on the acute treatmentof spasticity.

    EVALUATION

    Evaluation in the acute care setting

    Review

    There is literature that has demonstratedthe importance and efficacy of a rehabilita-tion evaluation in the acute care setting. Wag-ner and colleagues have reported that earlyacute care rehabilitation consultation resultsin improved outcome at acute care discharge,in addition to shorter acute care length ofstay.1 The rehabilitation teams role in themanagement of spasticity especially amongthose with more severe injuries may be mostcritical.

    Hinderer has proposed that the clinicalevaluation of such patients be divided into5 components: clinical history, stretch re-flex examination, passive motion examina-tion, active motion examination, and func-tional examination.2While all of the above are

    important, it may be challenging to evaluatefunctional status in the acute care setting andmaintaining range of motion is often the pri-mary focus in this setting. A careful evalua-tion of the history and care record will help

    Figure 1. Evaluation of the acute TBI patient with spasticity.

    to delineate risk factors and issues that mayinterfere with therapy. Care should be takento discuss prior functional history and deter-mine prior neurological or gait dysfunction.

    Risk factors for early spasticity include im-mobilization, motor dysfunction (hemiplegiaor tetraplegia), associated hypoxic ischemicinjury, spinal cord injury, and age.2,3 While re-specting range of motion precautions as wellas spinal stability a comprehensive evaluationof both the active and passive range of mo-tion of each available extremity should be per-formed. At this time it is helpful to also notethe perceived tone and range of motion ateach major joint. The positioning of a venti-lator may make shoulder evaluation difficultat times. Serial assessments of tone are oftenof value since severity may vary with stimuli(ie, prior medications, procedures that day).Tone is an important issue for the rehabilita-tion team to address as the acute care teamrarely attends to this issue and it may play arole in ultimate outcome and disposition. Itis recommended that some objective measureof tone such as the Ashworth Scale be used to

    quantify tone at each joint.The evaluation of patients with spasticity

    requires clinicians to adopt an organized sys-tematic approach to the problem (see Fig 1).After documenting tone and range of motion

  • 8/3/2019 zafonte_tbi

    3/12

    Acute Care Management of Post-TBI Spasticity 91

    the next step is to evaluate for any second-ary factors, such as occult injury (fracture,neurodysfunction [hydrocephalus or occultcerebral lesion], infection, heterotopic ossi-

    fication, and pain that may be exacerbatingthe motor dysfunction.4 Several medicationscan impact the evaluation of spasticity in theperson with TBI. In the intensive care set-ting the use of neuroparalytic agents, opioids,benzodiazepines, and propofol may mask theseverity of the spasticity that exists at base-line. Neuroparalytics remove almost all tone,while opioids and benzodiazepines appear todecrease tone. Propofol is a unique lipid solu-ble agent that produces a sedative effect, yetcan be quickly stopped to allow for motor andcognitive evaluation.5

    Chronology of spasticity

    One does not have a thorough understand-ing of how to evaluate spasticity, unless oneunderstands the importance of chronologyof spasticity and severity of the motor dys-function. The development of early spasticitycarries with it a more challenging prognosis.

    Those with more severe brain injuries, as wellas those with associated spinal cord injury, hy-poxic ischemic injury, and/or associated au-tonomic dysfunction, tend to develop earliermore aggressive forms of the dysfunction. Wepropose the term malignant spasticity to fur-ther define those persons in the acute care pe-riod whose spasticity is so severe that withoutaggressive early intervention care cannot beprovided and loss of functional range of mo-tion is almost certain.

    Case discussion

    A systematic evaluation of the patients situ-ation yields some concerning clinical data. The

    patients spasticity developed very early in hisclinical course and over a 2-day period of timehis tone progressed rapidly. The patient showsevidence of flexor tone in the upper extremitiesand extensor tone in the lower extremities. Serialevaluation of tone in the right upper and lower ex-tremities noted Ashworth 2 tone and Ashworth 3

    tone respectively. The patient had dysautonomicepisodes and severe posturing. Adductor tonewas already profound. Our team noted early de-

    crease in range of motion at the right ankle andelbow. His symptoms had been relieved only withsedatory therapy.

    TREATMENTS

    Physical modalities

    Physical modalities offer treatment withoutthe risk of systemic side effects that can occurwith systemic medications. Passive stretchingremains a core therapy in the management ofspasticity as well as in the prevention of con-tracture formation. This technique has beenshown to decrease spasticity in persons withstroke6 and cerebral palsy.6 Standing has alsobeen used as a form of prolonged stretch toreduce spasticity in those with spinal cordinjuries.7 An examination in a population ofbrain injured patients indicated that a seriesof passive flexion and extension movementsof the elbow result in a short-term reductionof tone.8 Clinically, the stretching program istypically performed 1 to 2 times a day. This isa relatively easy modality that can be taught

    to family members and selected patients toaid in spasticity management. One of themost accessible adjunctive therapies to pas-sive stretch is cryotherapy, the application ofcold acts to reduce stretch reflex activity9,10

    and induce analgesia. Treatment should be re-stricted to a maximum of 20 minutes at atime to reduce the risk of skin injury.11 Un-fortunately, the reduction of spasticity withcold application alone is limited to a half anhour.

    Splinting and serial casting, while morecommonly used in both inpatient and outpa-tient rehabilitation, can also be a valuable toolin the acute care setting. Casting in the up-per extremity has been shown to decrease the vibratory inhibitory index of the H-reflex.12

    After 5 weeks of casting, Singer et al founddecreased torque and reflexive stretch re-sponses in adults with spastic equinovarusdeformities due to brain injury.13 In addition

    to the physiological evidence of the benefitsof casting, studies have demonstrated an im-provement in function after the application

  • 8/3/2019 zafonte_tbi

    4/12

    92 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    of casts to spastic limbs.14,15 Casting does re-quire a significant length of time before im-provements are seen15,16; however, constant vigilance on the part of physicians, nurses,

    and therapists is required to mitigate pres-sure sore formation. Complication rates canbe decreased with a reduction of the cast-ing interval from 57 days to 14 days withno detriment to the improvement in range ofmotion.16 While treatment of patients withspasticity with casts in a population of pa-tients with a recent TBI (

  • 8/3/2019 zafonte_tbi

    5/12

    Acute Care Management of Post-TBI Spasticity 93

    of the agents when they are used in the TBIpatient.

    Dantrolene sodium: (Dantrium)

    Dantrolene is used in patients with mus-cle spasticity and is the only known effec-tive treatment for malignant hyperthermia.However, its effects on muscle relaxationand energetics are unknown and mayhave important consequences in diaphrag-matic function.26 Dantrolene sodium, 1([5-(nitrophenyl)furfurylidend]amino) hydan-toin sodium hydrate, is indicated for use inchronic disorders characterized by skeletalmuscle spasticity, such as spinal cord injury,stroke, cerebral palsy, and multiple sclerosis.Dantrolene is believed to act directly on thecontractile mechanism of skeletal muscle todecrease the force of contraction in the ab-sence of any demonstrated effects on neuralpathways, on the neuromuscular junction,or on the excitable properties of the musclefiber membranes.27 Dantrolene acts at thescaroplasmic reticulum by inhibiting calciumactivity. It has been evaluated in the treat-

    ment of those with cerebral palsy, multiplesclerosis, and spinal cord injury. Its role inthe treatment of severe spasticity in theacute care setting has not been adequatelyexplored.

    Laboratory evaluation of the potential roleof dantrolene in the management of acutecare spasticity raises some compelling issues.Cho et al evaluated the hemodynamiceffects of dantrolene in dogs that whereinstrumented to further define cardiac

    parameters.28 They noted that dantrolenereverses the hypotensive action produced bypropofol and causes an increase in coronaryblood flow with a decrease in coronary vascular resistance, but does not signifi-cantly change the negative inotropic effectsof propofol. In a laboratory investigation,Nakayama et al noted that intracerebroven-tricular dantrolene prevents delayed neuronalloss in the rat hippocampal CA1 region

    subjected to transient ischemia.29

    Popescuhas demonstrated that dantrolene sodiummay protect against kanic acid induced

    apoptosis.30 This agent does appear to crossthe blood brain barrier but only in modestfashion. While the evidence is relatively weakthere may be some theoretical advantages of

    dantrolene sodium in the acute care setting.Little literature exists to refine the clin-

    ical use of dantrolene among those withbrain injury. Seinberg et al studied the ef-fects of dantrolene sodium in 23 patients withhemiplegic spasticity of various etiologies.31

    The drug was most effective in reducing orabolishing clonus and somewhat less effica-cious in decreasing the resistance to stretchand the tendon reflexes. These authors re-ported that gait was improved and the pa-tients found it easier to take care of theirpersonal needs and in general, motor perfor-mance was improved.31 Of interest is a studyof enteral administration by Meyler, in which25 patients with spasticity underwent evalu-ation of the pharmacokinetics and effects ofdantrolene sodium after prolonged adminis-tration. The results were better on 100 mgdaily than on a higher daily dose and an in-crease of the daily dose from 200 to 400 mg

    was not associated with higher blood levels.32

    Weis has evaluated the differences in pharma-cokinetics between enteral and intravenousadministration of dantrolene.33 Thus changesin dosages are appropriate when intravenousroute administration is considered and theauthors administer it based on the patientsweight.

    In the acute care setting, enteral treatmentoften begins with low-dose therapy via na-sogastic tube or percutaneous gastrostomy.

    Doses begin at 25 to 50 mg and increase by25 to 50 mg every 4 to 5 days. Dantrolenecarries the advantage of minimal cognitiveside effects and only modest pharmacologicinteractions. The authors have occasionallyemployed the use of intravenous dantrolenetherapy for severe spasticity in persons withTBI; however, this should be done with greatcaution. Dantrolene therapy has been asso-ciated with hepatic toxicity.34 This is espe-

    cially true, when other agents that may facili-tate hepatotoxicity and careful monitoring ofliver function profiles are warranted. Durham

  • 8/3/2019 zafonte_tbi

    6/12

    94 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    has reported that the hepatotoxicity is notrelated to biotransformation issues,34 whileChan has discussed that those at highest riskare females older than 40 years and receiv-

    ing more than 300 mg of dantrolene.35

    Severaldrugs may impact the effects of dantroleneGilman has reported that metochlorpromideincreases the bioavailability of dantrolene,36

    while Chou has discussed that dantrolene in-hibits platelet activation mainly due to sup-pression of phosphoinositide breakdown.37

    This could be a concern in the post multi-trauma patient where bleeding is an issue.

    Despite its limitationsside effectstheauthors consider dantrolene to be the first-line pharmacologic agent for the treatmentof severe spasticity resulting from TBI. Inthe acute care setting dantrolene is likely ap-propriate in those with severe or malignantspasticty.

    Baclofen (Lioresal)

    Baclofen is active at the GABA b receptorboth presynaptically and postsynaptically. Ithas been employed for the treatment of spas-

    ticity in persons with spinal cord injury (SCI),cerebral palsy, multiple sclerosis, stroke, andbrain injury. Its utility in the treatment of spas-ticity in the acute care setting has not beenwell evaluated.

    Baclofen has been noted to produce seda-tion, especially among those with cerebral dis-orders. Concerns including asthenia, depres-sion, as well as altered attention and memoryadditional concerns have been raised in ani-mal models and observed with humans. GABA

    a facilitatory agents may impair recovery.38

    Wallace et al have reported that agents thatact postsynaptically at the GABA receptor canbe associated with impaired functional recov-ery in an animal model.39 The phenomenonhas not been evaluated in a model utilizing in-trathecal delivery systems. Memory dysfunc-tion has also been associated with baclofentherapy in both animals and humans.40,41

    Additional concerns with baclofen include

    lowered seizure threshold and withdrawalsyndrome.42 Of particular concern in theacute care setting is the fact that baclofen

    can increase bronchoconstriction and inhibitthe cough reflex. Such responses may makeventilatory weaning more challenging, espe-cially in those with a history of respiratory

    compromise.The majority of studies demonstrating the

    efficacy of baclofen have been conducted inpersons with SCI and multiple sclerosis.43 Ba-clofen appears effective at improving flexorspasms among those with spinal cord dysfunc-tion, yet little data exist to show improved ac-tivities of daily living or ambulatory capacity.Among those with spasticity of cerebral originlittle efficacy has been demonstrated. How-ever, enteral baclofen therapy may be war-ranted for those with severe spasticity whocannot tolerate dantrolene sodium and facethe risk of severe contracture or loss of rangeof motion.

    Early intrathecal baclofen (ITB) therapy hasnot been evaluated. ITB therapy has beengenerally considered only after 6 months ofinjury.44 Several studies have demonstratedthe efficacy of ITB in cerebral spasticity.4547

    This treatment has also been noted to

    have beneficial effects on sphincter function,sleep, and voluntary movement. In those withsevere spasticity who are not responding toother treatments, perhaps earlier intervention with ITB therapy is warranted. While issuesregarding the role of baclofen, GABA, andneurorecovery must be weighed, ITB treat-ment may be preferable to long-term contrac-tures and its associated complications. Theseissues are often made more difficult becausemany such patients have concomitant infec-

    tion making surgical implantation a difficultchoice. In such cases our own experience hasconsidered the role of intermittent bolus ther-apy or delivery via lumbar catheter to helpmaintain range of motion.

    Diazepam (Valium)

    Diazepam is a benzodiazepine and its ac-tion is mediated via the GABA a receptorand appears to increase presynaptic inhibi-

    tion of polysynaptic and monosynaptic re-flexes. These agents may be administered en-terally or via intravenous administration. Since

  • 8/3/2019 zafonte_tbi

    7/12

    Acute Care Management of Post-TBI Spasticity 95

    the action of this medication is also mediatedthrough the GABA system in a similar fashionto baclofen, concerns are raised regarding po-tential negative impact on neurorecovery.48

    This agent can produce sedation and impairedcognitive processing; intoxication with thisagent can produce somnolence and evencoma.

    The majority of data in support of this agentexists among those with spasticity of spinalorigin.49 In a study of persons with cerebralspasticity ambulation speed and grip strengthdeteriorated. In the acute care setting thisagent may have value as a temporizing agentamong those with severe spasticity and devel-oping contracture.

    Medications in the intensive care setting

    Propofol is an intravenous anesthetic agenthaving anticonvulsant property. It has uniquelipophilic properties and is ultra rapidlycleared after discontinuation. This agent pro-vides sedation that can be ultra rapidly clearedfrom the system. It has been used to treatmyoclonus that appears after therapeutic hy-

    pothermia treatment in acute TBI and canbe employed to temporarily treat severe spas-ticity while additional agents are employed.However, Cremer et al have noted that 5 pa-tients with TBI who had received propofolhad experienced cardiac arrest and may haveexperienced propofol infusion syndrome.50

    That work has suggested that propofol infu-sion at rates higher than 5 mg/kg per hourshould be discouraged for long-term sedationin the ICU. In the most severe cases neuropar-

    alytic agents can be considered. Midazolamis a unique water-soluble benzodiazepine thathas action at the GABA and opioid receptors.Dahm et al have reported the use of intrathe-cal midazolam for spasticity.51 Of note, mida-zolam therapy if given as a continuous intra-venous infusion may carry a specific benefitfor those with pain as an exacerbating fac-tor to their severe spasticity.51 Neurorecoveryand cognitive related issues remain with the

    use of midazolam.Rossier et al have reported on the role of

    magnesium in the treatment of severe spas-

    ticity in a woman with paraplegia.52 Magne-sium may also have anticonvulsant effects andhas a potential role in ameliorating a portionof the acute excitotoxic process after TBI via

    its action as an N-methyl-D-aspartate (NMDA)antagonist. Morphine is a water-soluble opi-oid characterized by its slow absorption. Ithas substantial tone relieving properties andhas been used via intrathecal route for spastic-ity. Morphine can however produce respira-tory depression and hypotension.53 Many pa-tients with the most severe forms of acutespasticty following TBI also have marked auto-nomic dysfunction. When medically feasibleour own experience has noted that the treat-ment of such patients with lipophilic betablockers often results in a substantial declinein Ashworth score and improvement in dysau-tonomic symptoms.

    Imidazolines

    Both clonidine (Catapress) and tizanidine(Zanaflex ) act as agonists at the alpha-2adrenergic receptor, which inhibits excitatoryamino acid release. They also facilitate the ac-

    tion of glycine, an inhibitor neurotransmitter.While clonidine is the older of the two agents,its clinical efficacy has been studied primar-ily in the spinal cord injury population.5456

    However, in a case series, Dall and colleaguesreported good results in the control of cere-bral spasticity with clonidine.57 Peak plasmalevels of clonidine occur at 3 to 5 hours af-ter oral administration, and its half-life is 5 to19 hours. The starting dose can be 0.1 mga day. Clonidines transdermal form provides

    an additional effective option for medica-tion delivery.58,59 Studies comparing tizani-dine with baclofen suggest equal or better ef-ficacy in control of spasticity in persons withmultiple sclerosis and strokes.60,61 A doubleblind, placebo-controlled trial in brain injuredpatients demonstrated tizanidines success intreating spasticity in this population.62 Thestarting dose is 2 to 4 mg at bedtime, increas-ing to a maximum of 36 mg per day. Tizani-

    dines peak effect is in 1 to 2 hours, and itshalf-life is 2.5 hours. Both clonidine and tizani-dine carry significant side effects of dizziness,

  • 8/3/2019 zafonte_tbi

    8/12

    96 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    drowsiness, and hypotension. Some careshould be given in the consideration of us-ing these medications; animal research63 andstudies in human stroke survivors64 suggest

    alpha-2 agonists may slow recovery from braininsult. Thus in the authors opinion the roleof these agents in the acute care setting islimited.

    Gabapentin (Neurontin)

    While its mechanism of action is unclear,the anticonvulsant gabapentin has been sug-gested for use as an antispastic agent. In stud-ies of persons with multiple sclerosis6567 andspinal cord injury,68,69 gabapentin was effec-tive in reducing spasticity with minimal ef-fects on cognition. This agents pain control-ling effects may play a role in its ability toimprove spasticity. It has also been used forthe treatment of dementia-related agitation70

    and the mentally ill71; however, Childers andHolland72 reported 2 cases of increased agi-tation during its use in TBI patients. The ef-fective dose for the treatment of spasticity is400 mg 3 times a day or higher, to a maxi-

    mum of 3600 mg a day total. Since this agentis mostly metabolized via the renal system itmay be helpful in those with hepatic dysfunc-tion or those where multiple drug interactionsare a concern. Gabapentin is limited by thefact that no intravenous preparation exists;however, it may also be a useful choice inthose with seizures who need anticonvulsanttherapy.

    Neurolytic therapy

    Cognitive side effects or tolerance issueslimit many systemic therapies. Thus in thosewith severe spasticity a reasonable portfolioof therapy might turn to interventional proce-dures, especially when it is focal. Phenol canbe delivered via indirect mixed nerve block-ade or via motor point block.73 Phenol via per-ineural injection causes a denaturation of pro-tein. The cost of this agent is minimal. It hasimmediate anesthetic effects and it impacts all

    neural fibers. Concerns include sensory loss,dysestheias, and vascular complications.74,75

    In the acute care setting perineural injection

    in the treatment of lower extremity adductorand elbow flexor tone may offer the advan-tage of easy access, reasonable positioning,and immediate onset. If the patient can toler-

    ate prolonged side lying then the tibial nervecan also be accessed. In experienced handsphenol can be an extremely effective treat-ment for focal spasticity and helps to maintainfunctional range in the acute care setting.75

    Phenol chemoneurolysis can also be used tofacilitate the application of a serial casting pro-gram. The duration of action varies with prox-imity of delivery and concentration of phenol.In some cases duration of action of greaterthan 1 year is not desired and may cause con-cern. Alcohol at low concentrations acts as alocal anesthetic and at higher concentrationsit appears to cause degeneration of neuronsand fibrosis.75 Pain, vascular complications,and skin discomfort have also been reportedwith the use of alcohol.

    Chemoneurolysis with Botulinum toxintherapy is now available as 2 differentserotypes: Botulinum toxin A and B.76 Theseagents have been approved for the treatment

    of dystonia but have been employed success-fully for the treatment of focal spasticity asan off-label use.7682 Development of futureserotypes may allow for varying duration ofaction that may show benefit for the intensivecare setting. Toxins are reasonable to delivereven in the acute care setting and have theadvantage of a relatively short duration of ac-tion. Of concern is the potential interaction with neuroparalytic agents and aminioglyco-side antibiotics. While cost has been cited as

    a reason not to utilize these agents during anacute inpatient stay, further evaluation mayprove that they are cost-efficacious in prevent-ing secondary complications. Phenol and al-cohol have a very quick, almost immediate,onset of action, which is not the case withthe toxins where weeks may pass prior tofull treatment effect.83 Another serious prob-lem is the ceiling effect of safe administra-tion of this agent. This is a specific concern

    when there is widespread tone. Botulinumtoxin can only be one of the tools used in themanagement of spasticity and will need to be

  • 8/3/2019 zafonte_tbi

    9/12

    Acute Care Management of Post-TBI Spasticity 97

    supplemented with phenol chemoneurolysisand/or systemic medications.

    Case discussion

    Our patients tone continued to worsen despiteaggressive range of motion and splinting ther-apy. Propofol therapy was weaned to allow for in-creased arousal and respiratory independence. Atrial of dantrolene therapy was begun and carefulmonitoring of hepatic enzymes were undertaken.Given the extent of the patients brain injury it waschosen to avoid baclofen as well as immediate im-idazoline therapy. The team also chose to closelymonitor the patients magnesium and keep the pa-tient at the higher end of normal magnesium level.He was placed on beta blocker therapy to ame-liorate severe dysautnomia, resulting in an as-sociated amelioration of the posturing episodes.Tone appeared to improve to Ashworth 2 in theright lower extremity, but adductor tone remained

    severe bilaterally as did right elbow flexor tone.During the course of treatment the patient hadrecently received aminoglycoside antibiotic ther-apy secondary to sepsis. Thus, we chose to per-form phenol neurolytic blockade of the obturatornerves and resulted in marked decline in adduc-tor tone. A phenol motor point block of the right

    biceps was undertaken and produced a reductionin right elbow flexor tone. This therapy was com-plemented by the addition of a right elbow serialcast.

    Fortunately over the next year the patient pro-gressed to recover cognitively and regained sig-nificant motor function in his right side. While no

    one can be sure, we believe that early aggressiveintervention was helpful in preventing secondarycomplications.

    CONCLUSION AND FUTURE DIRECTION

    The management of spasticity in the acutecare unit remains problematic. There is only very limited research to guide clinicians intheir decision process. Thus each case is anindividual decision making process. We haveknowledge concerning potential limitationsconcerning our early intervention efforts, butonly limited knowledge of what is efficacious.Much work still needs to be done to develop

    interventions with proven efficacy and ac-ceptable risk/benefit ratios. Progressive lossof range of motion and the development ofcontractures may impel the clinician to utilizemedications that could have potentially nega-tive consequences on recovery. ITB adminis-tration performed early on may prevent con-tractures and viscoelastic changes with onlylimited risks to motor recovery because ofthe low cerebral concentrations. Finally, re-

    search addressing the potential to increase theefficacy of botulinum toxin injections withincreased dilution or endplate localization,which may increase the amount of spasticitythat can be treated, as well as lowering costmay increase its utility as a spasticity interven-tion in the acute care environment.

    REFERENCES

    1. Wagner A, Fabio T, Zafonte R, Goldberg G, MarionD, Peitzman A. Physical medicine and rehabilitation

    consultation: relationship with functional outcome,

    length of stay, and discharge planning after traumatic

    brain injur y. Am J PMR. 2003;82:526536.

    2. Hinderer S, Dixon K. Physiologic and clinical moni-

    toring of spastic hypertonia. Phys Med Rehabil Clin

    N Am. 2001;12:733746.

    3. Dalyan, M, ShermanA, Cardenas D. Factors associated

    with contractures in acute spinal cord injury. Spinal

    Cord. 1998;36:405408.

    4. Keren O, Reznik J, Grosswasser Z. Combined mo-

    tor disturbances following severe traumatic brain in-

    jury: an integrative long-term approach. Brain Inj.2001;15:633638.

    5. Mizuno J, Sugimoto S, Tsutsui T, Machida K. Effi-

    cacy of propofol in controlling myoclonus duringrewarming in a brain hypothermia patient. Masui.

    2002;15:781790.

    6. Starring D, Grossman M, Nicholson G, et al. Compari-

    son of cyclic and sustained passive stretching using a

    mechanical device to increase resting length of ham-

    string muscles. Phys Ther. 1988;68:314.

    7. Tremblay F, Malouin F, Richards C, et al. Effects of pro-

    longed muscle stretch on reflex and voluntary mus-

    cle activations in children with spastic cerebral palsy.

    Scand J Rehabil Med. 1990;22:171.

    8. Singer BJ, Singer KP, Allison GT. Evaluation of extensi-

    bility, passive torque and stretch reflex responses in

    triceps surae muscles following serial casting to cor-rect spastic equinovarus deformity. Brain Inj. April

    2003;17(4):309324.

  • 8/3/2019 zafonte_tbi

    10/12

    98 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    9. King TI II. Plaster splinting as a means of reducing el-

    bow flexor spasticity: a case study.Am J Occup Ther.

    October 1982;36:671673.

    10. Knutsson E. Topical cryotherapy in spasticity. Scand

    J Rehabil Med. 1970;2(4):159163.

    11. Miglietta O. Action of cold on spasticity. Am J PhysMed. August 1973;52:198205.

    12. Childers MK, Biswas SS, Petroski G, Merveille O. In-

    hibitory casting decreases a vibratory inhibition in-

    dex of the H-reflex in the spastic upper limb. Arch

    Phys Med Rehabil. June 1999;80:714716.

    13. Singer BJ, Jegasothy GM, Singer KP, Allison GT. Eval-

    uation of serial casting to correct equinovarus de-

    formity of the ankle after acquired brain injury in

    adults. Arch Phys Med Rehabil. April 2003;84:483

    491.

    14. Otis JC, Root L, Kroll MA. Measurement of plantar

    flexor spasticity during treatment with tone-

    reducing casts. J Pediatr Orthop. November/

    December 1985;5:682686.

    15. Pohl M, Ruckriem S, Mehrholz J, Ritschel C, Strik H,

    Pause MR. Effectiveness of serial casting in patients

    with severe cerebral spasticity: a comparison study.

    Arch Phys Med Rehabil. June 2002;83:784790.

    16. Pohl M, Mehrholz J, Ruckriem S. The influence of ill-

    ness duration and level of consciousness on the treat-

    ment effect and complication rate of serial casting in

    patients with severe cerebral spasticity. Clin Reha-

    bil. July 2003;17:373379.

    17. Barnard P, Dill H, Eldredge P, Held JM, Judd DL,

    Nalette E. Reduction of hypertonicity by early castingin a comatose head-injured individual. A case report.

    Phys Ther. October 1984;64:15401542.

    18. Mills VM. Electromyographic results of inhibitory

    splinting. Phys Ther. February 1984;64:190193.

    19. Lannin NA, Horsley SA, Herbert R, McCluskey A,

    Cusick A. Splinting the hand in the functional po-

    sition after brain impairment: a randomized, con-

    trolled trial. Arch Phys Med Rehabil. February

    2003;84:297302.

    20. Gracies JM, Marosszeky JE, Renton R, Sandanam J,

    Gandevia SC, Burke D. Short-term effects of dynamic

    lycra splints on upper limb in hemiplegic patients.

    Arch Phys Med Rehabil. December 2000;81:15471555.

    21. Gracies JM, Fitzpatrick R, Wilson L, Burke D,

    Gandevia SC. Lycra garments designed for patients

    with upper limb spasticity: mechanical effects in

    normal subjects. Arch Phys Med Rehabil. October

    1997;78:10661071.

    22. Bajd T, Gregoric M, Vodovnik L, Benko H. Electri-

    cal stimulation in treating spasticity resulting from

    spinal cord injury. Arch Phys Med Rehabil. August

    1985;66:515517.

    23. Dewald JP, Given JD, Rymer WZ. Long-lasting re-

    ductions of spasticity induced by skin electricalstimulation. IEEE Trans Rehabil Eng. December

    1996;4:231242.

    24. Seib TP, Price R, Reyes MR, Lehmann JF. The quanti-

    tative measurement of spasticity: effect of cutaneous

    electrical stimulation. Arch Phys Med Rehabil. July

    1994;75:746750.

    25. Scheker LR, Chesher SP, Ramirez S. Neuromuscular

    electrical stimulation and dynamic bracing as a treat-ment for upper-extremity spasticity in children with

    cerebral palsy.J Hand Surg (Br). April 1999;24:226

    232.

    26. Langeron O, Coirault C, Fratea S, Orliaguet G,

    Coriat P, Riou B. The effects of dantrolene on the con-

    traction, relaxation, and energetics of the diaphragm

    muscle. Anesth Analg. August 1999;89:466471.

    27. Elovic E. Priciples of pharmaceutical management of

    spastic hypertonia. Phys Med Rehabil Clin N Am.

    2001;12:793816.

    28. Cho S, Zhang S, Ureshino H, Hara T, Tomiyasu S,

    Sumikawa K. Hemodynamic interactions of propo-

    fol and dantrolene in chronically instrumented dogs.

    Anesth Analg. 2003;96(5):13691373.

    29. Nakayama R, Yano T, Ushijima K, Abe E, Terasaki E.

    Effects of dantrolene on extracellular glutamate con-

    centration and neuronal death in the rat hippocam-

    pal CA1 region subjected to transient ischemia.Anes-

    thesiology. March 2002;96:705710.

    30. Popescu BO, Oprica M, Sajin M, et al. Dantrolene

    protects neurons against kainic acid induced apop-

    tosis in vitro and in vivo. J Cell Mol Med. Octo-

    ber/December 2002;6:555569.

    31. Seinberg F, Ferguson K. Effect of dantolene sodium

    on spasticity associated with hemiplegia. J Am Geri-atr Soc. 1975;23:7073.

    32. Meyler W, Mols-Thurkow H, Wesseling H. Relation-

    ship between plasma concentration and effect of

    dantrolene sodium in man. Eur J Clin Pharmacol.

    1979;16:203209.

    33. Weis EW, Vree TB, Van Der Kleijn E. Pharmacokinet-

    ics of intravenously administered dantrolene and its

    5-hydroxy metabolite in dogs. Int J Clin Pharmacol

    Res. 1990;10(4):203210.

    34. Durham JA, Gandolfi AJ, Bentley JB. Hepatotoxico-

    logical evaluation of dantrolene sodium. Drug Chem

    Toxicol. 1984;7(1):2340.

    35. Chan C. Dantrolene sodium and hepatic injury. Neu-rology. 1990;40;14271432.

    36. Gilman T, Segal J, Brunnemann S. Metoclopramide in-

    creases the bioavaliability of dantolene in spinal cord

    injury. J Clin Pharmacol. 1996;36:6471.

    37. Chou TC, Li CY. Inhibitory mechanisms of dantro-

    lene on platelet aggregation. Thromb Res. November

    1999;96:299307.

    38. Goldstein LB. Neuropharmacology of TBI-induced

    plasticity. Brain Inj. August 2003;17:685694.

    39. Wallace A, Kline A, Montanez S, Hernandez T. Imapct

    of the novel anticonvulsant vigabatrin on functional

    recovery following brain lesion. Restor Neurol Neu-rosci. 199;14:3545.

    40. Sandyknb R, Gillman M. Balcofen induced memory

  • 8/3/2019 zafonte_tbi

    11/12

    Acute Care Management of Post-TBI Spasticity 99

    impairment. Clin Neuropharmacology. 1885;8:

    294295.

    41. Castellano C, Brioni J, Nagahara A, McGaugh J. Post-

    training systemic and intra amygdla administration of

    the GABA B agonist baclofen impairs retention Be-

    hav Neural. 1989;52:170179.42. Terrence C, FrommG, Roussan M. Baclofen its effect

    on seizure frequency. Arch Neurol. 1983;40:2829.

    43. Taricco M, Adone R, Pagliacci C, Telaro E. Phar-

    macological interventions for spasticity following

    spinal cord injury. Cochrane Database Syst Rev.

    2000;(2):CD001131.

    44. Coffey E, Edgar T, Francisco G, et al. Abrupt with-

    drawal from intrathechal baclofen: recogntion and

    management of potentially life threatening syn-

    drome. Arch Phys Med Rehabil. 2002;83:735741.

    45. Meyhtaler J, Guin-Renfroe S, Grabb P, Hadley M.

    Long term continuously infused intrathecal baclofen

    for spastic dystonia hypertonia in trumatic brain in-

    jury: 1-year experience. Arch Phys Med Rehabil.

    1999;80:1319.

    46. Albright A, Gilmartin R, Swift D, Krach L, Ivan-

    hoe C, McLaughlin J. Long term intrathecal baclofen

    therapy for severe spasticity of cerebral origin.J Neu-

    rosurg. 2003;98:291295.

    47. Gertzen P, Albright A, Barry M. Effect on ambulation

    of continous intrathecahl baclofen infusion Pediatr

    Neurosurg. 1997;27:4044.

    48. ODellDM, Gibson CJ, Wilson MS, DeFord SM, Hamm

    RJ. Positive and negative modulation of the GABA(A)

    receptor and outcome after traumatic brain injury inrats. Brain Res. April 2000;861:325332.

    49. Corbett M, Frankel H, Michaelis L. A double blind

    cross over trial of valium in the treatment of spas-

    ticity. Paraplegia. 1972;10:19.

    50. Cremer OL, Moons KG, Bouman EA, Kruijswijk JE,

    de Smet AM, Kalkman CJ. Long-term propofol in-

    fusion and cardiac failure in adult head-injured pa-

    tients. Lancet. January 2001;357:117118.

    51. Dahm LS, Beric A, Dimitrijevic MR, Wall PD. Di-

    rect spinal effect of a benzodiazepine (midazo-

    lam) on spasticity in man. Stereotact Funct Neuro-

    surg. 1989;53(2):8594. Eur J Neurol. November

    2000;7:741744.52. Rossier P, van Erven S, Wade DT. The effect of mag-

    nesium oral therapy on spasticity in a patient with

    multiple sclerosis. Eur J Neurol. 2000;7:741744.

    53. Soni B, Mani R, Oo T, Vaidyanathan S. Treat-

    ment of spasticity in a spinal cord injured patient

    with intrathecal baclofen tolerance. Spinal Cord.

    2003;41:586589.

    54. Donovan WH, Carter RE, Rossi CD, Wilkerson MA.

    Clonidine effect on spasticity: a clinical trial. Arch

    Phys Med Rehabil. March 1988;69(3 pt 1):193194.

    55. Nance PW. A comparison of clonidine, cyprohepta-

    dine and baclofen in spastic spinal cord injured pa-tients. J Am Paraplegia Soc. July 1994;17:150156.

    56. Maynard FM. Early clinical experience with clonidine

    in spinal spasticity. Paraplegia. June 1986;24:175

    182.

    57. Dall JT, Harmon RL, Quinn CM. Use of clonidine for

    treatment of spasticity arising from various forms

    of brain injury: a case series. Brain Inj. June

    1996;10:453458.58. Weingarden SI, Belen JG. Clonidine transdermal sys-

    tem for treatment of spasticity in spinal cord injury.

    Arch Phys Med Rehabil. September 1992;73:876

    877.

    59. Yablon SA, Sipski ML. Effect of transdermal clonidine

    on spinal spasticity. A case series. Am J Phys Med

    Rehabil. June 1993;72:154157.

    60. Groves L, Shellenberger MK, Davis CS. Tizanidine

    treatment of spasticity: a meta-analysis of controlled,

    double-blind, comparative studies with baclofen

    and diazepam. Adv Ther. July/August 1998;15:241

    251.

    61. Medici M, Pebet M, Ciblis D. A double-blind, long-

    term study of tizanidine (Sirdalud) in spasticity due

    to cerebrovascular lesions. Curr Med Res Opin.

    1989;11(6):398407.

    62. Meythaler JM, Guin-Renfroe S, Johnson A, Brunner

    RM. Prospective assessment of tizanidine for spastic-

    ity due to acquired brain injury. Arch Phys Med Re-

    habil. September 2001;82:11551163.

    63. Feeney DM, Westerberg VS. Norepinephrine and

    brain damage: alpha noradrenergic pharmacology al-

    ters functional recovery after cortical trauma. Can J

    Psychol. June 1990;44:233252.

    64. Goldstein LB. Common drugs may influence motorrecovery after stroke. The Sygen In Acute Stroke

    Study Investigators. Neurology. May 1995;45:865

    871.

    65. Cutter NC, Scott DD, Johnson JC, Whiteneck G.

    Gabapentin effect on spasticity in multiple sclero-

    sis: a placebo-controlled, randomized trial.Arch Phys

    Med Rehabil. February 2000;81:164169.

    66. Dunevsky A, Perel AB. Gabapentin for relief of spas-

    ticity associated with multiple sclerosis. Am J Phys

    Med Rehabil. September/October 1998;77:451

    454.

    67. Mueller ME, Gruenthal M, Olson WL, Olson WH.

    Gabapentin for relief of upper motor neuron symp-toms in multiple sclerosis. Arch Phys Med Rehabil.

    May 1997;78:521524.

    68. Gruenthal M, Mueller M, Olson WL, Priebe MM,

    Sherwood AM, Olson WH. Gabapentin for the treat-

    ment of spasticity in patients with spinal cord injury.

    Spinal Cord. October 1997;35:686689.

    69. Priebe MM, Sherwood AM, Graves DE, Mueller M,

    Olson WH. Effectiveness of gabapentin in controlling

    spasticity: a quantitative study. Spinal Cord. March

    1997;35:171175.

    70. Miller LJ. Gabapentin for treatment of behavioral and

    psychological symptoms of dementia. Ann Pharma-cother. April 2001;35:427431.

    71. Megna JL, Devitt PJ, Sauro MD, Dewan MJ.

  • 8/3/2019 zafonte_tbi

    12/12

    100 JOURNAL OF HEAD TRAUMA REHABILITATION/MARCHAPRIL 2004

    Gabapentins effect on agitation in severely and

    persistently mentally ill patients. Ann Pharma-

    cother. January 2002;36:1216.

    72. Childers MK, Holland D. Psychomotor agitation

    following gabapentin use in brain injury. Brain Inj.

    July 1997;11:537540.73. McGuire JR. Effective use of chemodenervation

    and chemical neurolysis in the management of

    poststroke spasticity Top Stroke Rehabil. Spring

    2001;8:4755.

    74. Albert TA, Yelnik A, Bonan I, Lebreton F, Bussel B.

    Effectiveness of femoral nerve selective block in pa-

    tients with spasticity: preliminary results. Arch Phys

    Med Rehabil. 2002;83:692696.

    75. Zafonte RD, Munin MC. Phenol and alcohol blocks

    for the treatment of spasticityPhys Med Rehabil Clin

    N Am. 2001;12:817832.

    76. Barnes M. Botulinum toxinmechanisms of ac-

    tion and clinical use in spasticity. J Rehabil Med.

    2003;35:9899.

    77. Ward AB, Aguilar M, De Beyl Z, et al. Use of

    botulinum toxin type A in management of adult

    spasticitya European consensus statement.J Reha-

    bil Med. 2003;35:9899.

    78. Brashear A, McAfee AL, Kuhn ER, Ambrosius WT.

    Treatment with botulinum toxin type B for upper-

    limb spasticity. Arch Phys Med Rehabil. 2003;84:

    103107.

    79. Richardson D, Sheaan G, Werring D, et al. Evaluating

    the role of botulinum toxin in the management offocal hypertonia in adults. J Neurol Neurosurg Psy-

    chiatry. 200;69:499504.

    80. Smith S, Ellis E, White S, et al. A double blind placebo

    controlled study of botulinum toxin in upper limb

    spasticity after stroke or head injury. Clin Rehabil.

    2000;14:513.

    81. Brashear A, Gordon M, Elovic E, et al. A double blind

    randomized placebo controlled study of botulinum

    toxin type A (BOTOX) in the treatment of upper limb

    spasticity following stroke. NEJM. 2002.

    82. Francisco GE, Boake C, Vaughn A. Botulinum toxin

    in upper limb spasticity after acquired brain injury:

    a randomized trial comparing dilution techniques.

    Arch Phys Med Rehabil. 2002;81:355363.

    83. Glenn M, Elovic E. Chemical denervation for the

    treatment of hypertonia and related disorders: phe-

    nol and botulinum toxin. J Head Trauma Rehabil.

    1997;12:40.