Introduction
Stroke is the leading cause of neurological disability in the U.S. and worldwide. Remarkable advances have been made over the last 20 years in acute vascular treatments to reduce infarct size and improve neurological outcome. Substantially less progress has been made in treatments to enhance neurological recovery after stroke. However, a number of promising research directions have been identified for treatments targeting stroke recovery and are the focus of the current review (Figure 1).
Figure 1.
Summary of current experimental treatments for stroke recovery and their readiness for incorporation into clinical practice based on synthesis of current evidence. Readiness for clinical application ranges from preclinical development (i.e. in animal studies only, red) to broad clinical application (green).
Epochs of time post-stroke can be divided into acute (hours-days), subacute (days-weeks), and chronic (months-years) based on the tissue response and biological programs that emerge during these time windows1. In contrast to acute stroke treatments such as tPA and thrombectomy, for which the target is a clot and the goal is to salvage threatened brain tissue to limit injury, recovery treatments target surviving neural tissue with the goal of promoting neural repair. Recovery trials are thus fundamentally different from acute stroke trials2. First, the time windows available for administering recovery-based treatments span much longer periods. These time windows enable detailed within-subject analyses before, during, and after treatment. Second, stroke deficits are multi-modal (e.g., combinations of motor, language, and other deficits) and recovery is modality-specific, meaning that different deficits recover with different rates and extents3. Accordingly, recovery trials often employ modality-specific endpoints (such as the Fugl-Meyer Motor (FM) Scale), in addition to global outcome measures (such as the modified Rankin Scale (mRS) or the NIH Stroke Scale (NIHSS)) that lump together all aspects of neurological function and are used in acute stroke trials. Third, recovery trials have issues often not shared with other stroke research domains, such as variable standards of care, patient access and retention, difficulty with experimental blinding (particularly in the study of behavioral interventions), and importance of concomitant experiences4 [Table 1].
Table 1.
Main points
• Stroke recovery trials target a different biology with a different time course as compared to acute stroke trials |
∘ Stroke recovery trials target brain tissue not vasculature |
∘ Acute phase treatments (reperfusion, neuroprotection) have a time window measured in minutes and hours, while recovery treatments have time windows measured in days, week, months, or years. |
∘ Recovery treatments are not targeted to rescue dying brain tissue, but rather to promote neural repair in remaining tissue. |
• Many new promising strategies are on the horizon for stroke recovery. |
• Trials of treatments targeting stroke recovery benefit from employing modality-specific endpoints in order to obtain the granularity needed to measure differences in recovery across different neural systems (e.g., recovery of language versus gait). |
• The time window for many recovery trials affords the opportunity to measure behavior at baseline and thus change in behavior, which in turn enables within-subject assessment of recovery. |
• The choice of the study population for recovery studies can strongly influence how well preclinical results are accurately translated and how well study hypotheses are truly tested. |
Spontaneous behavioral recovery occurs after stroke, but is variable. The molecular and cellular mechanisms of this recovery have been extensively reviewed. Several treatment approaches in clinical translation aim to improve stroke recovery. Here we review contemporary approaches to therapeutically enhancing stroke recovery, focusing on recent trials. For this review, we define stroke recovery treatments as non-vascular treatments initiated in the subacute to chronic phases (days to years) after stroke. Our intent is not to be comprehensive, but to highlight select examples of promising approaches and directions with an acknowledged emphasis on motor recovery. It should be noted that to date no small molecule or biologic has been approved by the FDA to promote stroke recovery.
Conventional Therapies
Conventional therapies for patients recovering from stroke include physical, occupational, and speech therapy. These are delivered across numerous care settings. The duration, intensity, and type of conventional therapy are highly variable in the US, complicating the design of control groups in stroke rehabilitation trials. Indeed, one of the research priorities for stroke rehabilitation and recovery is better reporting and standardization of usual care in trials1.
Two specific interventions for stroke recovery with promising initial evidence emerging from conventional therapies include mirror therapy and constraint-induced movement therapy (CIMT)5. Mirror therapy, or prism adaptation therapy, uses simple equipment to focus a patient’s attention on a neglected hemifield. Mirror therapy, generally provided as an adjunct to conventional therapy, can improve motor function and reduce pain6 and might improve activities of daily living7. Mirror therapy studies have been limited by small sample sizes and methodological limitations. Larger, more rigorous trials are needed. CIMT involves intensive rehabilitation therapy to overcome learned disuse by an affected arm, with concomitant constraint of the unaffected arm. EXCITE8 was a phase 3 trial that found evidence that CIMT improved motor function, surpassing the minimally clinically important difference (MCID) in the Wolf Motor Function Test among patients with intact motor control in early chronic stroke. The timing of CIMT is important, as very early application (10 days post-stroke) was not more effective than traditional therapy9. Overall, there is moderate evidence that CIMT may be effective10 for post-stroke recovery, but specific protocols and timing have yet to be defined for widespread clinical adoption.
Small molecules
The two classes of drugs that have been most investigated as stroke recovery treatments to date are serotonergic and dopaminergic. Building on prior smaller studies, the FLAME study was a double-blind, placebo-controlled trial in which 118 patients with weakness after ischemic stroke were randomized to 3-months of oral fluoxetine or placebo, 5–10 days post-stroke11. Patients with clinical depression were excluded. Those randomized to fluoxetine showed significantly greater motor recovery to 90 days post-stroke on the FM Motor Scale than those receiving placebo (a 9.8 point boost in recovery on the 100 point total FM for upper+lower extremities, largely driven by the upper extremity where fluoxetine-related gains exceeded the MCID of 5.25 points in chronic stroke12) as well as a significantly less disability as measured by the mRS; interpretation is complicated by mild baseline imbalances favoring the fluoxetine group. A meta-analysis of 52 trials of SSRIs after stroke found that at the end of treatment, patients receiving an SSRI were less likely to be dependent, disabled, neurologically impaired, depressed, or anxious, and that favorable effects were greater in participants who were depressed at randomization13. Phase III trials examining SSRIs for stroke recovery are ongoing. Several mechanisms might account for the potential efficacy of SSRIs for stroke recovery, including reducing neural inflammation, enhancing neurotropic effects, and modulating excitatory/inhibitory synaptic and cortical network balance14, 15.
Given their well-established role in reward, learning, and neural plasticity, dopaminergic drugs have also been investigated for post-stroke recovery. In 2001, a single-center (inpatient rehabilitation facility), randomized, placebo-controlled study of 53 patients with ischemic stroke that occurred 3-weeks to 6-months previously showed that a dopaminergic drug (Sinemet 100 mg/day) improved motor recovery measured using the Rivermead Motor Assessment16. More recently, the Dopamine Augmented Rehabilitation in Stroke trial was a multicenter, randomized, double-blinded, placebo-controlled trial with some pragmatic design features that examined 6-weeks of Sinemet 100 mg/day given prior to rehabilitation therapy in 593 patients recruited 5–42 days post-stroke and found no difference between groups in the primary endpoint, walking independently17. Overall, stroke recovery studies with dopaminergic drugs have been mixed, likely reflecting complex interactions between learning, reward, environment, psychology, cognition, and genetics18. Stroke recovery is not a one-size-fits-all science, and so these results suggest the need to identify the optimal target population and conditions for therapies targeting dopaminergic neurotransmission.
Other small-molecules, including noradrenergic19 and cholinergic20 drugs, have recently been explored for post-stroke recovery. Amphetamines have been extensively studied, with mixed results, and a meta-analysis suggested that too few patients have been studied to draw conclusions21. Building off favorable preclinical studies22, a phase IIa trial in France and Spain (Clinicaltrials.gov NCT02928393) of Basmisanil, a GABA negative allosteric modulatory drug, was initiated but was terminated prematurely after 9 months (enrollment was slow--only 5 patients had been enrolled towards the goal of 95). A key insight from preclinical studies of neurotransmitter strategies is that a drug that improves recovery when initiated days after stroke onset can cause poorer outcomes if initiated too early,22, 23, reaffirming a longstanding observation in stroke recovery pharmacology that recovery drugs often have biphasic effects depending on time of initiation post-stroke.
Few traditional and alternative medicines have been studied using formal research methods. One exception is the Chinese Medicine Neuroaid Efficacy on Stroke recovery (CHIMES) study24, a multicenter, double-blind, placebo-controlled clinical trial that randomized 1,100 patients to 3 months of Neuroaid versus placebo, provided in the form of three daily doses of 4 oral capsules each. Neuroaid is a traditional Chinese medicine containing extracts of 9 herbal and 5 animal components. Patients started therapy within 72 hours of stroke onset. No difference between treatment arms was seen in the primary outcome measure, shift in the modified Rankin Scale at 3 months.
In summary, some small molecules are ready for late phase trials and indeed are underway, while others are in earlier stages of development or require study with respect to specific patient subgroups or conditions. Timing of drug initiation is critical and can be informed by preclinical studies as well as time window-finding studies in humans. Personalized medicine approaches can likely maximize benefit from small molecules for promoting stroke recovery, for example, enrolling subjects with attention to key genetic factors or pairing drug exposure with optimized reward or training. Combining small molecules with appropriate repetitive neurorehabilitation training may also be promising.
Growth factors
After stroke, many growth factors show increased levels for several weeks, playing a critical role in spontaneous neural repair through mechanisms that include neural sprouting and new synapse formation, angiogenesis, reduced apoptosis, stem cell proliferation, and immunomodulation25. One strategy to promote stroke recovery, therefore, is to increase levels of key growth factors26. Numerous preclinical stroke studies suggest that administration of exogenous growth factors ≥24 hours post-stroke significantly improves outcome, e.g., using fibroblast growth factor27, brain-derived neurotrophic factor28, epidermal growth factor plus erythropoietin29, or b-hCG followed by erythropoietin30.
Systemic erythropoietin enters the brain, and when introduced with a delay after stroke (e.g. ~24 hours), can improve outcomes31. Favorable effects of this growth factor have been described in other preclinical studies that used sequential administration of epidermal growth factor29 or b-hCG30 followed by erythropoietin, beginning 1–7 days after stroke. This approach was studied in the B-hCG+ErythropoieTin in Acute Stroke (BETAS) study, a single-dose, open-label, non-controlled safety trial of B-hCG (1–2 days post-stroke) followed by IV erythropoietin (7–8 days post-stroke), which successfully demonstrated safety32. The REGENESIS Study33 was intended to be a randomized, placebo-controlled, double-blind proof of concept study of sequential b-hCG and erythropoietin using the BETAS study treatment schedule. This trial was put on hold due to concerns arising from a previous acute neuroprotection stroke trial34 that initiated erythropoietin less than 6 hours after stroke onset and found increased drug-related mortality rates, despite the fact that the REGENESIS trial targeted a different time window and biology (erythropoietin initiated less than 6 hours after stroke in the acute trial versus REGENESIS trial which waited until 7–8 days post-stroke). As a result, the REGENESIS trial was modified to be a dose-ranging safety study, and treatment groups did not differ in NIHSS score change at Day 90. Growth factors are likely appropriate for further clinical study— development would benefit from clearer direction from preclinical studies for insights into mechanism of action (informing target human population), dosing, and optimal timing.
Monoclonal antibodies
In the CNS, three inhibitors (myelin-associated glycoprotein [MAG], oligo-myelin glycoprotein, and Nogo-A) increase levels post-stroke and result in lack of a permissive growth environment35. Monoclonal antibodies can neutralize these inhibitors by modulating activity in targeted signaling pathways, thus promoting axonal growth and potentially recovery. One study36 randomized 42 patients with stroke to two rounds (24–72 hours post-stroke and 9 days later) of placebo versus one of three dose levels of IV GSK249320, a humanized IgG1 monoclonal antibody to MAG with disabled Fc region. No safety concerns were identified, and results suggested that the antibody improved gait recovery. A subsequent phase II study37 compared the highest IV GSK249320 dose level to placebo in 134 patients with stroke 24–72 hours prior. The antibody reduced free serum MAG levels, but the primary endpoint, change in gait velocity from baseline to day 90, did not differ between groups. Effective antibody activity was confirmed in the periphery but was not assessed in the CNS. In summary, monoclonal antibodies would benefit from further preclinical development followed by further early phase clinical study.
Cellular Therapy
Cell treatments have multiple effects in the post-stroke brain. Transplanted cells might integrate into the existing brain architecture replacing cells lost to stroke; secrete cytokines, growth factors, and extracellular matrix proteins; or modify systemic immune responses to stroke aiding functional recovery. In a randomized, double-blind, placebo-controlled Phase 2 study, Hess and colleagues delivered allogeneic marrow-derived cells intravenously to 126 patients early (24–48 hours) after ischemic stroke38. Cell administration was safe, but no significant efficacy signal was seen in the primary endpoint: excellent outcome at day 90 (mRS score 0–1, NIHSS score 0–1, and Barthel score ≥95). Post hoc analysis at one-year found a difference in excellent outcome (23% cell-treated versus 8% placebo, p=0.02).
In a Phase 1/2 study, Kalladka and collaborators39 stereotaxically implanted allogeneic immortalized human fetal neurons into the ipsilesional putamen adjacent to focal infarcts in 11 men with stable neurological deficits 6–60 months after ischemic stroke. There were three serious adverse events during the first month post-procedure, two asymptomatic intracranial hematomas attributed to the procedure and one symptomatic infarct attributed to pre-procedure suspension of antiplatelet drugs. Non-serious adverse events were mostly mild. Several patients showed treatment-related improvement, including on NIHSS, Ashworth scale, and Barthel Index.
In a single-arm, open-label study, Steinberg et al40 stereotaxically implanted allogeneic modified marrow-derived mesenchymal cells into tissue surrounding focal infarcts in 18 patients with stable deficits 6–60 months after ischemic stroke. Cell implantation appeared safe, with reversible adverse events largely related to the surgical procedure. Over one-year follow-up, significant improvements were seen in the European Stroke Scale, NIHSS, and FM scores, with no significant changes in mRS.
In summary, early studies suggest that systemic and intracranial cell administration is feasible, and may prove safe and effective as treatments for promoting neurological recovery early and late after stroke. Optimal dose, route, and timing of cell administration remain underexplored. Further randomized, double-blind, placebo-controlled trials are needed and are ongoing.
Brain Stimulation
Several forms of brain stimulation have been investigated to enhance recovery after stroke, most in small studies. Some aim to increase activity, e.g., with high-frequency repetitive transcranial magnetic stimulation (rTMS)41, while others aim to reduce activity in areas thought to have an undesired suppressive effect, e.g., with low-frequency repetitive rTMS42. Similar approaches have been advanced for transcranial DC stimulation (tDCS)43, where electrodes broadly apply current through the scalp in the treatment of several different types of post-stroke deficit including motor, language, memory, and neglect. An advantage of tDCS is it is readily applied during rehabilitation. Meta-analyses have concluded that more evidence is needed to establish benefit44, 45. Two recent studies showing positive effects of tDCS after stroke raise enthusiasm, one targeting dysphagia during the acute stroke hospitalization46 and the other targeting chronic aphasia47. These highlight issues of capitalizing on anatomical substrates of neurologic function (e.g., bihemispheric control, for swallowing; unilateral dominant hemisphere control, for language) as well as importance of considering timing of intervention post-stroke.
Other approaches for brain stimulation are under study, including deep brain stimulation. A phase III trial did not find a significant benefit from epidural cortical stimulation combined with rehabilitation compared to rehabilitation alone in patients with chronic stroke48. Vagus nerve stimulation has been hypothesized to enhance brain plasticity, possibly by modulating activity in neurotransmitter systems such as acetylcholine and norepinephrine49. A recent randomized human clinical trial of vagal nerve stimulation paired with rehabilitation demonstrated safety and feasibility50, and efficacy trials are ongoing.
To advance brain and nervous system stimulation for the treatment of stroke recovery, further clinical and possibly preclinical studies (preclinical studies have been limited to date) are needed to inform mechanism, dose, intensity, and optimal targets in order to maximize effects.
Robotics
Robots are promising for stroke rehabilitation because they can deliver consistent, programmable, and high-intensity motor therapy, and furthermore can use sensors to measure movement kinematics. Numerous robotic systems have been studied51. A 2010 multicenter study of 127 patients conducted within the VHA system showed that robot-assisted therapy did not significantly improve motor status at 12 weeks, the primary endpoint, as compared with usual care or intensive therapy. A delayed favorable effect as compared with usual care, however, was suggested in secondary analyses over 36 weeks52. This study may have been limited by its choice of study population, enrolling patients with very severe deficits in the chronic phase post-stroke, among whom producing substantial benefits is challenging. Numerous other studies spanning different recovery stages over the past 20 years have demonstrated economic feasibility53 and therapeutic benefits of robot therapy. Benefits of robot-assisted therapy for the upper extremity, however, are attributable to the intensity and repetition of movement practice54, and robot-mediated therapy has not demonstrated clear additional benefits when compared to physical therapy when intensity and number of movement repetitions have been matched55. Moreover, a recent Cochrane review of robot-assisted and electromechanical arm training devices for improving arm function after stroke found only low quality evidence that such interventions improve activities of daily living56.
For the lower extremity, meta-analyses have shown that adding electromechanical-assistance (robot-assistance) to body-weight support increases the odds of patients becoming independent with walking57. However, a large clinical trial did not find body-weight supported treadmill training to be superior to progressive exercise at home managed by a physical therapist in patients two-months post-stroke58. AHA/ASA Guidelines describe Class IIb evidence supporting mechanically assisted walking with body weight support for patients who are non-ambulatory after stroke59.
Overall, Phase 2 and possibly Phase 3 studies of robotics for stroke recovery are needed that enroll populations most likely to benefit, or that test engineering approaches targeting improved functional outcomes using methods that build on the unique strengths of robots. Further development of robotic interventions might also include adjusting robot movements to patterns of disrupted movements and linking robot assistance to cortical activity (i.e. brain-computer interfaces).
Brain-Computer Interfaces
When a lesion is severe, one potential approach to restoring voluntary behavior is to capture signals from intact cortex and bypass the lesion. Some advocate a surgical approach. A recent study in patients with spastic arm paresis, due to various diagnoses (25% were due to stroke), an average of 15 years after the event, showed that C7 nerve transfer from the nonparalyzed to paralyzed body side was associated with a 15 point increase in the FM arm score in the affected arm, exceeding the MCID for the upper extremity of 5.25 points in chronic stroke12, and with sustained improvements as compared to controls (rehabilitation alone) at one year60.
A non-invasive approach to lesion bypass is a brain-computer interface (BCI). A BCI is a system that translates CNS signals into command signals for a technological device. The components of a BCI include devices for recording neural signals (sensors), the process of translating neural signals into command signals (neural decoding), devices for executing command signals (effectors), and the feedback provided to the user (feedback). Using BCI systems, people with neurologic disease have successfully controlled computer cursors to regain communication and robotic prosthetics to restore movement61. A complementary goal of BCI is to enhance rehabilitation therapy effects62. The hypothesis is that training patients to produce normal brain activity patterns through feedback reinforcement or linking neural activity directly to sensory feedback of intended actions engages activity-dependent Hebbian neuroplasticity mechanisms and thereby restores native neurologic functions. Initial studies in patients with chronic stroke have been promising, e.g., EEG-BCI training with an upper extremity orthosis produced a 3.4-point significant positive difference in the FM arm motor score compared to controls. Though shy of the MCID, behavioral gains in the experimental group correlated with adaptive changes in fMRI neural activity pattern, arguing that BCIs may induce neuroplastic changes63. Another recent study64 showed that use of an EEG-driven exoskeleton to open and close the affected hand using signals from the contralesional (unaffected) hemisphere significantly improved arm function (6.2 point increase on the Action Research Arm Test score, exceeding the 5.7 point MCID65 in chronic stroke). This study was notable given the use of signals from the unaffected hemisphere, and delivery of therapy at home. Advances in BCI will benefit from a deeper understanding of how to facilitate personalized neural signals to enhance neuroplasticity, as well as a more nuanced understanding of circuit function in spontaneous recovery after stroke. More generally, the paradigm of closed-loop neurotechnological devices, in which sensing neural activity is coupled with effecting change in the nervous system, is a promising neurorehabilitation approach. Further preclinical study and early phase clinical development are needed.
Telerehabilitation
Task-based practice improves outcomes after stroke in a dose-dependent manner, and also provides the behavioral component of the experience-dependent plasticity that is facilitated by many restorative therapies. However, patients often do not receive large doses of supervised practice due to issues such as cost, difficulty traveling to appointments, and regional provider shortages. Telehealth might provide new means to achieve high doses of therapy in an accessible manner using communication technologies. One recent study evaluated a home-based telerehabilitation system in patients with chronic hemiparetic stroke with onset 3–24 months prior66. Enrollees received 28-days of telerehabilitation prescribed by licensed therapists using a system delivered to the patient’s home, each day consisting of one structured hour focused on individualized exercises and games, stroke education, plus an hour of free play. Compliance was excellent: participants engaged in therapy on 97.9% of assigned days, a substantial improvement over traditional home exercise programs. Arm repetitions averaged 879/day, far exceeding the 32 repetitions found with standard of care therapy67 and approximating the several hundred movements/day found in primate studies as important to achieving optimal post-stroke motor cortex plasticity68. Arm motor status showed significant gains, with half of the participants exceeding the arm motor FM score MCID. This approach did not require computer skills, levels of which were unrelated to motor gains or system use. A multisite phase II trial is now underway (Clinicaltrials.gov NCT02360488). Meta-analyses of telerehabilitation after stroke have found either better or similar effects when compared with conventional in-person therapy, but have noted high heterogeneity in treatment content across studies69, 70. Further clinical development of telerehabilitation to optimize dose and standardize protocols is warranted before late-phase clinical trials.
Virtual Reality and Movement Sensors
There is converging evidence that high-dose, repetitive, task-oriented, and task-specific training is beneficial, at least for selected patients after stroke71, 72. The key is not simply a greater number of repetitions, but also task salience and specific attention to time post-stroke and external environment73–76. Virtual reality (VR), ranging from non-immersive to fully-immersive depending on the degree to which the user is isolated from his or her physical surroundings when interacting virtually, has been used as an alternative and as an adjunct to conventional rehabilitation. VR provides a scalable means to deliver high-intensity, task-specific training as well as real-time feedback (i.e. rewards for positive trials) potentially with enhanced salience. However, the evidence for efficacy of VR to date has been mixed77–80. Well-designed studies that build on prior work are needed, possibly maximizing salience to individual patient factors.
Movement sensors are another means of providing feedback with the goal of motivating greater skills practice and thus improving rehabilitation. In this regard, lower extremity sensors providing simple measures of physical activity and gait are more developed than upper extremity sensors, for which accurately characterizing reaching and grasping movements represents an ongoing computational challenge81. In one lower extremity sensor feasibility study, participants were given feedback of walking metrics via ankle sensors. This augmented feedback did not increase time spent practicing or improve walking outcomes in a rehabilitation setting as compared to therapists providing feedback on walking speed alone (without the use of ankle sensors)82. A recent review found some evidence that lower extremity wearable sensors improved outcomes, but the design of studies included was highly variable83. As with VR, additional earlier phase studies are needed, particularly those building on principles of personalized intervention.
Summary and Future Directions
This review highlighted contemporary treatment approaches aimed at improving recovery after stroke. Development of stroke recovery treatments will depend on increased understanding of the complex events underlying recovery and development of methods to measure treatment effects in human patients. A key issue is optimal treatment timing, as specific critical periods for most interventions have not yet been defined. Trial design is also paramount, for example, endpoints targeting a single domain (motor, language, etc.) may be more informative than global outcome measures that combine data from multiple different domains. Outcome measures benefit from having sufficient granularity to detect useful functional gains and being informative about the biology of recovery— global outcome measures such as the mRS may have critical shortcomings in this regard. As the efficacy and mechanism of individual treatments targeting stroke recovery are better understood, combinations of existing and novel therapies will warrant investigation.
Stroke recovery trials offer the opportunity for rigorous testing, e.g., at baseline as entry criteria or stratification variables. Serum, genetic, imaging, and neurophysiologic data may be useful as predictors84. For example, motor response to TMS85 differentiates patients likely to experience spontaneous motor recovery from those who will not. An imaging-based measure of corticospinal tract injury can prospectively identify responders to rehabilitation therapy in chronic stroke86. Some of these measures can be serially collected as biomarkers of treatment effect.
The field of stroke recovery continues to benefit from the combination of perspectives of clinicians caring for stroke patients as well as the scientists elucidating biological mechanisms of recovery and developing new recovery-promoting technologies. These groups bring invaluable insights.
Acknowledgments
Sources of Funding
This work was supported by grants from the National Institutes of Health (R25NS065743, R44NS095381, and K24HD074722).
Footnotes
Disclosures
Dr. Lin has consulted for Boehringer Ingelheim; Dr. Cramer, for MicroTransponder, Roche, Dart Neuroscience, Neurolutions, Regenera, Abbvie, SanBio, and TRCare; Dr. Finklestein, for Constant Pharmaceuticals and is a Principal in Stemetix.
References
- 1.Bernhardt J, Hayward KS, Kwakkel G, Ward NS, Wolf SL, Borschmann K, et al. Agreed definitions and a shared vision for new standards in stroke recovery research: The stroke recovery and rehabilitation roundtable taskforce. Int J Stroke. 2017;12:444–450 [DOI] [PubMed] [Google Scholar]
- 2.Dobkin BH, Carmichael ST. The specific requirements of neural repair trials for stroke. Neurorehabilitation and Neural Repair. 2016;30:470–478 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cramer SC, Koroshetz WJ, Finklestein SP. The case for modality-specific outcome measures in clinical trials of stroke recovery-promoting agents. Stroke. 2007;38:1393–1395 [DOI] [PubMed] [Google Scholar]
- 4.Cramer SC, Wolf SL, Adams HP Jr., Chen D, Dromerick AW, Dunning K, et al. Stroke recovery and rehabilitation research: Issues, opportunities, and the National Institutes of Health StrokeNet. Stroke. 2017;48:813–819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pollock A, Farmer SE, Brady MC, Langhorne P, Mead GE, Mehrholz J, et al. Interventions for improving upper limb function after stroke. Cochrane database systematic reviews. 2014;11:CD010820-CD010820 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Thieme H, Mehrholz J, Pohl M, Behrens J, Dohle C. Mirror therapy for improving motor function after stroke. Cochrane Database Systematic Reviews. 2012:CD008449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Champod AS, Frank RC, Taylor K, Eskes GA. The effects of prism adaptation on daily life activities in patients with visuospatial neglect: A systematic review. Neuropsychological Rehabilitation. 2018;28:491–514 [DOI] [PubMed] [Google Scholar]
- 8.Wolf SL, Winstein CJ, Miller JP, Taub E, Uswatte G, Morris D, et al. Effect of constraint-induced movement therapy on upper extremity function 3 to 9 months after stroke. JAMA. 2006;296:2095. [DOI] [PubMed] [Google Scholar]
- 9.Dromerick AW, Lang CE, Birkenmeier RL, Wagner JM, Miller JP, Videen TO, et al. Very early constraint-induced movement during stroke rehabilitation (VECTORS): A single-center RCT. Neurology. 2009;73:195–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kwakkel G, Veerbeek JM, van Wegen EEH, Wolf SL. Constraint-induced movement therapy after stroke. The Lancet Neurology. 2015;14:224–234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chollet F, Tardy J, Albucher JF, Thalamas C, Berard E, Lamy C, et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): A randomised placebo-controlled trial. Lancet Neurol. 2011;10:123–130 [DOI] [PubMed] [Google Scholar]
- 12.Page SJ, Fulk GD, Boyne P. Clinically important differences for the upper-extremity Fugl-Meyer scale in people with minimal to moderate impairment due to chronic stroke. Phys Ther. 2012;92:791–798 [DOI] [PubMed] [Google Scholar]
- 13.Mead GE, Hsieh CF, Lee R, Kutlubaev M, Claxton A, Hankey GJ, et al. Selective serotonin reuptake inhibitors for stroke recovery: A systematic review and meta-analysis. Stroke. 2013;44:844–850 [DOI] [PubMed] [Google Scholar]
- 14.Maes M, Leonard B, Fernandez A, Kubera M, Nowak G, Veerhuis R, et al. (neuro)inflammation and neuroprogression as new pathways and drug targets in depression: From antioxidants to kinase inhibitors. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:659–663 [DOI] [PubMed] [Google Scholar]
- 15.Maya Vetencourt JF, Sale A, Viegi A, Baroncelli L, De Pasquale R, O’Leary OF, et al. The antidepressant fluoxetine restores plasticity in the adult visual cortex. Science. 2008;320:385–388 [DOI] [PubMed] [Google Scholar]
- 16.Scheidtmann K, Fries W, Muller F, Koenig E. Effect of levodopa in combination with physiotherapy on functional motor recovery after stroke: A prospective, randomised, double-blind study. Lancet. 2001;358:787–790 [DOI] [PubMed] [Google Scholar]
- 17.Ford G, Farrin A, Hartley S, Cozens A, Holloway I, Meads D, et al. The DARS (Dopamine Augmented Rehabilitation in Stroke) trial. 10th UK Stroke Forum Conference 2015 (abstract) [Google Scholar]
- 18.Pearson-Fuhrhop KM, Minton B, Acevedo D, Shahbaba B, Cramer SC. Genetic variation in the human brain dopamine system influences motor learning and its modulation by L-Dopa. PLoS One. 2013;8:e61197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wang LE, Fink GR, Diekhoff S, Rehme AK, Eickhoff SB, Grefkes C. Noradrenergic enhancement improves motor network connectivity in stroke patients. Ann Neurol. 2011;69:375–388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Barrett KM, Brott TG, Brown RD Jr., Carter RE, Geske JR, Graff-Radford NR, et al. Enhancing recovery after acute ischemic stroke with donepezil as an adjuvant therapy to standard medical care: Results of a phase iia clinical trial. Journal of stroke and cerebrovascular diseases. 2011;20:177–182 [DOI] [PubMed] [Google Scholar]
- 21.Martinsson L, Hardemark H, Eksborg S. Amphetamines for improving recovery after stroke. Cochrane Database Syst Rev. 2007:CD002090. [DOI] [PubMed] [Google Scholar]
- 22.Clarkson AN, Huang BS, Macisaac SE, Mody I, Carmichael ST. Reducing excessive GABA-mediated tonic inhibition promotes functional recovery after stroke. Nature. 2010;468:305–309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Clarkson AN, Overman JJ, Zhong S, Mueller R, Lynch G, Carmichael ST. AMPA receptor-induced local brain-derived neurotrophic factor signaling mediates motor recovery after stroke. J Neurosci. 2011;31:3766–3775 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Chen CL, Young SH, Gan HH, Singh R, Lao AY, Baroque AC, 2nd, et al. Chinese medicine neuroaid efficacy on stroke recovery: A double-blind, placebo-controlled, randomized study. Stroke. 2013;44:2093–2100 [DOI] [PubMed] [Google Scholar]
- 25.Finklestein SP, Caday CG, Kano M, Berlove DJ, Hsu CY, Moskowitz M, et al. Growth factor expression after stroke. Stroke. 1990;21:III122-124 [PubMed] [Google Scholar]
- 26.Finklestein S, Ren J. Growth factors as treatments for stroke In: Cramer SC, Nudo RJ, eds. Brain repair after stroke. Cambridge, UK: Cambridge University Press; 2010:259–266. [Google Scholar]
- 27.Kawamata T, Dietrich WD, Schallert T, Gotts JE, Cocke RR, Benowitz LI, et al. Intracisternal basic fibroblast growth factor enhances functional recovery and up-regulates the expression of a molecular marker of neuronal sprouting following focal cerebral infarction. PNAS. 1997;94:8179–8184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ren JM, Finklestein SP. Growth factor treatment of stroke. Current drug targets. CNS and neurological disorders. 2005;4:121–125 [DOI] [PubMed] [Google Scholar]
- 29.Kolb B, Morshead C, Gonzalez C, Kim M, Gregg C, Shingo T, et al. Growth factor-stimulated generation of new cortical tissue and functional recovery after stroke damage to the motor cortex of rats. J Cereb Blood Flow Metab. 2007;27:983–997 [DOI] [PubMed] [Google Scholar]
- 30.Belayev L, Khoutorova L, Zhao KL, Davidoff AW, Moore AF, Cramer SC. A novel neurotrophic therapeutic strategy for experimental stroke. Brain Res. 2009;1280:117–123 [DOI] [PubMed] [Google Scholar]
- 31.Jerndal M, Forsberg K, Sena ES, Macleod MR, O’Collins VE, Linden T, et al. A systematic review and meta-analysis of erythropoietin in experimental stroke. J Cereb Blood Flow Metab. 2010;30:961–968 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cramer SC, Fitzpatrick C, Warren M, Hill MD, Brown D, Whitaker L, et al. The beta-hcg+erythropoietin in acute stroke (BETAS) study: A 3-center, single-dose, open-label, noncontrolled, phase iia safety trial. Stroke. 2010;41:927–931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cramer SC, Hill MD. Human choriogonadotropin and epoetin alfa in acute ischemic stroke patients (REGENESIS-LED trial). International journal of stroke. 2014;9:321–327 [DOI] [PubMed] [Google Scholar]
- 34.Ehrenreich H, Weissenborn K, Prange H, Schneider D, Weimar C, Wartenberg K, et al. Recombinant human erythropoietin in the treatment of acute ischemic stroke. Stroke. 2009;40:e647-656 [DOI] [PubMed] [Google Scholar]
- 35.Li S, Carmichael ST. Growth-associated gene and protein expression in the region of axonal sprouting in the aged brain after stroke. Neurobiol Dis. 2006;23:362–373 [DOI] [PubMed] [Google Scholar]
- 36.Cramer SC, Abila B, Scott NE, Simeoni M, Enney LA. Safety, pharmacokinetics, and pharmacodynamics of escalating repeat doses of GSK249320 in patients with stroke. Stroke. 2013;44:1337–1342 [DOI] [PubMed] [Google Scholar]
- 37.Cramer SC, Enney LA, Russell CK, Simeoni M, Thompson TR. Proof-of-concept randomized trial of the monoclonal antibody GSK249320 versus placebo in stroke patients. Stroke. 2017;48:692–698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hess DC, Wechsler LR, Clark WM, Savitz SI, Ford GA, Chiu D, et al. Safety and efficacy of multipotent adult progenitor cells in acute ischaemic stroke (MASTERS): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Neurol. 2017;16:360–368 [DOI] [PubMed] [Google Scholar]
- 39.Kalladka D, Sinden J, Pollock K, Haig C, McLean J, Smith W, et al. Human neural stem cells in patients with chronic ischaemic stroke (PISCES): A phase 1, first-in-man study. Lancet. 2016;388:787–796 [DOI] [PubMed] [Google Scholar]
- 40.Steinberg GK, Kondziolka D, Wechsler LR, Lunsford LD, Coburn ML, Billigen JB, et al. Clinical outcomes of transplanted modified bone marrow-derived mesenchymal stem cells in stroke: A phase 1/2a study. Stroke. 2016;47:1817–1824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Khedr EM, Ahmed MA, Fathy N, Rothwell JC. Therapeutic trial of repetitive transcranial magnetic stimulation after acute ischemic stroke. Neurology. 2005;65:466–468 [DOI] [PubMed] [Google Scholar]
- 42.Kirton A, Chen R, Friefeld S, Gunraj C, Pontigon AM, Deveber G. Contralesional repetitive transcranial magnetic stimulation for chronic hemiparesis in subcortical paediatric stroke: A randomised trial. Lancet Neurol. 2008;7:507–513 [DOI] [PubMed] [Google Scholar]
- 43.Hummel F, Celnik P, Giraux P, Floel A, Wu WH, Gerloff C, et al. Effects of non-invasive cortical stimulation on skilled motor function in chronic stroke. Brain. 2005;128:490–499 [DOI] [PubMed] [Google Scholar]
- 44.Hao Z, Wang D, Zeng Y, Liu M. Repetitive transcranial magnetic stimulation for improving function after stroke. Cochrane Database Syst Rev. 2013;5: CD008862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Elsner B, Kugler J, Pohl M, Mehrholz J. Transcranial direct current stimulation (tDCS) for improving activities of daily living, and physical and cognitive functioning, in people after stroke. Cochrane Database Systematic Reviews. 2016;3:CD009645-CD009645 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Suntrup-Krueger S, Ringmaier C, Muhle P, Wollbrink A, Kemmling A, Hanning U, et al. Randomized trial of transcranial direct current stimulation for poststroke dysphagia. Ann Neurol. 2018;83:328–340 [DOI] [PubMed] [Google Scholar]
- 47.Fridriksson J, Rorden C, Elm J, Sen S, George MS, Bonilha L. Transcranial direct current stimulation vs sham stimulation to treat aphasia after stroke: A randomized clinical trial. [published online Aug 20, 2018] JAMA Neurol. 2018. https://jamanetwork.com/journals/jamaneurology/fullarticle/2696529. Accessed October 4, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Levy RM, Harvey RL, Kissela BM, Winstein CJ, Lutsep HL, Parrish TB, et al. Epidural electrical stimulation for stroke rehabilitation: Results of the prospective, multicenter, randomized, single-blinded everest trial. Neurorehabil Neural Repair. 2016;30:107–119 [DOI] [PubMed] [Google Scholar]
- 49.Meyers EC, Solorzano BR, James J, Ganzer PD, Lai ES, Rennaker RL 2nd, et al. Vagus nerve stimulation enhances stable plasticity and generalization of stroke recovery. Stroke. 2018;49:710–717 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dawson J, Pierce D, Dixit A, Kimberley TJ, Robertson M, Tarver B, et al. Safety, feasibility, and efficacy of vagus nerve stimulation paired with upper-limb rehabilitation after ischemic stroke. Stroke. 2016;47:143–150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Veerbeek JM, Langbroek-Amersfoort AC, van Wegen EE, Meskers CG, Kwakkel G. Effects of robot-assisted therapy for the upper limb after stroke. Neurorehab Neural Re. 2017;31:107–121 [DOI] [PubMed] [Google Scholar]
- 52.Lo AC, Guarino PD, Richards LG, Haselkorn JK, Wittenberg GF, Federman DG, et al. Robot-assisted therapy for long-term upper-limb impairment after stroke. N Engl J Med. 2010;362:1772–1783 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wagner TH, Lo AC, Peduzzi P, Bravata DM, Huang GD, Krebs HI, et al. An economic analysis of robot-assisted therapy for long-term upper-limb impairment after stroke. Stroke. 2011;42:2630–2632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Volpe BT, Lynch D, Rykman-Berland A, Ferraro M, Galgano M, Hogan N, et al. Intensive sensorimotor arm training mediated by therapist or robot improves hemiparesis in patients with chronic stroke. Neurorehabilitation and neural repair. 2008;22:305–310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Klamroth-Marganska V, Blanco J, Campen K, Curt A, Dietz V, Ettlin T, et al. Three-dimensional, task-specific robot therapy of the arm after stroke: A multicentre, parallel-group randomised trial. Lancet Neurology. 2014;13:159–166 [DOI] [PubMed] [Google Scholar]
- 56.Mehrholz J, Pohl M, Platz T, Kugler J, Elsner B. Electromechanical and robot-assisted arm training for improving activities of daily living, arm function, and arm muscle strength after stroke. The Cochrane database systematic reviews. 2015;11:CD006876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mehrholz J, Thomas S, Werner C, Kugler J, Pohl M, Elsner B. Electromechanical-assisted training for walking after stroke: A major update of the evidence. Stroke. 2017:188–190 [DOI] [PubMed] [Google Scholar]
- 58.Duncan PW, Sullivan KJ, Behrman AL, Azen SP, Wu SS, Nadeau SE, et al. Body-weight-supported treadmill rehabilitation after stroke. New Engl J Med. 2011;364:2026–2036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Winstein CJ, Stein J, Arena R, Bates B, Cherney LR, Cramer SC, et al. Guidelines for adult stroke rehabilitation and recovery: A guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2016;47:e98–e169 [DOI] [PubMed] [Google Scholar]
- 60.Zheng M-X, Hua X-Y, Feng J-T, Li T, Lu Y-C, Shen Y-D, et al. Trial of contralateral seventh cervical nerve transfer for spastic arm paralysis. New Engl J Med. 2018;378:22–34 [DOI] [PubMed] [Google Scholar]
- 61.Hochberg LR, Bacher D, Jarosiewicz B, Masse NY, Simeral JD, Vogel J, et al. Reach and grasp by people with tetraplegia using a neurally controlled robotic arm. Nature. 2012;485:372–375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chaudhary U, Birbaumer N, Ramos-Murguialday A. Brain-computer interfaces for communication and rehabilitation. Nat Rev Neurol. 2016;12:513–525 [DOI] [PubMed] [Google Scholar]
- 63.Ramos-Murguialday A, Broetz D, Rea M, Laer L, Yilmaz O, Brasil FL, et al. Brain-machine interface in chronic stroke rehabilitation: A controlled study. Ann Neurol. 2013;74:100–108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bundy DT, Souders L, Baranyai K, Leonard L, Schalk G, Coker R, et al. Contralesional brain-computer interface control of a powered exoskeleton for motor recovery in chronic stroke survivors. Stroke. 2017;48:1908–1915 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.van der Lee J, Beckerman H, Lankhorst G, Bouter L. The responsiveness of the Action Research Arm Test and the Fugl-Meyer assessment scale in chronic stroke patients. J Rehabil Med. 2001;33:110–113. [DOI] [PubMed] [Google Scholar]
- 66.Dodakian L, McKenzie AL, Le V, See J, Pearson-Fuhrhop K, Burke Quinlan E, et al. A home-based telerehabilitation program for patients with stroke. Neurorehabil Neural Repair. 2017;31:923–933 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lang CE, Macdonald JR, Reisman DS, Boyd L, Jacobson Kimberley T, Schindler-Ivens SM, et al. Observation of amounts of movement practice provided during stroke rehabilitation. Arch Phys Med Rehabil. 2009;90:1692–1698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Nudo R, Wise B, SiFuentes F, Milliken G. Neural substrates for the effects of rehabilitative training on motor recovery after ischemic infarct. Science. 1996;272:1791–1794 [DOI] [PubMed] [Google Scholar]
- 69.Chen J, Jin W, Zhang X-X, Xu W, Liu X-N, Ren C-C. Telerehabilitation approaches for stroke patients: Systematic review and meta-analysis of randomized controlled trials. Journal of Stroke Cerebrovascular Diseases. 2015;24:2660–2668 [DOI] [PubMed] [Google Scholar]
- 70.Sarfo FS, Ulasavets U, Opare-Sem OK, Ovbiagele B. Tele-rehabilitation after stroke: An updated systematic review of the literature. [published online June 4, 2018]. Journal of Stroke Cerebrovascular Diseases. 2018. https://www.sciencedirect.com/science/article/pii/S1052305718302313. Accessed October 4, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Veerbeek JM, Van Wegen E, Van Peppen R, Van Der Wees PJ, Hendriks E, Rietberg M, et al. What is the evidence for physical therapy poststroke? A systematic review and meta-analysis. PLoS One. 2014;9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Winstein CJ, Wolf SL, Dromerick AW, Lane CJ, Nelsen MA, Lewthwaite R, et al. Effect of a task-oriented rehabilitation program on upper extremity recovery following motor stroke: The ICARE randomized clinical trial. JAMA. 2016;315:571–581 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lewthwaite R, Winstein CJ, Lane CJ, Blanton S, Wagenheim BR, Nelsen MA, et al. Accelerating stroke recovery: Body structures and functions, activities, participation, and quality of life outcomes from a large rehabilitation trial. Neurorehabilitation Neural Repair. 2018;32:150–165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Basso DM, Lang CE. Consideration of dose and timing when applying interventions after stroke and spinal cord injury. Journal Neurologic Physical Therapy. 2017;41:S24-S31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Jeffers MS, Corbett D. Synergistic effects of enriched environment and task-specific reach training on poststroke recovery of motor function. Stroke. 2018;49:1496–1503 [DOI] [PubMed] [Google Scholar]
- 76.Biernaskie J, Chernenko G, Corbett D. Efficacy of rehabilitative experience declines with time after focal ischemic brain injury. Journal Neuroscience. 2004;24:1245–1254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Saposnik G, Cohen LG, Mamdani M, Pooyania S, Ploughman M, Cheung D, et al. Efficacy and safety of non-immersive virtual reality exercising in stroke rehabilitation (EVREST): A randomised, multicentre, single-blind, controlled trial. Lancet Neurology. 2016;15:1019–1027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Laver KE, Lange B, George S, Deutsch JE, Saposnik G, Crotty M. Virtual reality for stroke rehabilitation. Stroke. 2018;49:e160-e161 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kiper P, Szczudlik A, Agostini M, Opara J, Nowobilski R, Ventura L, et al. Virtual reality for upper limb rehabilitation in sub-acute and chronic stroke: A randomized controlled trial. Archives Physical Medicine and Rehabilitation. 2018;99:834–842 [DOI] [PubMed] [Google Scholar]
- 80.Aminov A, Rogers JM, Middleton S, Caeyenberghs K, Wilson PH. What do randomized controlled trials say about virtual rehabilitation in stroke? A systematic literature review and meta-analysis of upper-limb and cognitive outcomes. Journal NeuroEngineering and Rehabilitation. 2018;15:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Dobkin BH, Dorsch AK. The evolution of personalized behavioral intervention technology. Stroke. 2017;48:2329–2334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dorsch AK, Thomas S, Xu X, Kaiser W, Dobkin BH. Sirract: An international randomized clinical trial of activity feedback during inpatient stroke rehabilitation enabled by wireless sensing. Neurorehabilitation Neural Repair. 2015;29:407–415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Powell L, Parker J, Martyn St-James M, Mawson S. The effectiveness of lower-limb wearable technology for improving activity and participation in adult stroke survivors: A systematic review. Journal medical Internet research. 2016;18:e259-e259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Boyd LA, Hayward KS, Ward NS, Stinear CM, Rosso C, Fisher RJ, et al. Biomarkers of stroke recovery: Consensus-based core recommendations from the stroke recovery and rehabilitation roundtable. Int J Stroke. 2017;12:480–493 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Stinear CM, Byblow WD, Ackerley SJ, Smith MC, Borges VM, Barber PA. Prep2: A biomarker-based algorithm for predicting upper limb function after stroke. Ann Clin Transl Neurol. 2017;4:811–820 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Cassidy JM, Tran G, Quinlan EB, Cramer SC. Neuroimaging identifies patients most likely to respond to a restorative stroke therapy. Stroke. 2018;49:433–438 [DOI] [PMC free article] [PubMed] [Google Scholar]