5 Clinical Results in Brain Injury Trials - Another Perspective

Embed Size (px)

Citation preview

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    1/19

    333

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    Copyright 2015 Undersea & Hyperbaric Medical Society, Inc.

    Clinical results in brain injury trials using HBO2therapy:

    Another perspective

    Xavier A. Figueroa, Ph.D.1, James K. Wright, M.D.2

    1 The Brain Health & Healing Foundation, Seattle, Washington U.S.

    2 Swedish Medical Center-Edmonds, Wound Healing & Hyperbarics, Edmonds, Washington U.S.

    CORRESPONDINGAUTHOR: Xavier Figueroa [email protected]

    ______________________________________________________________________________________________________________________________________________________

    ABSTRACT

    ______________________________________________________________________________________________________________________________________________________

    The current debate surrounding the use of hyperbaric oxy-

    gen (HBO2) for neurological indications, specically mild

    to moderate chronic traumatic brain injury (mTBI) and

    post-concussion syndrome (PCS), is mired in confusion

    due to the use of non-validated controls and an unfamil-

    iarity by many practitioners of HBO2 therapy with the

    experimental literature. In the past 40 years, the use of

    an air sham (21% oxygen, 1.14-1.5 atmospheres absolute/

    atm abs) in clinical and animal studies, instead of obser-

    vational or crossover controls, has led to false acceptance

    of the null hypothesis (declaring no effect when one is

    present), due to the biological activity of these sham

    controls. The recent Department of Defense/Veterans

    Administration (DoD/VA) sponsored trials, previous

    published reports on the use of HBO2therapy on stroke

    and mTBI and preliminary reports from the HOPPS

    Army trial, have helped to highlight the biological ac-

    tivity of pressurized air, validate the development of a

    convincing control for future studies and demonstrate

    the effectiveness of a hyperbaric intervention for mTBI/

    PCS. Approval of HBO2 for neurological indications,

    especially for mTBI/PCS, should be granted at the federal,

    state and certifying body levels as a safe and viable

    treatment for recovery in the post-acute phase.

    _______________________________________________________________________________________________________________________

    KEYWORDS: traumatic brain injury, hyperbaric oxygen, cognitive function, TBI, HBO, HBO2,

    _______________________________________________________________________________________________________________________

    ABBREVIATIONS: amyloid precursor protein (APP); tissue plasminogen activator (tPA); traumatic brain injury (TBI); mild to mod-

    erate traumatic brain injury (mTBI); acquired brain injury (ABI); post-traumatic stress disorder (PTSD); post-concussion syndrome

    (PCS); single photon emission computed tomography (SPECT); diffusion tensor imaging (DTI); regional cerebral blood flow

    (rCBF); Rivermead Post-Concussion Symptom Questionnaire (RPQ); PTSD Checklist Military Version (PCL-M); Department of

    Defense (DoD); Veterans Administration (VA); standard of care (SoC); hyperbaric oxygen (HBO2); Hyperbaric Oxygen Therapy for

    Persistent Post-Concussive Symptoms (HOPPS). National Institutes of Health Stroke Scale (NIHSS); activities of daily living (ADL);

    evaluation of quality of life (EQ); evaluation by the EQ-5D descriptive system (EQ-5D); EQ visual analogue scale (EQ-VAS).

    INTRODUCTION

    The conclusions of the DoD/VA sponsored studies [1-

    4] on the benets of hyperbaric oxygen (HBO2) therapy

    for service members diagnosed with post-concussion

    syndrome have recommended ceasing the development

    of a promising therapy. The published conclusions of

    the DoD/VA sponsored studies recommend the view

    that there is no benet of HBO2for mild to moderate

    traumatic brain injury (mTBI) and post-concussion

    syndrome (PCS). The DoD/VA studies have added con-

    fusion to the debate on controls for HBO2trials and have

    failed to account for within-group pre- and post-treat-

    ment outcomes, biasing analysis and reporting towardthe conclusion that hyperbaric oxygen is ineffective

    for the treatment of symptoms of mTBI and PCS. The

    assumption that pressurized air is a sham intervention

    has led to erroneous conclusions of non-activity, spe-

    cically when the conclusions are due to comparison

    between sham and HBO2 treatment as the terminal

    decision point [5]. A review of the past literature will

    show that this has been a recurring problem and has

    led to incorrect conclusions of non-efcacy for HBO2.

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    2/19

    X.A. Figueroa, J.K. Wright

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    334

    The varied biological effects of both low- and high-

    pressure delivery of pure and mixed gases are substan-

    tial, with a large literature in experimental and applied

    therapies. The current evidence from cell culture,

    animal studies, case reports and clinical trials for stroke

    and mTBI treatment is consistent with a reparative

    effect of HBO2on the nervous system, including low-

    pressure air treatments. Furthermore, the overall safety

    of this intervention is not called into doubt. The use

    of HBO2 for mTBI or PCS should be sanctioned and

    given provisional approval under Centers for Medi-

    care and Medicaid Services (CMS) coverage with evi-

    dence guidelines, as well as for the VA and TRICARE.

    Biological effects of hyperbaric oxygen

    The human body is sensitive to both decreases and

    increases in oxygen levels, brought about by changesin total oxygen concentration and changes in absolute

    pressure. Any change in cellular oxygen concentration

    will have potential effects on cellular function. Indeed,

    both hypobaric and hyperbaric oxygen can induce pre-

    conditioning effects that can protect myocardial and

    neural tissue from damaging changes in oxygen tension

    [6-11]. HBO2 preconditioning has been used in coro-

    nary artery bypass graft surgery and for post-operative

    recovery [12-14], reducing cognitive impairments and

    neurological sequelae. The application of oxygen under

    pressure has been shown to induce reparative mechan-isms in a number of injury and disease induced con-

    ditions.

    Understanding the mechanisms that are activated

    by HBO2 is essential for applying this clinical tool

    correctly. In the last 30 years numerous physiological

    and cellular effects of HBO2 have been identied

    and elucidated. The effects are varied, occurring

    simultaneously, in many cases long-lasting and extensive

    in the responses that they elicit.

    Stem cell upregulation

    In the past 10 years, reports of increased circulating

    endothelial progenitor cells in mouse [21] and humans

    [22] have shed light on mechanisms stimulated by

    HBO2 for wound healing and angiogenesis [23]. In

    humans and mice, the process of increased stem cell

    release is a nitric oxide (NO) mediated process that

    is triggered by HBO2. Animal models of TBI have

    effectively shown that angiogenesis and neurogenesis

    are increased with HBO2in the injured hemisphere and

    are correlated with improvements in performance and

    recovery vs. the non-HBO2-treated [24,25]. In adult

    cerebral stroke models in rats, neurogenesis via stem

    cell migration was seen in the cortex, and the duration

    of HBO2 treatment was correlated with recovery of

    function [26]. Stem cell migration has been observed

    in wound healing clinical studies, ranging from diabetic

    foot ulcers [27] to skin wounds [28] that are treated by

    HBO2. When the injured brain of those suffering from

    PCS, TBI or stroke is considered as a type of wound,

    the data supporting a model of stem cell recruitment to

    damaged areas of the brain [29] becomes better under-

    stood.

    SummaryHow stem cell migration aids in the restoration of func-

    tion and symptom alleviation in mTBI is not fully un-

    derstood, as there are no direct studies linking these

    phenomena together in humans. Possible mechanisms

    are a restoration of the vascular network in damaged

    or low-ow areas of the brain and repair of dam-

    ages axons. Stem cell upregulation is a direct effect

    of HBO2 treatment and should be considered as a re-

    parative function for acquired brain injuries. HBO2

    has the untapped potential as a ready and accessible

    source for stem cell recruitment in future therapies.The mechanism of action remains to be elucidated,

    and further work should be done, including studies of

    the effect of varied pressures or oxygen concentrations

    on stem cell numbers in peripheral blood and tissues.

    Modulation of inammatory responses by HBO2

    One of the initial responses to a TBI is an increase in

    inammation to the brain (in animal models and in

    humans). HBO2 has been shown to effectively re-

    duce inammatory markers in blast- and blunt force

    trauma-induced TBI [30-33]. Mouse models of TBI

    have shown that head injuries induce widespread in-

    ammatory responses; HBO2 exposure can decrease

    apoptosis, reactive neuroinammation [33] and inhib-

    its microglial activation and production of pro-inam-

    matory cytokines [31]. New evidence suggests that

    mild to moderate TBI has a strong inammatory com-

    ponent that mediates acute and long-term injury [34].

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    3/19

    X.A. Figueroa, J.K. Wright

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    335

    The role of HBO2induced vasoconstriction is a well-

    known effect in reducing cerebral edema [35,36]. In

    clinical research with severe TBI study subjects, HBO2

    reduces swelling and intracranial pressure [37,38]

    as well as increasing survival. More recent work has

    shown that HBO2 reduces the rate and the number

    of inltrating neutrophils into injury areas [39-46].

    Another effect of HBO2 is its ability to modulate the

    sensation of pain. In rat models of neuropathic pain

    [47-49], HBO2was able to reduce the pain sensation;

    it appears to be a NO-mediated process in mice [48].

    In the case of severe traumatic brain injury, inam-

    matory markers have been detected months to years

    after the TBI incident in humans [50]. A lower level

    of inammation may be present in mTBI and PCS

    sufferers [34]. The recent discovery of -amyloid

    (1-42) and APP increasing after blast-induced braininjury in animal models [51], and severe brain injury

    in humans [52] and the increased risk of Alzheimers

    disease and cumulative traumatic encephalopathy with

    a history of head trauma [53-56], point to the role of a

    chronic inammatory response in mediating long-term

    damage in TBI and dementia of the Alzheimers type.

    Summary

    Inammation is a recognized component of TBIs of all

    types. HBO2 has well-established anti-inammatory

    capabilities in neuroinammation, suggesting that therole of HBO2in mTBI and PCS treatment extend much

    further than oxygenating tissues. Mixed approaches in

    controlling inammation, such as Omega-3 polyunsatu-

    rated fatty acid supplementation [57-63] may enhance

    recovery and alleviate symptoms of mTBI and PCS,

    with adjunct HBO2. Chronic administration of NSAIDs,

    specically ibuprofen, have negative correlations with

    TBI recovery in animal models [64], but inhibition of

    specic inammatory mediators have reparative effects

    [65-67].

    Cellular repair and protection

    Over 8000 known biochemical processes are dependent

    on oxygen in the atmosphere to carry out metabolic

    processes [68]. Changes in the concentration of oxygen

    in the ambient environment, hypoxic or hyperoxic, an

    increase due to an elevation in air pressure, supple-

    mental oxygen administration or hyperbaric oxygen

    will more than likely result in changes in cellular pro-

    cesses, some of which may be profound and long lasting.

    In the past few years there has been a signicant in-

    crease in the scientic understanding of how oxygen

    works on a cellular level. Many of these cellular pro-

    cesses are signaled by reactive oxygen species (ROS)

    [69-72] and others are signaled by oxygen itself [73-76].

    Initial exposures to HBO2can induce DNA strand breaks

    [77] and increased lipid peroxidation of cell mem-

    branes [78], but cellular defenses are activated that

    repair and protect from further damage [77]. Although

    concerns of overproduction are warranted, most organ-

    isms have a wide array of defenses to limit the over-

    production of oxygen radical species [79]. Cellular

    ROS activity is under tight control and is involved in

    cell signaling, protein trafcking and regulation of en-

    zyme, receptor, transporter and transcription factors[69]. ROS produced by HBO2 induce mitochondrial

    biogenesis [80] and may play a role in cell repair

    and protection from neurological injuries [81].

    Additionally each organelle within the cell, including

    mitochondria, has different ROS regulation character-

    istics that govern the function of each organelle.

    Mechanisms associated with repair processes, such as

    -integrin b1 receptors mediating collagen synthesis

    [69] and DNA methylation (which alters the expression

    of genes in cells as they divide and differentiate) affects

    gene transcription and are signaled by ROS [69]. ROShave a role in signaling histone acetylation and deacety-

    lation which are also key processes of gene expression

    [69] and repair processes. Hyperbaric oxygen adminis-

    tration alters the ROS in the cellular environment and

    its known mechanisms of action in cellular inamma-

    tion, repair and apoptosis regulation are activated [82].

    Derangements in ROS regulation and oxidative stress

    have been implicated in a variety of neurologic diseases,

    including TBI [69]. In TBI, oxidative stress plays a role

    in the pathology of neuronal dysfunction, while HBO2

    upregulates the neuronal repair processes required to

    restore function [83,84]. Furthermore, HBO2 upregu-

    lates anti-apoptotic mechanisms (such as the Bcl-2

    family of proteins) that protect neuronal cells from

    dying [85,86] and help maintain mitochondrial function

    [85,87,88]. While it is not known precisely how Bcl-2

    signals cellular repair via ROS, while hypoxia-induced

    ROS cause inammation and cellular degeneration and

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    4/19

    336

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    apoptosis, it is theorized that the changes in cellular

    oxygen levels, and hence ROS (termed oxygen cy-

    cling) cause a perceived hypoxia by the cell which

    in turn signals repair processes in the presence of ade-

    quate oxygen levels for the repair to proceed [84].

    It appears that it is not the actual level of oxygen

    present, but the difference in the homeostatic equilib-

    rium of oxygen that is sensed and the same ROS that

    are produced when the equilibrium swings too far from

    the set-point. The ROS act as signaling agents, inde-

    pendent of hypoxic, normoxic or hyperoxic states, but

    the context of the signaling (the actual level of oxygen)

    may mediate the activation of context specic mech-

    anisms that interpret the ROS signaling.

    Summary

    HBO2-produced ROS are mitigated by the cells abilityto quench ROS overproduction. HBO2activates numer-

    ous cellular mechanisms of action which are neuro-

    protective and assist the injured brain to heal. The

    effects are broad and protective at the cellular level,

    including protection of mitochondrial potential [85,87,

    88]. Furthermore, the anti-apoptotic proteins, Bcl-2 and

    Bcl-xL, are upregulated in neurons in contusion models

    of rats after HBO2exposure and the downstream effects

    that these proteins exert to inhibit apoptosis [89].

    Cellular oxygen utilization: mitochondrialregulation and idling neurons

    Disruption or reduction of mitochondrial activity in

    nervous tissue is a reported effect of stroke [90-92] and

    TBI [93]. In the case of TBI, the deregulation of mito-

    chondrial activity is complex and can lead to changes

    in metabolic function [37,93-96] and changes in blood

    ow [97]. HBO2 has shown the ability to restore

    mitochondrial activity in animal models of TBI [80,

    85,87,88,96,98,99]. In the case of blast-induced TBI,

    cerebral blood ow and metabolic function were

    shown to improve after HBO2in a rabbit model [30].

    The hypothesis of hypometabolic neural tissue in a

    penumbra or an area of idling neurons in brain in-

    jury was promulgated by Dr. Neubauer [100-102], but

    was considered untested, mostly due to a lack of an es-

    tablished mechanism for neural tissue entering into a

    state of hibernation. The evidence of neural tissue that

    can be rescued is supported by recovery of function

    (neurological testing, EEG or SPECT) in stroke studies

    that used HBO2 to detect brain tissue that was viable

    in post-acute stroke with 10-15 HBO2 treatments [103]

    or even a single HBO2 treatment [104]. Another case

    report of post-acute stroke patients treated with HBO2

    and imaged with single photon emission computer

    tomography (SPECT), reported improved neurological

    function [105] and increased regional cerebral blood

    ow (rCBF) in the penumbra of the affected region.

    The development of diffusion tensor imaging of the

    brain in concussion patients appears to validate the

    presence of a penumbra of non-functional damaged

    axons after injury [106]. These studies help to further

    validate the improved outcomes (National Institutes

    of Health Stroke Scale, NIHSS; activities of daily

    living, ADL; evaluation of quality of life, EQ) of

    the recent post-acute stroke recovery study usingHBO2 by Efrati, et al. [107] and buttress the case

    for reactivation of hypometabolic neural tissue.

    The landmark studies of Blackstone, et al. [108,109]

    were the rst to provide a plausible mechanism for

    idling neurons. Blackstone, et al. demonstrated the

    ability to downregulate mitochondrial respiration by

    simultaneously exposing mice to gaseous H2S and re-

    ducing oxygen levels. Not only did the mice survive

    periods of reduced oxygen tension of > 6 hour at 5%

    oxygen, there was no measurable neurological or myo-

    cardial damage when they were restored to normal oxy-gen levels and metabolic function [108, 109]. The exact

    mechanism of neural tissue protection remains unestab-

    lished, but the principle is present for future investiga-

    tion. Furthermore, the discovery of a hydrogen sulde

    (H2S) mechanism modulating long-term potentiation

    (LTP) in neurons [110] suggests that this signaling gas

    is an integral and conserved mechanism in mammals

    [111-115]. From a mechanism perspective, enzymatic

    systems in mammalian neural tissues produce H2S

    locally and could induce a local hypometabolic state by

    direct regulation of mitochondrial activity [116-120].

    Summary

    The hypothesis that neurons can remain viable but not

    electrically active after a traumatic brain injury is sup-

    ported by SPECT imaging of TBI patients in the post-

    acute phase. Individuals diagnosed with a TBI pres-

    ent with reduced rCBF [121,122] when compared to a

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    5/19

    337

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    healthy, non-injured population. Diffusion tensor im-

    aging in traumatic brain injury provides an anatomic

    picture of idling neurons with loss of axonal integrity

    demonstrable for years after injury [123]. The effects

    of axonal remyelination after chronic TBI using HBO2

    saw a signicant restoration of axonal tracts and motor

    improvements in rats [124]. Restoration or elevation of

    rCBF is seen with HBO2 treatment of TBI study par-

    ticipants [125,126] with a concomitant improvement

    in cognitive function and symptom improvement in

    bomb-blast-induced and study subjects (civilian and

    veterans). The existence of hypometabolic neural

    tissue provides a plausible mechanism for the recovery

    of function seen in both clinical TBI and stroke studies

    that tested HBO2 as a treatment. Although the exact

    mechanisms for reactivation are not elucidated, it appears

    that increases from the 5% to 21% oxygen concentrationcan have an effect on the mitochondria of hypometabolic

    neural tissue, restoring them to baseline function [108,

    109].

    The unintended oxygen dosing trials

    In the literature, a sham in HBO2 (room air at 1.1-1.4

    atmospheres absolute/atm abs) has been commonly

    used as a comparator against a treatment dose ( 1.5 atm

    abs, 100% oxygen). Yet, the sham remains a point of

    contention within the hyperbaric medical research

    eld. By denition HBO2is a two component therapy:increased pressure and increased oxygen concentration.

    A sham, as established for all clinical trial interven-

    tions, is a treatment or procedure that is performed as

    a control and which is similar to but omits a key thera-

    peutic element of the treatment or procedure under

    investigation. Therefore, controlling the oxygen con-

    centration (keeping oxygen at a level equivalent to 21%

    at sea level) is essential to developing a sham that

    controls for oxygen. To effectively create a sham for

    a treatment of 100% oxygen at 1.5 atm abs would

    require a breathing mixture of 14% oxygen (and the

    remainder presumably nitrogen) delivered at 1.5 atm

    abs. This would deliver an oxygen equivalent dose to

    21% oxygen breathed at sea level (1.0 atm abs) but

    would also deliver an increased dose of nitrogen which

    could have biological effects. Use of pressurized air at

    1.2 to 1.3 atm abs would not be a true sham treatment

    for oxygen, as the subjects would be breathing a 20%-

    30% increase in oxygen than that breathed at sea level.

    This increase in oxygen (and also possibly the nitrogen

    increase) would be expected to have cellular effects.

    Pressurized air shams are not inactive. In actuality the

    use of pressurized air in studies of hyperbaric oxygen

    treatments constitutes a dosing trial rather than a sham.

    The issue facing hyperbaric medical research boils

    down to dening what constitutes a medically relevant

    dose of oxygen and fully understanding the biology of

    oxygen. Changes in air pressure smaller than 0.1 atm abs

    have induced growth factor production and an increase

    in cell division [129] in epithelial cell cultures when

    compared to the control cultures. Studies of smooth

    muscle cells derived from the human aorta reported

    that 1.1 atm abs of room air (an increase of 0.1 atm

    abs) was sufcient to increase growth rates [130,131].

    Cells (as demonstrated in vitro) detect minute change inoxygen concentration (up or down) and the 20%-30%

    increase in dissolved oxygen concentration in reported

    shams as comparators in HBO2 trials represent a

    therapeutic dose.

    The cell culture data is supported by evidence from

    studies of lung function and oxygenation with chronic

    obstructive pulmonary disease, cystic brosis, pulmo-

    nary brosis and pulmonary hypertension (thromboem-

    bolic). Moving patients from cities located 800 meters

    above sea level (0.91 atm abs, ~21% oxygen) to loca-

    tions 400 meters below sea-level (1.05 atm abs, ~21%oxygen) improved oxygenation and lung function

    [132,133]. In a presentation of pilot study data looking

    at the effects of scuba diving on veterans diagnosed with

    post-traumatic stress disorder (PTSD), a large reduction

    (~80%) in PTSD symptoms (PCL-M) was observed

    [134], with pressure ranges from 1.3 to 1.5 atm abs

    with nitrox (32%-36% oxygen/68%-64% nitrogen) sus-

    tained while diving. The sham in the Wolf, et al. [4]

    report dissolved ~30% more oxygen into the plasma

    than just breathing room air at 1.0 atm abs, inducing

    a signicant change in symptoms scores (IMPACT

    symptoms score and the PCL-M), which are in agree-

    ment with Kaplin, et al. [134], and the HOPPS trial

    [135] results, both of which reported a therapeutic

    effect of breathing pressurized air.

    Illustrative of the sham issues are two published

    studies by Anderson, et al. [5] and Nighoghossian, et al.

    [136]. Both papers were designed as double-blind

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    6/19

    338

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    HBO2 (1.5 atm abs, 100% oxygen) vs. air sham cere-bral stroke studies. The air sham showed an equivalent

    effect when compared to HBO2 in the Anderson, et al.

    study (Figure 1, left graph: Air: Nstart=19, Nend=11;

    HBO2: Nstart=20, Nend=14), while Nighoghossian, et al.

    reported statistically signicant results between the

    sham and the HBO2 treatment, with the sham show-

    ing superior improvement in two out of three measures

    (Figure 1, right graph). Lower scores on the Orgogozo

    and Rankin scales and higher scores on the Trouillas

    scale are measures of better outcomes (N=17; both

    groups; Figure 1, left graphs). In both studies it wasassumed that the shams would not be biologically

    active. The authors compared HBO2 treatment to the

    supposed inactive sham, resulting in an invalid

    analysis. Re-examination of the pre- and post-neuro-

    logical scores revealed statistically signicant differ-

    ences in both groups (before vs. after treatment)

    using a two-tailed T-test (=0.05, Figure 1). Had a

    third comparator been included, such as a standard

    of care group, the analysis and conclusions would

    likely have been markedly different.

    The use of an air sham requires the assumption of

    inactivity of the sham [137], but with sham treatments

    consistently showing better results than higher treat-

    ment pressures (100% oxygen, 1.14 atm abs vs. 2.5 atm

    abs; Figure 2 [left side]) the sham treatment shows

    biological activity. Outcomes at 90 days involved four

    stroke scales with good outcomes dened as follows:

    Reanalysis of pre- and post-HBO2therapy measures of sham vs. HBO2treatment in stroke study participants. Left box:

    Anderson, et al. study. Right box: Nighoghossian, et al. Columns are final mean values. Error bars are standard deviations.

    __________________________________________________________________________________________________________________________________________________________

    Figure 1

    Pre-HBO2Post-HBO2

    1.5 atm abs 1.5 atm abs

    1.2 atm abs 1.5 atm abs 1.2 atm abs 1.5 atm abs 1.2 atm abs 1.5 atm abs

    an NIHSS score 1; a Barthel Index score of 95 or 100;

    a modied Rankin Scale score 1; and

    a Glasgow Outcome Scale score of 5

    (Nsham=11; NHBO2=16).

    Lacking in the Rusyniak, et al. study [137] was a com-

    parator for standard of care (SoC). As can be compared

    in Figure 2 (right side), published results [138,139]

    of non-related studies (using the same post-stroke

    outcome denition criteria as Rusyniak, et al.) give

    a clear indication that air shams are not inactive, and

    higher pressures of oxygen are not necessarily betterfor treatment in post-acute stroke. Some of the problems

    in selecting an appropriate sham or control in a hyper-

    baric oxygen trial are illustrated in Table 1.

    Summary

    Pressurized air is not inactive, and comparison between

    different pressure regimes suggest that a dose response

    to neuronal healing is present with different levels of

    oxygen concentration and pressure. The 2.5-atm abs

    (at 100% oxygen) treatments appear to have very little

    effect when compared to standard of care (Figure 2),

    but mild pressurization (1.14 atm abs, 100% oxygen)

    appears to have an equivalent clinical response as tPA

    therapy for stroke and a superior response than 2.5 atm

    abs It is no surprise that subsequent DoD/VA spon-

    sored reports [1,2] used a different control group: An

    oxygen sham that controlled for oxygen concentration

    (10.5% O2 at 2.0 atm abs=21% oxygen equivalent).

    neurologic

    score

    Anderson, et al. Orgogozo RankinTrouillas

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    7/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    339X.A. Figueroa, J.K. Wright

    Graph of pre- and post-HBO2therapy measures of sham (1.14 atm abs, 100% oxygen) vs. 2.5 atm abs in stroke study

    participants (left graph) Columns are the end mean values. Standard of care for stroke and of tPA intervention (right graph).

    __________________________________________________________________________________________________________________________________________________________

    Figure 2

    1.14 atm abs

    2.5 atm abs

    ___________________________________________________________________________________________________________________________________________________________

    Table 1

    type of control purported effect advantage(s) disadvantage(s) potential error as control___________________________________________________________________________________________________________________________________________________________

    pressurized air at no oxygen effect allows for pressure effect increased concentration ignores cellular effects of

    1.2-1.5 atm abs in experimental subjects of oxygen and nitrogen increased oxygen and nitrogen

    have biological effects concentration___________________________________________________________________________________________________________________________________________________________

    no treatment none allows for monitoring subjects may experience enrolling in a study may have

    observation at rare subjects with minimal Hawthorne effect a treatment effect intervals placebo or Hawthorne effect___________________________________________________________________________________________________________________________________________________________

    pressurization with mixed none for oxygen control oxygen concentration pressure effects, regular participation in study

    gas to give room air at room air (1 atm abs) increased nitrogen protocol, increased nitrogen

    (1 atm abs) equivalent equivalent effects, placebo and concentration may have an

    oxygen concentration Hawthorne effects effect___________________________________________________________________________________________________________________________________________________________

    delay in treatment/ none during delay all participants can be subjects may improve subjects may change for

    crossover study offered HBO2therapy or deteriorate during better or worse during delay

    delay, placebo and and differ from treatment

    Hawthorne effects may group

    be operational during delay___________________________________________________________________________________________________________________________________________________________

    historical controls none allows for monitoring the data may be insufficient

    untreated course of the and too varied for

    disease adequate comparison,

    no possibility of measuring

    placebo or Hawthorne

    effects___________________________________________________________________________________________________________________________________________________________

    standard of care healing through allows comparison with standard of care may not excludes intervention, placebo

    supportive therapy historical data and comparison actually be therapeutic and Hawthorne effects

    or accepted standard with new treatments

    therapy___________________________________________________________________________________________________________________________________________________________

    Comparison of different controls used in HBO2clinical trials for TBI/PCS and PTSD.

    p=0.11 p=0.02 p=0.01 p=0.04

    percentagepfparticipants

    with

    goodoutcomemeasures

    Barthel Modified Glasgow NIHHS Barthel Modified NIHHS

    Index Rankin Outcome Index Rankin

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    8/19

    340

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    Reinterpreting the DoD/VA results

    The single DoD/VA sponsored clinical trial by Cifu,

    et al. was a three-arm design [1,2] and reported on

    one group of Marines (N=61). Study participant were

    randomized into three different treatments groups

    (2.0 atm abs: 10.5% oxygen, 75% oxygen and 100%

    oxygen), with participants and researchers blinded to

    treatment. The participants were given the exact same

    pressure (2.0 atm abs) but given different oxygen mix-

    tures to simulate different treatments: 21% oxygen;

    1.5 atm abs at 100% oxygen; 2.0 atm abs at 100% oxy-

    gen. The two primary measures they looked at were

    the Rivermead Post-Concussion Questionnaire (RPQ)

    and the PTSD Checklist Military Version (PCL-M).

    In each of the articles by Cifu, et al. [1,2], the con-

    clusions were that there was no effect from HBO2,

    partially contradicting the reported results from the

    Wolf, et al. study [4]. The Wolf, et al. study reports a

    statistically signicant effect with hyperbaric air (1.3

    atm abs, 21% oxygen). The effects were large in the

    Wolf, et al. publication (41% difference between pre-

    and post-testing on the RPQ, Figure 3), but they were

    not reported as signicant in the aggregate scores,

    although nine symptoms showed signicant differenc-

    es in the sham treatment group (Figure 3). The PCL-

    M scores are also very close in overall effect between

    pre- and post-treatment: 16% improvement in the Wolf

    __________________________________________________________________________________________________________________________________________________________

    Figure 3

    Pre-HBO2Post-HBO2

    Wolf, et al.

    Meansymptoms

    core

    Cifu, et al. Harch, et al.

    sham

    2.4atm

    sham

    2.4atm

    10.5

    %

    75%

    100%

    10.5

    %

    75%

    100%

    1.4atm

    1.5atm

    RPQ PTSD RPQ PTSD RPQ PTSD

    Cross-study analysis of results from the Rivermead Post-Concussion Questionnaire (RPQ) and the PTSD Checklist-Military Version

    (PCL-M). Wolf: Nsham=24, NHBO2=24; Cifu: N10.5%= 21, N75%= 18, N100%=21; Harch: N=16. *= p 0.05; ?=unreported value of P.

    study and 14% in the Cifu study. The Cifu, et al. studies

    conclude that the results were clinically non-signicant,

    but the aggregate PCL-M scores were statistically sig-

    nicant, just like in the Wolf, et al. study (see Figure 3).

    The lack of change in the RPQ and PCL-M scores

    in the sham and 1.5 atm abs equivalent groups for

    the Cifu study could be due to insufcient number

    of study participants, the use of paper and pencil vs.

    computerized testing (time variations in response times

    are subject to analysis in computer tests), subject

    selection (all Marines were in a Warrior Transition

    Unit) or other external factors associated with study

    location. The Wolf, et al. study had six and three more

    study participants per group, respectively (N=24 for

    both the sham and 2.4 atm abs groups) than the Cifu

    study (N=18 for the 75% oxygen and N=21 for the

    100% group). Given how close the results were be-

    tween both studies, the addition of six and three more

    study participants by the Wolf, et al. study could ex-

    plain the difference seen between both articles. The Cifu

    et al. studies [1,2] also appears to have been under-

    powered, a point they acknowledge in their publication.

    Another factor at play is the relatively high pressures

    used by the DoD/VA sponsored studies [1,2,4]. Global-

    ly, a consensus is building for the use of lower pressure

    treatments, ideally 1.5 atm abs or lower, for central

    nervous system injuries. For the DoD/VA sponsored

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    9/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    341X.A. Figueroa, J.K. Wright

    studies [1,2,4], the HBO2 intervention doses were

    at or above 2.0 atm abs in pressure. The lower sham

    treatment [4] was much closer to the ideal pressure

    that proponents of HBO2normally use for neurological

    treatments [125,126,140,141].

    The improvements measured for RPQ and PCL-M

    outcomes in the Wolf and Cifu studies are in agreement

    with another published report by Harch, et al. [126].

    Harch, et al. reported on the treatment of 16 Marines

    with bomb-blast-induced mild TBIs (mTBI) and pro-

    duced results that were in agreement with the DoD/VA

    sponsored study results [1,2,4], if not their conclusions

    (Figure 3). The conclusion of Cifu, et al. that HBO2 is

    ineffective on mTBI is not supported by the data they

    acquired. All three studies measured statistically signi-

    cant improvements in the PCL-M scores [1,3,126] and

    are in agreement with previously published outcomes

    [142] in case reports of HBO2 reducing PTSD symp-

    toms. The overall trend between the published studies

    using HBO2 as an intervention on chronic mTBI/PCS

    favors a clinically positive outcome with HBO2 treat-

    ment across a variety of measures. Adding to this dis-

    cussion is a presentation of an Army study, HOPPS

    [135] (Clinicaltrials.gov, NCT01306968). The HOPPS

    study (Hyperbaric Oxygen Therapy for Persistent Post-

    Concussive Symptoms) saw a statistically and clinically

    __________________________________________________________________________________________________________________________________________________________

    Figure 4

    SoC N = 23

    1.5 atm N = 24

    sham N = 25

    SoC

    1.5atm

    sham

    SoC

    1.5atm

    sham

    SoC 1.5 atm sham

    Result of HOPPS study. (Left side) RPQ and PCL-M (PTSD) results compared to standard of care (SoC), 1.5 atm abs

    (100% oxygen) and sham (1.3 atm abs, 21% oxygen). PTSD scores that exceed 8 points (*C) are considered clinically significant.

    (Right side) The results of the health survey (SF36) of HOPPS study.

    RPQ PTSD

    signicant improvement six months post-treatment

    in the Rivermead Symptoms Questionnaire (RPQ;

    p< 0.05) and PCL-M scores (*C; >8 point improvement;

    Figure 4). Six-month post treatment measures (using

    the Short Form Health Survey [143]) revealed that im-

    provements were greater than standard of care (SoC),

    but it also saw a graded improvement between sham

    and HBO2 treatments. The sham produced results

    equivalent to or better than the 1.5 atm abs treatments.

    HOPPS has a one-year follow-up to perform and other

    measures, such as cognitive, neurological and psycho-

    motor outcomes remain to be reported, but based on

    previous clinical experience with TBI and PCS sufferers

    we expect the one-year follow-up results to retain

    their signicance.

    Relevant to our discussion regarding the use of

    HBO2for neurological injuries is the necessity to ensure

    that studies use a well-controlled population of study

    subjects. The Churchill, et al. study [144] tested a

    mixed population of acquired brain injury study subjects

    (ABI; stroke, anoxia and trauma) and saw a mix of posi-

    tive and inconclusive results. The TBI population was

    skewed toward the severe TBI category (21/27), with

    six study subjects in the mild-to-moderate TBI category

    (Figure 4, top graph). Illustrative of the difculties in

    testing a heterogeneous ABI population are the Wechsler

    Short Form Health Survey (SF36)HOPPS

    pointimprovement

    fromb

    aseline

    unfavorable

    favorable

    14

    12

    10

    8

    6

    4

    2

    0

    20

    15

    10

    5

    0

    -5

    -10

    -15

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    10/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright342

    Abbreviated Scale of Intelligence (WASI: Figure 5,

    bottom graph) scores. Although the WASI reported

    signicant improvements in aggregate, when broken

    down by groups (TBI, stroke and anoxia), only theTBI and stroke showed statistically signicant changes

    (Figure 4, bottom graphs). The anoxia group saw no

    statistically signicant improvements using the WASI.

    Further testing via other parameters produced results

    that were, at best, difcult to interpret. Although there

    is a temptation to conclude that HYBOBI (hyper-

    baric oxygen therapy in chronic stable brain injury;

    NCT00830453) is another publication pointing to the

    failure of HBO2 for treating neurological conditions,

    we would agree with the authors concluding remarks:

    Many clinical trials incorporate patient reported

    outcomes, but future studies should use a standard-

    ized brain injury symptom questionnaire that has

    been robustly validated in the population of interest

    [underline is ours]. Unfortunately, such a measure,

    validated across all brain injury etiologies and

    severities, was not available for this study.

    Dening the population that will be treated and grading

    the injury level are important steps in assessing and

    validating the effectiveness of a therapy.

    Summary

    The DoD/VA sponsored studies [1,2,4] reached conclu-

    sions that were technically correct, but failed to recog-

    nize the clinical relevance in the pre- and post-treatment

    values; one study [4] relied on the difference between

    treatment groups and a questionable sham intervention

    to reach a questionable conclusion. Published case

    reports and clinical studies that have utilized HBO2as

    an intervention in mild to moderate TBI and PCS have

    shown consistent positive results [125,126,141,145,

    146]. The conclusions of the DoD/VA sponsored tri-

    als received the widest media coverage [147], giv-

    ing the impression to the public and general medicine

    that HBO2 was ineffective for the treatment of mTBI/

    PCS. In reality, the DoD/VA study results provid-

    ed a boon to HBO2 clinical research at many levels.

    __________________________________________________________________________________________________________________________________________________________

    Figure 5

    Graphical representation of HYBOBI results. Top graph, distribution of acquired brain injuries.

    Bottom graphs, WASI intelligence scores divided by injury type. P-values are pre- and post-HBO 2analysis at six months.

    WechslerAbbreviated

    Scale

    ofIntelligence

    numberofstudy

    participants

    Pre-HBO2

    Post-HBO2

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    11/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright 343

    The missing placebo

    The placebo effect is a well-recognized component of

    medical treatment, sometimes superseding the effects

    of the actual treatment [148-152]. Controls are an es-

    sential component of clinical trials, but sometimes the

    use of specic controls (a sham or placebo) is just not

    feasible or too costly to set up (especially with HBO2).

    Placebos are not always used (cancer studies compare

    new drugs or procedures against standard of care) and

    comparing pre- and post-treatment measures (including

    crossover designs), although not deemed the most

    stringent, are acceptable and valuable in medicine

    [153]. As we have discussed above, the use of a non-

    validated sham (pressurized air) in many of the previ-

    ous HBO2 studies have clouded the debate regarding

    HBO2 effects and efcacy for neurological injuries.

    The assumption of a placebo effect with studies thatused pressurized air as a sham control is awed. Using

    a comparator that has biological activity equal to the

    treatment group is not a sham or a placebo: It is a bad

    study design.

    Although poorly controlled, some useful results can

    be extracted from existing studies. An example is

    the magnitude of the placebo effect in HBO2 studies.

    Unreported in the discussions of the Cifu, et al. publi-

    cations [1,2] was the lack of any measurable placebo

    or Hawthorne effect in the 2.0 atm abs, 10.5% oxygen

    group (the 21% oxygen equivalent; Figure 3). TheCifu, et al. study demonstrated that being in the pres-

    ence of a hyperbaric chamber and sensing an increase

    in pressure did not induce a placebo effect in their

    21% oxygen equivalent sham group (Figure 3). The

    increased attention by the study technician and doctors

    did not produce a Hawthorne effect on the study par-

    ticipants. On the contrary:

    ...we found that the sham and the 1.5 ATA

    equivalent groups demonstrated nonsignicant

    [underline is ours] ... (worsening) in their raw

    total RPQ scores... (P.18) [2]

    The 2.0 atm abs, 10.5% oxygen group did not produce

    any statistically signicant changes in the RPQ or

    PTSD scores (placebo or nocebo) from baseline

    measures, as would be expected when simulating a

    21% oxygen equivalent.

    In lieu of this analysis, a serious reconsideration of

    the published literature must occur. Previous studies that

    have a crossover design, that compare pre- and post-

    treatment measures and have avoided the sham assump-

    tion (using compressed air at 1.14-1.5 atm abs) should

    be given equal consideration when analyzed side by

    side with the sham or placebo controlled studies. The

    assumption that pressurized air has negligible or no

    biological activity is refuted by cell culture, animal

    studies and clinical reports. The requirement for a sham

    control is an important component for clinical trials,

    but the absolute requirement for a sham should be

    tempered by the results of previous trials, the realiza-

    tion that pressurized air is biologically active and the

    inclusion of other controls (such as standard of care).

    Taken as a whole, these results support the idea that we

    are witnessing a real and powerful neurological repair/

    reactivation mechanism for mTBI with oxygen, even

    under low positive-pressure room air.

    Summary

    The result from the rst DoD/VA study [4] must be

    viewed as an early entry in the arena of low-pressure

    therapy results, as the sham group is not a true sham.

    The results from the other DoD/VA sponsored stud-

    ies [1,2], highlight the lack of a measurable placebo/

    Hawthorne effect and the ability of HBO2 to improve

    PTSD symptoms in blast-induced PCS. The HOPPS

    study [135] supports the results of the Wolf, et al. study

    [4] and the other published HBO2outcome for TBI/PCS[125,126,141,145,146]. Pressurized air (1.3 stm abs,

    21% oxygen) can produce a treatment response that

    improves measured outcomes in TBI/PCS and PTSD

    symptoms and is superior to standard of care for

    TBI/PCS [135].

    The urgency of now

    A large fraction of the current epidemic of active duty

    U.S. military suicides (22+ service members a day take

    their lives, 44 attempt suicide) [154] is possibly due to

    misdiagnosing the military TBI population with PTSD

    or failing to appropriately diagnose other mental health

    conditions [155]. In the civilian population as well, the

    issue of misdiagnosis of TBI as PTSD is similar to that

    for the U.S. military [156]. The DoD and VA have spent

    at least $9.2 billion since 2010 [157] treating PTSD

    but have been unable to stem the epidemic of suicide

    and mental illness in veterans. Drug interventions are

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    12/19

    344

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    inadequate, and studies continue to nd that pharmaco-

    logical interventions are not effective in treating the

    symptoms of TBI or PTSD [158-160]. Many cases of

    veteran suicide have been linked to prescribed medi-

    cation [161,162].

    Currently, the debate regarding the utility of HBO2for

    treating mTBI, PCS or PTSD is mired in controversy. At

    the heart of this controversy is a simple question:

    Does it work?

    Based on information from case reports and phase I

    studies HBO2 does work as a therapy for mTBI/PCS

    and PTSD as they are currently diagnosed. None of

    the prescribed therapies for mTBI or PCS for service

    members (or civilians) has undergone a phase III RCT.

    Currently, the best that medicine and therapy can offer

    is the chance for a new normal, a term that is utilized

    in rehabilitation to inform those with a brain injury that

    they have to live with lower expectations and abilities.

    For individuals who are living with a chronic brain

    injury and surviving the symptoms, being told that they

    need to adapt to reduced abilities can be a demoralizing

    diagnosis. The literature in English for mTBI, PCS and

    PTSD case reports and clinical trials has the weight of

    evidence in favor of HBO2 having a reparative

    effect (Table 2).

    Realistically, not all patients will benet from HBO2.

    Yet, is it worthwhile to consider HBO2 as a therapy

    for mTBI/PCS if it can improve the lives of 10%, 25%

    or 50% of affected individuals? Would a new new

    normal that is, 25%-50% better than the old new

    normal be worth the effort? At what point will the

    general medical community draw the line for the

    potential treatment of more than 5 million Americans?

    The fundamental questions that should be asked by

    physicians are the following:

    1. Who are the most likely candidates to benet

    from HBO2?

    2. What are the minimum treatments that can elicit

    a clinically relevant improvement?

    _____________________________________________________________________________________________________________________________________

    Table 2

    authors year Dx symptoms neurocog pre-/post- between type of # of sham HBO2 HBO2 # of time since tests HBO2 tests design arms press. press. dives subj. injury

    A75% O2 100% O2 totalB10.5% O2

    C(Med Air)_____________________________________________________________________________________________________________________________________

    Eovaldi, et al. 2005 PTSD yes N/A N/A N/A pre- to post- 1 N/A 2.4 atm 7 1 4 days_____________________________________________________________________________________________________________________________________ Hardy, et al. 2007 mTBI yes yes yes N/A pre- to post- 1 N/A 2 atm 20/60 1 12 mos.

    _____________________________________________________________________________________________________________________________________ Lin, et al. 2008 SoCno N/A N/A SoCno yes RCT w/ 2 N/A 1.5 atm 22/22

    /yes /yes SoC grp. (44)_____________________________________________________________________________________________________________________________________ Wright et al. 2009 mTBI yes yes yes N/A pre- to post- 1 N/A 1.5 atm 40 2 8 mos.

    _____________________________________________________________________________________________________________________________________ Harch et al. 2009 mTBI yes yes yes N/A pre- to post- 1 N/A 1.5 atm 40 1

    _____________________________________________________________________________________________________________________________________ Harch et al. 2009 mTBI yes yes yes N/A pre- to post- 1 N/A 1.5 atm 40 12 ~ 2 yrs.

    _____________________________________________________________________________________________________________________________________ Stoller et al. 2011 mTBI yes yes yes N/A pre- to post- 1 N/A 1.5 atm 40 3 3 mos./20 yrs.

    _____________________________________________________________________________________________________________________________________ HOPPS 2012 PCS SoCno/ N/A SoCno/ SoCyes/ RCT w/sham 3 C1.5 atm 1.5 atm 40 23/24 ~ 2 yrs.

    Ayes/yes Ayes/yes Ano/no /SoC /25 (72)_____________________________________________________________________________________________________________________________________ Wolf et al. 2012 PCS *no *no *no *no RCT w/ 2 C1.3 atm 2.4 atm 30 24/24 3-71 mos.

    sham (48)

    _____________________________________________________________________________________________________________________________________ Boussi- 2013 mTBI yes/no yes/no yes/no yes//no crossover 2 N.A 1.5 atm 40 32/24 ~34.6 mos./

    Gross, et al. //yes //yes //yes //24 (56) ~31.7 mos._____________________________________________________________________________________________________________________________________ Cifu et al.+ 2013 PCS Bno/Ano/ Bno/Ano/ Bno/Ano/ Bno/Ano/ RCT w/ 3 B2 atm/ 2 atm 40 B21/A18 ~8.5 mos.

    Walker et al. no* no* no* no sham A2 atm/ /21 (60)_____________________________________________________________________________________________________________________________________

    Filled light-grey cells are for reports of positive outcomes with HBO2. Dark-grey cells are for outcome with no discernible change.

    SoC = standard of care. Starred outcomes (*) for Symptoms, Neurocog Tests, Pre-/Post-HBO2and Between Groups are for stud-

    ies that concluded no significant result, but statistical analysis contradicts conclusion with results that are statistically significant

    between pre- and post-testing within groups.

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    13/19

    345

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    3. What combination of treatments can produce

    the maximum improvement?

    Ideally, a set of RCTs with true shams and standard of

    care comparators will settle this long-standing issue.

    But we are far from the ideal. People with an mTBI or

    PCS need treatment now. Given the published super-

    iority of HBO2over the standard of care (in a military

    and civilian population), HBO2 should be covered

    or given provisional approval. If, in the future, the

    medical community can replicate the presumptive

    placebo effect induced by HBO2 chambers and fa-

    cilities, this can become the standard of care.

    The contradictory outcomes from published HBO2

    studies have muddied the waters and added to the con-

    fusion of clinically relevant results. Replication of

    results remains difcult to accomplish with the ob-

    stacles of cost, ill-dened controls and non-standardassessment scales. HBO2is not a eld known for deep

    research pockets, industrial/academic collaboration or

    a dedicated budget from the Federal government for

    research. Clinical trials in diffuse neurological injuries

    are attempts at placing parameters on complex medical

    cases with varied history of injury, genetic background,

    nutritional variations, socioeconomic factors and previ-

    ous attempts at medical intervention. There is no single

    standard for testing recovery in a general TBI popula-

    tion, the ideal measure of efcacy being the return to a

    productive life for the study subjects and the long-termcessation of brain injury sequelae. For the majority of

    HBO2 studies approximations vs. replications of the

    trials are par for the course. Variations are to be

    expected with the current low number of subjects

    recruited in U.S., Israeli, Taiwanese and Chinese

    HBO2studies on TBI recovery.

    The Wolf, et al. [4], Cifu, et al. [1,2] and Army [143]

    results have added to the growing database of effects

    that HBO2 has on neurological injuries. These are ac-

    complishments that should be roundly applauded and

    commended. We do, however, disagree with the con-

    clusions reached by the authors and their interpretation

    of their results. We urge the UHMS Committee to grant

    preliminary approval for the use of HBO2 in the treat-

    ment of chronic mild to moderate TBI/PCS. Given the

    history of using non-validated shams and the emerging

    data comparing HBO2 to standard of care, HBO2has a

    superior outcome in phase I studies. The clinical reality

    is stark: currently there are no treatments that can im-

    prove symptom and cognitive performance for chronic

    mild to moderate TBI or PCS, especially one year or

    longer from the time of injury. HBO2 is safe, with few

    side effects, has durable treatment outcomes, has avail-ability in many metropolitan centers and is managed

    by both hospital-based and independent clinics. The

    evidence for improving long-term symptoms (includ-

    ing PTSD symptoms) and cognitive performance is

    substantial in this regard. Approval of HBO2 on a

    tentative basis would allow for the collection of

    results in a large patient population, and we recom-

    mend this be done. Even if some of the improvement

    seen could be attributed to placebo or Hawthorne

    effects, it is still durable and should be considered

    treatment. No other intervention can offer these resultsor this degree of hope for the mTBI/PCS patient. HBO2

    should be part of the approved treatment armamen-

    tarium, along with other appropriate and proven thera-

    pies, for mild to moderate TBI and PCS in civilian,

    active-duty and veteran populations.

    Conict of interest

    The authors report no conict of interest with this

    submission. n

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    14/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    346 X.A. Figueroa, J.K. Wright

    1. Cifu DX, et al. Hyperbaric oxygen for blast-related

    postconcussion syndrome: Three-month outcomes. Ann

    Neurol, 2014. 75(2): p. 277-286.

    2. Cifu DX, et al. The effect of hyperbaric oxygen onpersistent postconcussion symptoms. J Head Trauma

    Rehabil, 2014. 29(1): p. 11-20.

    3. Walker WC, et al. Randomized, sham-controlled,

    feasibility trial of hyperbaric oxygen for service members

    with postconcussion syndrome: cognitive and psychomotor

    outcomes 1 week postintervention. Neurorehabil Neural

    Repair, 2013.

    4. Wolf G, et al. The effect of hyperbaric oxygen on symp-

    toms after mild traumatic brain injury. J Neurotrauma, 2012.

    29(17): p. 2606-2612.

    5. Anderson DC, et al. A pilot study of hyperbaric oxygen

    in the treatment of human stroke. Stroke, 1991. 22(9): p.1137-42.

    6. Stetler RA, et al. Preconditioning provides neuro-

    protection in models of CNS disease: Paradigms and clinical

    signicance. Prog Neurobiol, 2014.

    7. Soejima Y, et al. Hyperbaric oxygen preconditioning

    attenuates hyperglycemia-enhanced hemorrhagic trans-

    formation by inhibiting matrix metalloproteinases in focal

    cerebral ischemia in rats. Experimental Neurology, 2013.

    247: p. 737-743.

    8. Zeng Y, et al. Hyperbaric oxygen preconditioning

    protects cortical neurons against oxygen-glucose deprivation

    injury: role of peroxisome proliferator-activated receptor-

    gamma. Brain Res, 2012. 1452: p. 140-150.

    9. Li Z, et al. Mechanism of hyperbaric oxygen precondi-

    tioning in neonatal hypoxia-ischemia rat model. Brain Res,

    2008. 1196: p. 151-156.

    10. Gu GJ, et al., Mechanism of ischemic tolerance induced

    by hyperbaric oxygen preconditioning involves upregulation

    of hypoxia-inducible factor-1alpha and erythropoietin in rats.

    J Appl Physiol, 2008. 104(4): p. 1185-1191.

    11. Freiberger JJ, et al. A comparison of hyperbaric oxygen

    versus hypoxic cerebral preconditioning in neonatal rats.

    Brain Res, 2006. 1075(1): p. 213-222.

    12. Li Y, et al. Preconditioning with repeated hyperbaricoxygen induces myocardial and cerebral protection in

    patients undergoing coronary artery bypass graft surgery:

    a prospective, randomized, controlled clinical trial.

    J Cardiothorac Vasc Anesth, 2011. 25(6): p. 908-916.

    13. Jeysen ZY, et al. Research report: the effects of

    hyperbaric oxygen preconditioning on myocardial

    biomarkers of cardioprotection in patients having coronary

    artery bypass graft surgery. Undersea Hyperb Med, 2011.

    38(3): p. 175-185.

    14. Yogaratnam JZ, et al.,Hyperbaric oxygen precondition-

    ing improves myocardial function, reduces length of

    intensive care stay, and limits complications post coronaryartery bypass graft surgery. Cardiovasc Revasc Med, 2010.

    11(1): p. 8-19.

    15. Goldstein LJ, Hyperbaric oxygen for chronic wounds.

    Dermatol Ther, 2013. 26(3): p. 207-214.

    16. Game FL, et al. A systematic review of interventions to

    enhance the healing of chronic ulcers of the foot in diabetes.

    Diabetes Metab Res Rev, 2012. 28 Suppl 1: p. 119-141.

    17. Thom SR, Hyperbaric oxygen: its mechanisms and

    efcacy. Plastic and Reconstructive Surgery, 2011. 127

    Suppl 1: p. 131S-141S.

    18. Garcia-Lechuz J, Bouza E. Treatment recommendations

    and strategies for the management of bone and jointinfections. Expert Opin Pharmacother, 2009. 10(1): p. 35-55.

    19. Cimsit M, Uzun G, Yildiz S. Hyperbaric oxygen therapy

    as an anti-infective agent. Expert Rev Anti Infect Ther, 2009.

    7(8): p. 1015-1026.

    20. Brown RB. Sands M. Infectious disease indications

    for hyperbaric oxygen therapy. Compr Ther, 1995. 21(11):

    p. 663-667.

    21. Goldstein LJ, et al. Endothelial progenitor cell release

    into circulation is triggered by hyperoxia-induced increases

    in bone marrow nitric oxide. Stem Cells, 2006. 24(10):

    p. 2309-2318.

    22. Thom SR, et al. Stem cell mobilization by hyperbaricoxygen. Am J Physiol Heart Circ Physiol, 2006. 290(4): p.

    H1378-86.

    23. Velazquez OC. Angiogenesis and vasculogenesis:

    inducing the growth of new blood vessels and wound healing

    by stimulation of bone marrow-derived progenitor cell mobi-

    lization and homing. J Vasc Surg, 2007. 45 Suppl A:

    p. A39-47.

    24. Lin KC, et al. Attenuating inammation but stimulating

    both angiogenesis and neurogenesis using hyperbaric oxygen

    in rats with traumatic brain injury. J Trauma Acute Care

    Surg, 2012. 72(3): p. 650-659.

    25. Harch PG, et al. Hyperbaric oxygen therapy improvesspatial learning and memory in a rat model of chronic

    traumatic brain injury. Brain Res, 2007. 1174: p. 120-129.

    26. Lee YS, et al. Long course hyperbaric oxygen

    stimulates neurogenesis and attenuates inammation after

    ischemic stroke. Mediators Inamm, 2013. 2013: p. 512978.

    27. Liu ZJ, Velazquez OC. Hyperoxia, endothelial

    progenitor cell mobilization, and diabetic wound healing.

    Antioxid Redox Signal, 2008. 10(11): p. 1869-1882.

    _________________________________________________________________________________________________________________________________________________________

    REFERENCES

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    15/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    347X.A. Figueroa, J.K. Wright

    28. Thom SR, et al. Vasculogenic stem cell mobilization

    and wound recruitment in diabetic patients: increased cell

    number and intracellular regulatory protein content

    associated with hyperbaric oxygen therapy. Wound Repair

    Regen, 2011. 19(2): p. 149-161.

    29. Zhang K, Zhu L, Fan M. Oxygen, a key factor

    regulating cell behavior during neurogenesis and cerebraldiseases. Front Mol Neurosci, 2011. 4: p. 5.

    30. Zhang Y, et al. Hyperbaric oxygen therapy ameliorates

    local brain metabolism, brain edema and inammatory

    response in a blast-induced traumatic brain injury model

    in rabbits. Neurochem Res, 2014. 39(5): p. 950-960.

    31. Lim SW, et al. Microglial activation induced by

    traumatic brain injury is suppressed by postinjury treatment

    with hyperbaric oxygen therapy. J Surg Res, 2013.

    32. Wilson HD, et al. Hyperbaric oxygen treatment is

    comparable to acetylsalicylic acid treatment in an animal

    model of arthritis. J Pain, 2007. 8(12): p. 924-930.

    33. Vlodavsky E, Palzur E, Soustiel JF. Hyperbaric oxygentherapy reduces neuroinammation and expression of matrix

    metalloproteinase-9 in the rat model of traumatic brain

    injury. Neuropathol Appl Neurobiol, 2006. 32(1): p. 40-50.

    34. Roth TL, et al., Transcranial amelioration of inamma-

    tion and cell death after brain injury. Nature, 2014. 505

    (7482): p. 223-228.

    35. Niklas A, et al. Continuous measurements of cerebral

    tissue oxygen pressure during hyperbaric oxygenation--HBO

    effects on brain edema and necrosis after severe brain trauma

    in rabbits. J Neurol Sci, 2004. 219(1-2): p. 77-782.

    36. Rockswold SB, et al. A prospective, randomized phase

    II clinical trial to evaluate the effect of combined hyperbaricand normobaric hyperoxia on cerebral metabolism,

    intracranial pressure, oxygen toxicity, and clinical outcome

    in severe traumatic brain injury. J Neurosurg, 2013. 118(6):

    p. 1317-1328.

    37. Rockswold SB, et al. A prospective, randomized

    clinical trial to compare the effect of hyperbaric to normo-

    baric hyperoxia on cerebral metabolism, intracranial

    pressure, and oxygen toxicity in severe traumatic brain

    injury. J Neurosurg, 2010. 112(5): p. 1080-1094.

    38. Rockswold SB,Rockswold GL, Dello A. Hyperbaric

    oxygen in traumatic brain injury. Neurol Res, 2007. 29(2): p.

    162-172.39. Thom SR, et al. Nitric-oxide synthase-2 linkage to focal

    adhesion kinase in neutrophils inuences enzyme activity

    and beta2 integrin function. J Biol Chem, 2013. 288(7): p.

    4810-4818.

    40. Kendall AC, et al. Hyperbaric oxygen treatment reduces

    neutrophil-endothelial adhesion in chronic wound conditions

    through S-nitrosation. Wound Repair Regen, 2013. 21(6): p.

    860-868.

    41. Yogaratnam JZ, et al. Pharmacological preconditioning

    with hyperbaric oxygen: can this therapy attenuate myo-

    cardial ischemic reperfusion injury and induce myocardial

    protection via nitric oxide? J Surg Res, 2008. 149(1):

    p. 155-164.

    42. Khiabani KT, et al. A quantitative method for

    determining polarization of neutrophil adhesion moleculesassociated with ischemia reperfusion. Plastic and

    Reconstructive Surgery, 2004. 114(7): p. 1846-1850.

    43. Miljkovic-Lolic M, et al. Neuroprotective effects of

    hyperbaric oxygen treatment in experimental focal cerebral

    ischemia are associated with reduced brain leukocyte

    myeloperoxidase activity. Brain Res, 2003. 971(1): p. 90-94.

    44. Larson JL, Stephenson LL, Zamboni WA. Effect of

    hyperbaric oxygen on neutrophil CD18 expression. Plastic

    and Reconstructive Surgery, 2000. 105(4): p. 1375-1381.

    45. Buras JA, et al. Hyperbaric oxygen downregulates

    ICAM-1 expression induced by hypoxia and hypoglycemia:

    the role of NOS. Am J Physiol Cell Physiol, 2000. 278(2):

    p. C292-302.

    46. Atochin DN, et al. Neutrophil sequestration and the

    effect of hyperbaric oxygen in a rat model of temporary

    middle cerebral artery occlusion. Undersea Hyperb Med,

    2000. 27(4): p. 185-190.

    47. Gu N, et al. Hyperbaric oxygen therapy attenuates

    neuropathic hyperalgesia in rats and idiopathic trigeminal

    neuralgia in patients. Eur J Pain, 2012. 16(8): p. 1094-1105.

    48. Quock LP, et al. The acute antinociceptive effect of

    HBO is mediated by a NO-cyclic GMP-PKG-KATP channel

    pathway in mice. Brain Res, 2011. 1368: p. 102-107.

    49. Li F. et al. Hyperbaric oxygenation therapy alleviates

    chronic constrictive injury-induced neuropathic pain and

    reduces tumor necrosis factor-alpha production. Anesth

    Analg, 2011. 113(3): p. 626-633.

    50. Smith C. et al. The neuroinammatory response in

    humans after traumatic brain injury. Neuropathol Appl

    Neurobiol, 2013. 39(6): p. 654-666.

    51. Tompkins P. et al. Brain injury: neuro-inammation,

    cognitive decit, and magnetic resonance imaging in a

    model of blast induced traumatic brain injury. J Neurotrauma,

    2013. 30(22): p. 1888-1897.

    52. Olsson A. et al. Marked increase of beta-amyloid (1-42)

    and amyloid precursor protein in ventricular cerebrospinal

    uid after severe traumatic brain injury. J Neurol, 2004.

    251(7): p. 870-876.

    53. Dams-OConnor K. et al. Risk for late-life re-injury,

    dementia and death among individuals with traumatic brain

    injury: a population-based study. J Neurol Neurosurg

    Psychiatry, 2013. 84(2): p. 177-182.

    54. Moretti L, et al. Cognitive decline in older adults with

    a history of traumatic brain injury. Lancet Neurol, 2012.

    11(12): p. 1103-12.

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    16/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    348 X.A. Figueroa, J.K. Wright

    55. Jellinger KA, et al. Effects of closed traumatic brain

    injury and genetic factors on the development of Alzheimers

    disease. Eur J Neurol, 2001. 8(6): p. 707-710.

    56. Jellinger KA, et al. Traumatic brain injury as a risk

    factor for Alzheimer disease. Comparison of two retro-

    spective autopsy cohorts with evaluation of ApoE genotype.

    BMC Neurol, 2001. 1: p. 3.57. Desai A, Kevala K, Kim HY. Depletion of brain docosa-

    hexaenoic Acid impairs recovery from traumatic brain injury.

    PLoS One, 2014. 9(1): p. e86472.

    58. Pu H, et al. Omega-3 polyunsaturated fatty acid supple-

    mentation improves neurologic recovery and attenuates

    white matter injury after experimental traumatic brain injury.

    J Cereb Blood Flow Metab, 2013. 33(9): p. 1474-1484.

    59. Lewis M, Ghassemi P, Hibbeln J. Therapeutic use of

    omega-3 fatty acids in severe head trauma. Am J Emerg

    Med, 2013. 31(1): p. 273 e5-8.

    60. Hasadsri L, et al. Omega-3 fatty acids as a putative

    treatment for traumatic brain injury. J Neurotrauma, 2013.30(11): p. 897-906.

    61. Shin SS, Dixon CE. Oral sh oil restores striatal

    dopamine release after traumatic brain injury. Neurosci Lett,

    2011. 496(3): p. 168-171.

    62. Mills JD, et al. Omega-3 fatty acid supplementation and

    reduction of traumatic axonal injury in a rodent head injury

    model. J Neurosurg, 2011. 114(1): p. 77-84.

    63. Bailes JE, Mills JD. Docosahexaenoic acid reduces

    traumatic axonal injury in a rodent head injury model. J

    Neurotrauma, 2010. 27(9): p. 1617-1624.

    64. Browne KD, et al. Chronic ibuprofen administration

    worsens cognitive outcome following traumatic brain injuryin rats. Experimental Neurology, 2006. 201(2): p. 301-307.

    65. Tobinick E, et al. Selective TNF inhibition for chronic

    stroke and traumatic brain injury: an observational study

    involving 629 consecutive patients treated with perispinal

    etanercept. CNS Drugs, 2012. 26(12): p. 1051-70.

    66. Tobinick E. Perispinal etanercept: a new therapeutic

    paradigm in neurology. Expert Rev Neurother, 2010. 10(6):

    p. 985-1002.

    67. Gopez JJ, et al. Cyclooxygenase-2-specic inhibitor

    improves functional outcomes, provides neuroprotection,

    and reduces inammation in a rat model of traumatic brain

    injury. Neurosurgery, 2005. 56(3): p. 590-604.68. Godman CA, et al. Hyperbaric oxygen induces a cyto-

    protective and angiogenic response in human microvascular

    endothelial cells. Cell Stress Chaperones, 2010. 15(4): p.

    431-442.

    69. Kaludercic N, Deshwal S, Di Lisa F. Reactive oxygen

    species and redox compartmentalization. Front Physiol,

    2014. 5: p. 285.

    70. Schmeisser S, et al. Neuronal ROS signaling rather than

    AMPK/sirtuin-mediated energy sensing links dietary restric-

    tion to lifespan extension. Mol Metab, 2013. 2(2): p. 92-102.

    71. Wolin MS, Ahmad M, Gupte SA. Oxidant and redox

    signaling in vascular oxygen sensing mechanisms: basic

    concepts, current controversies, and potential importance

    of cytosolic NADPH. Am J Physiol Lung Cell Mol Physiol,2005. 289(2): p. L159-73.

    72. Waypa GB, et al. Mitochondrial reactive oxygen species

    trigger calcium increases during hypoxia in pulmonary arte-

    rial myocytes. Circ Res, 2002. 91(8): p. 719-26.

    73. Gilles-Gonzalez MA. Oxygen signal transduction.

    IUBMB Life, 2001. 51(3): p. 165-173.

    74. Aragones J. et al. Oxygen sensors at the crossroad of

    metabolism. Cell Metab, 2009. 9(1): p. 11-22.

    75. Chandel NS, Schumacker PT. Cellular oxygen sensing

    by mitochondria: old questions, new insight. J Appl Physiol

    (1985), 2000. 88(5): p. 1880-1889.

    76. Quaegebeur A, Carmeliet P. Oxygen sensing: a commoncrossroad in cancer and neurodegeneration. Curr Top

    Microbiol Immunol, 2010. 345: p. 71-103.

    77. Dennog C. et al. Detection of DNA damage after

    hyperbaric oxygen (HBO) therapy. Mutagenesis, 1996.

    11(6): p. 605-609.

    78. Hall ED. Lipid antioxidants in acute central nervous

    system injury. Annals of Emergency Medicine, 1993. 22(6):

    p. 1022-1027.

    79. Benedetti S, et al. Oxidative stress and antioxidant

    status in patients undergoing prolonged exposure to hyper-

    baric oxygen. Clin Biochem, 2004. 37(4): p. 312-317.

    80. Gutsaeva DR, et al. Oxygen-induced mitochondrial

    biogenesis in the rat hippocampus. Neuroscience, 2006.

    137(2): p. 493-504.

    81. Anne Stetler R, et al. The dynamics of the mitochondrial

    organelle as a potential therapeutic target. J Cereb Blood

    Flow Metab, 2013. 33(1): p. 22-32.

    82. Matchett GA, Martin RD, Zhang JH. Hyperbaric

    oxygen therapy and cerebral ischemia: neuroprotective

    mechanisms. Neurol Res, 2009. 31(2): p. 114-121.

    83. Leask A. Integrin 1: a mechanosignaling sensor

    essential for connective tissue deposition by broblasts.

    Adv Wound Care (New Rochelle), 2013. 2(4): p. 160-166.

    84. Kufer DP. The role of hyperbaric oxygen therapy in

    enhancing the rate of wound healing with a focus on axon

    regeneration. P R Health Sci J, 2011. 30(1): p. 35-42.

    85. Palzur E, et al. Neuroprotective effect of hyperbaric

    oxygen therapy in brain injury is mediated by preservation of

    mitochondrial membrane properties. Brain Res, 2008. 1221:

    p. 126-133.

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    17/19

    349

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    X.A. Figueroa, J.K. Wright

    86. Wada K, et al. Mn-SOD and Bcl-2 expression after

    repeated hyperbaric oxygenation. Acta Neurochir Suppl,

    2000. 76: p. 285-90.

    87. Soustiel JF, et al. The effect of oxygenation level on

    cerebral post-traumatic apoptotsis is modulated by the

    18-kDa translocator protein (also known as peripheral-type

    benzodiazepine receptor) in a rat model of corticalcontusion. Neuropathol Appl Neurobiol, 2008. 34(4):

    p. 412-423.

    88. Zhou Z, et al. Protection of mitochondrial function

    and improvement in cognitive recovery in rats treated with

    hyperbaric oxygen following lateral uid-percussion injury.

    J Neurosurg, 2007. 106(4): p. 687-94.

    89. Chen X, et al. Interleukin-10 mediates the neuro-

    protection of hyperbaric oxygen therapy against traumatic

    brain injury in mice. Neuroscience, 2014. 266: p. 235-243.

    90. Ito U, et al. Temporal prole of experimental ischemic

    edema after threshold amount of insult to induce infarction-

    -ultrastructure, gravimetry and Evans blue extravasation.Acta Neurochir Suppl, 2003. 86: p. 131-5.

    91. Livnat A, Barbiro-Michaely E, Mayevsky A.

    Mitochondrial function and cerebral blood ow variable

    responses to middle cerebral artery occlusion. J Neurosci

    Methods, 2010. 188(1): p. 76-82.

    92. Villa RF, et al. Energy metabolism of cerebral mito-

    chondria during aging, ischemia and post-ischemic recovery

    assessed by functional proteomics of enzymes. Neurochem

    Int, 2013. 63(8): p. 765-781.

    93. Mazzeo AT, et al. The role of mitochondrial transition

    pore, and its modulation, in traumatic brain injury and

    delayed neurodegeneration after TBI. ExperimentalNeurology, 2009. 218(2): p. 363-370.

    94. Sayeed I, et al. Direct inhibition of the mitochondrial

    permeability transition pore: a possible mechanism for better

    neuroprotective effects of allopregnanolone over progester-

    one. Brain Res, 2009. 1263: p. 165-173.

    95. Robertson CL, et al. The potential role of mitochondria

    in pediatric traumatic brain injury. Dev Neurosci, 2006.

    28(4-5): p. 432-46.

    96. Liu Z, et al. Effect of hyperbaric oxygen on cytochrome

    C, Bcl-2 and Bax expression after experimental traumatic

    brain injury in rats. Chin J Traumatol, 2006. 9(3): p. 168-174.

    97. Busija DW, Katakam PV. Mitochondrial mechanismsin cerebral vascular control: shared signaling pathways with

    preconditioning. J Vasc Res, 2014. 51(3): p. 175-189.

    98. Lou M, et al. Involvement of the mitochondrial ATP-

    sensitive potassium channel in the neuroprotective effect of

    hyperbaric oxygenation after cerebral ischemia. Brain Res

    Bull, 2006. 69(2): p. 109-116.

    99. Daugherty WP, et al. Effects of hyperbaric oxygen

    therapy on cerebral oxygenation and mitochondrial function

    following moderate lateral uid-percussion injury in rats.

    J Neurosurg, 2004. 101(3): p. 499-504.

    100. Neubauer RA, Gottlieb SF, Kagan RL. Enhancing

    idling neurons. Lancet, 1990. 335(8688): p. 542.

    101. Neubauer, R.A., Idling neurons. Lancet, 1990.335(8699): p. 1217.

    102. Neubauer RA, James P. Cerebral oxygenation and the

    recoverable brain. Neurol Res, 1998. 20 Suppl 1: p. S33-S36.

    103. Holbach KH, et al. Differentiation between reversible

    and irreversible post-stroke changes in brain tissue: its

    relevance for cerebrovascular surgery. Surg Neurol, 1977.

    7(6): p. 325-331.

    104. Rossi GF, et al. Neurological improvement in chronic

    ischemic stroke following surgical brain revascularization.

    Ital J Neurol Sci, 1987. 8(5): p. 465-75.

    105. Neubauer RA, Gottlieb SF, Miale A Jr. Identication of

    hypometabolic areas in the brain using brain imaging andhyperbaric oxygen. Clin Nucl Med, 1992. 17(6): p. 477-481.

    106. Shin SS, et al. Detection of white matter injury in

    concussion using high-denition ber tractography.

    Prog Neurol Surg, 2014. 28: p. 86-93.

    107. Efrati S, et al. Hyperbaric oxygen induces late neuro-

    plasticity in post stroke patients - randomized, prospective

    trial. PLoS One, 2013. 8(1): p. e53716.

    108. Blackstone E, Morrison M, Roth MB. H2S induces

    a suspended animation-like state in mice. Science, 2005.

    308(5721): p. 518.

    109. Blackstone E, Roth MB. Suspended animation-like

    state protects mice from lethal hypoxia. Shock, 2007. 27(4):p. 370-372.

    110. Eto K, et al. Hydrogen sulde is produced in response

    to neuronal excitation. J Neurosci, 2002. 22(9): p. 3386-91.

    111. Chen CQ, Xin H, Zhu YZ. Hydrogen sulde: third gas-

    eous transmitter, but with great pharmacological potential.

    Acta Pharmacol Sin, 2007. 28(11): p. 1709-1716.

    112. Qu K, et al. Hydrogen sulde: neurochemistry and

    neurobiology. Neurochem Int, 2008. 52(1-2): p. 155-165.

    113. Baumgart K, Radermacher P, Wagner F. Applying gases

    for microcirculatory and cellular oxygenation in sepsis:

    effects of nitric oxide, carbon monoxide, and hydrogen

    sulde. Curr Opin Anaesthesiol, 2009. 22(2): p. 168-176.

    114. Wagner F, et al. Bench-to-bedside review: Hydrogen

    sulde--the third gaseous transmitter: applications for

    critical care. Crit Care, 2009. 13(3): p. 213.

    115. Olson KR, Whiteld NL. Hydrogen sulde and oxygen

    sensing in the cardiovascular system. Antioxid Redox Signal,

    2010. 12(10): p. 1219-1234.

    116. Pun PB, et al. Gases in the mitochondria. Mitochon-

    drion, 2010. 10(2): p. 83-93.

  • 7/26/2019 5 Clinical Results in Brain Injury Trials - Another Perspective

    18/19

    UHM 2015, VOL. 42, NO. 4 CLINICALRESULTSINBRAININJURYTRIALS:ANOTHER PERSPECTIVE

    350 X.A. Figueroa, J.K. Wright

    117. Tay AS, et al . Hydrogen sulde protects neurons against

    hypoxic injury via stimulation of ATP-sensitive potassium

    channel/protein kinase C/extracellular signal-regulated

    kinase/heat shock protein 90 pathway. Neuroscience, 2010.

    167(2): p. 277-86.

    118. Yong QC, et al. Effect of hydrogen sulde on intra-

    cellular calcium homeostasis in neuronal cells. NeurochemInt, 2010. 56(3): p. 508-515.

    119. Mikami Y, et al. Hydrogen sulde protects the retina

    from light-induced degeneration by the modulation of Ca2+

    inux. J Biol Chem, 2011. 286(45): p. 39379-39386.

    120. Kimura H, Shibuya N. Kimura T. Hydrogen sulde is

    a signaling molecule and a cytoprotectant. Antioxid Redox

    Signal, 2012.

    121. Morbelli S. Arnaldi D. Brain perfusion SPECT provides

    new insight on neurobiological effects of hyperbaric hyper-

    oxia. Acta Physiol (Oxf), 2013.

    122. Barrett KF, et al. Regional CBF in chronic stable TBI

    treated with hyperbaric oxygen. Undersea Hyperb Med,2004. 31(4): p. 395-406.

    123. Kraus MF, et al. White matter integrity and cognition

    in chronic traumatic brain injury: a diffusion tensor imaging

    study. Brain, 2007. 130(Pt 10): p. 2508-2519.

    124. Kraitsy K, et al. Repetitive long-term hyperbaric

    oxygen treatment (HBOT) administered after experimental

    traumatic brain injury in rats induces signicant remyelina-

    tion and a recovery of sensorimotor function. PLoS One,

    2014. 9(5): p. e97750.

    125. Boussi-Gross R, et al. Hyperbaric oxygen therapy

    can improve post concussion syndrome years after mild

    traumatic brain injury - randomized prospective trial.PLoS One, 2013. 8(11): p. e79995.

    126. Harch PG, et al. A phase I study of low-pressure hyper-

    baric oxygen therapy for blast-induced post-concussion

    syndrome and post-traumatic stress disorder. J Neurotrauma,

    2012. 29(1): p. 168-85.

    127. Gesell LB. Hyperbaric oxygen therapy indications., in

    The Hyperbaric Oxygen Therapy Committee report. 2008,

    Undersea and Hyperbaric Medical Society: Durham, N.C.

    128. Acker T, Acker H. Cellular oxygen sensing need in CNS

    function: physiological and pathological implications. J Exp

    Biol, 2004. 207(Pt 18): p. 3171-3188.

    129. Acevedo AD, et al. Morphological and proliferativeresponses of endothelial cells to hydrostatic pressure: role

    of broblast growth factor. J Cell Physiol, 1993. 157(3):

    p. 603-614.

    130. Tokunaga O, Fan JL, Watanabe T. Atherosclerosis and

    endothelium. Part II. Properties of aortic endothelial and

    smooth muscle cells cultured at various ambient pressures.

    Acta Pathol Jpn, 1989. 39(6): p. 356-362.

    131. Tokunaga O, Watanabe T. Properties of endothelial cell

    and smooth muscle cell cultured in ambient pressure. In

    Vitro Cell Dev Biol, 1987. 23(8): p. 528-534.

    132. Kramer MR, et al. Effect of natural oxygen enrichment

    at low altitude on oxygen-dependent patients with end-stage

    lung disease. Ann Intern Med, 1994. 121(9): p. 658-662.

    133. Kramer MR, et al. Rehabilitation of hypoxemic patientswith COPD at low altitude at the Dead Sea, the lowest place

    on earth. Chest, 1998. 113(3): p. 571-575.

    134. Medicine JH. Scuba diving improves function of body,

    mind in vets with spinal cord injury. [Current News Release]

    2011 September 17, 2011 [cited 2014 August 22]; Johns

    Hopkins researchers nd dramatic results in small prelimi-

    nary study]. Available from: http://www.hopkinsmedicine.

    org/news/media/releases/scuba_diving_improves_function_

    of_body_mind_in_vets_with_spinal_cord_injury.

    135. Miller S. Primary outcomes from randomized, multi-

    center trial of HBOT 1.5 ATA for post concussion syndrome.

    2013, US Army Medical Materiel Development Activity:Ft. Detrick. p. 15.

    136. Nighoghossian N, et al. Hyperbaric oxygen in the treat-

    ment of acute ischemic stroke. A double-blind pilot study.

    Stroke, 1995. 26(8): p. 1369-72.

    137. Rusyniak DE, et al. Hyperbaric oxygen therapy in acute

    ischemic stroke: results of the hyperbaric oxygen in acute

    ischemic stroke trial pilot study. Stroke, 2003. 34(2):

    p. 571-574.

    138. Wilkinson PR, et al. Longer term quality of life and

    outcome in stroke patients: is the Barthel index alone an

    adequate measure of outcome? Qual Health Care, 1997. 6(3):

    p. 125-130.139. Wasserman S, Bryer A. Early outcomes of thrombolysis

    for acute ischaemic stroke in a South African tertiary care

    centre. S Afr Med J, 2012. 102(6): p. 541-544.

    140. Stoller KP. Hyperbaric oxygen therapy (1.5 ATA) in

    treating sports related TBI/CTE: two case reports. Med Gas

    Res, 2011. 1(1): p. 17.

    141. Wright JK, et al. Case report: treatment of mild trau-

    matic brain injury with hyperbaric oxygen. Undersea Hyperb

    Med, 2009. 36(6): p. 391-399.

    142. Eovaldi B, Zanetti C. Hyperbaric oxygen ameliorates

    worsening signs and symptoms of post-trauma