Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 1.
Published in final edited form as: Expert Rev Neurother. 2016 Jul 15;16(12):1397–1405. doi: 10.1080/14737175.2016.1207530

Potential new complication in drug therapy development for amyotrophic lateral sclerosis

Svitlana Garbuzova-Davis 1,2,3,4,*, Avery Thomson 5, Crupa Kurien 1, R Douglas Shytle 1,2, Paul R Sanberg 1,2,4,6
PMCID: PMC5164916  NIHMSID: NIHMS834820  PMID: 27362330

Abstract

Introduction

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron degeneration in the brain and spinal cord. Treatment development for ALS is complicated by complex underlying disease factors.

Areas covered

Numerous tested drug compounds have shown no benefits in ALS patients, although effective in animal models. Discrepant results of pre-clinical animal studies and clinical trials for ALS have primarily been attributed to limitations of ALS animal models for drug-screening studies and methodological inconsistencies in human trials. Current status of pre-clinical and clinical trials in ALS is summarized. Specific blood-CNS barrier damage in ALS patients, as a novel potential reason for the clinical failures in drug therapies, is discussed.

Expert opinion

Pathological perivascular collagen IV accumulation, one unique characteristic of barrier damage in ALS patients, could be hindering transport of therapeutics to the CNS. Restoration of B-CNS-B integrity would foster delivery of therapeutics to the CNS.

Keywords: ALS, animal models, blood-CNS barrier, clinical trials, drug therapy, patients

1. Introduction

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by gradual motor neuron degeneration throughout the neural axis. The rapid progression of debilitating symptoms of ALS leads to paralysis and fatality [1,2]. Approximately 50% of patients die within 30 months of disease symptom onset and only 20% of patients survive 5 to10 years after symptom onset [3]. In the United States, estimated ALS incidence and prevalence are 5.4/100,000 and 10.5/100,000, respectively [4,5] with an estimated 5,000 new cases annually [6]. ALS cases are 90–95% sporadic (SALS) with the remaining 5–10% of cases being genetically linked, familial (FALS). Peak ages for disease onset are 58–63 years for SALS and 47–52 years for FALS [7]. Men have a higher incidence of this disease than women. The overall population-based lifetime risk for ALS is 1:350 for men and 1:400 for women. Within FALS cases, approximately 20% result from missense mutations in the genes coding for Cu/Zn superoxide dismutase (SOD1) [8,9] and about 2–5% have mutations of the TARDBP gene encoding the TAR-DNA binding protein TDP-43 [10]. Additional gene mutations in FUS [11,12] and ANG [13], approximately 5% and 1% of FALS cases respectively, have been identified. Relatively recently, a mutation in the C9ORF72 gene was identified and has been associated with a significant number of FALS, FALS with frontotemporal dementia, and SALS cases [14,15]. The clinical presentation and underlying pathology of SALS and FALS are similar and treatment options for ALS patients are limited and mainly supportive. The only FDA approved drug to treat ALS is riluzole (Rilutek®), which blocks glutamate release and extends the lifespan of ALS patients by only a few months [16].

Since French neurologist Jean-Martin Charcot first described ALS as a rapidly progressive neuromuscular disease in 1869 [17], there have been growing clinical and scientific efforts to understand the disease pathogenesis. Numerous hypotheses about the etiopathology of ALS have been proposed (reviewed in [1829]), including glutamate excitotoxicity, mitochondria dysfunction, oxidative stress, glial cell pathology, impaired axonal transport, protein aggregations, immune reactivity, neurotrophic factor deficits, and neuroinflammation.

Development of an effective treatment for ALS is complicated by the diffuse nature of motor neuron degeneration and by the complexity of intrinsic and extrinsic factors underlying this disease. Although more than 30 drug compounds have been tested in ALS clinical trials, most drugs failed to show benefit in ALS patients [30,31].

The present review briefly summarizes current status and potential pitfalls of pre-clinical and clinical trials in ALS. A new potential cause for the observed ALS clinical trial failures is discussed.

2. Power of pre-clinical studies in successful clinical trials for ALS

The therapeutic strategies for ALS have primarily focused on pharmacological interventions to prevent and/or delay motor neuron deterioration and thereby slow or arrest disease progression. These disease-modifying neuroprotective strategies target motor neurons. However, none of the agents tested for their neuroprotective role in human studies demonstrated a significant benefit in survival or quality of life of ALS patients, although each of these compounds showed promising results in animal models. For example, brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) as potent drugs for ALS failed to demonstrate efficacy in clinical trials [3234], although improvements in motor neuron protection were demonstrated in preliminary studies of axotomized rats or wobbler mice as relevant models of ALS [3538].

Similarly, insulin-like growth factor-1 (IGF-1), including the recombinant form, delivered intrathecally or intraparenchymally into a superoxide dismutase 1 (SOD-1) rodent model of ALS demonstrated beneficial effects by prolonging survival and increasing motor function in a number of reports [3942]. However, the reported improvement in SOD-1 animal survival was controversial [43,44]. Despite the controversy, intrathecal [45] and subcutaneous [46] administration of IGF-1 were investigated in later clinical trials. Although some benefits were shown in patients receiving a high dose of IGF-1 via intrathecal delivery, ALS patients with subcutaneous drug treatment showed no improvement in Phase III clinical trials. It has been suggested that intrathecal IGF-1 treatment might have clinical significance but development of better drug delivery mechanisms strength “important for validating the utility of IGF-1 as a therapy for ALS” [47]. Detailed discussions regarding pre-clinical and clinical trials of the above mentioned and other growth factors (glial-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), etc.) are available [48,49]. It is believed that neurotrophic growth factors, which might promote motor neuron survival, are the most rational therapeutic approach for ALS.

Minocycline, a tetracycline antibiotic, also demonstrated positive effects in animal studies by delaying disease progression and extending lifespan in SOD-1 mutant mice [5052]. This beneficial drug effect resulted from attenuated microglial activation in the spinal cord of treated animals at the end stage of disease. However, results of a Phase III clinical trial were negative and escalated doses of minocycline even proved harmful [53,54]. A reasonable question was raised by Keller at al. [55] regarding glial cells as a potential therapeutic target in ALS. The authors performed a study to evaluate the effect of minocycline treatments initiated at different stages of disease in SOD-1 G93A mice. Results of this study, showing altered astrocyte reactivity and increased microgliosis at late stage of disease, suggest that minocycline lacks anti-inflammatory actions.

Similarly to minocycline treatment, creatine monohydrate, a potential cellular energy supplier, showed no obvious therapeutic benefit on survival or disease progression in ALS patients in double-blind placebo-controlled studies [5658], although benefitting motor function and survival of transgenic mice modeling ALS [59].

Additionally, celecoxib, a cyclooxygenase-2 inhibitor, which reduced astrocytic glutamate release, was effective in increasing motor neuron survival and prolonging lifespan of G93A mice [60], but showed no benefits in a double-blind, placebo-controlled clinical trial [61]. Another compound, ceftriaxone, efficiently prolonged survival of G93A animals via increased glial glutamate transporter expression in astrocytes [62]. Nevertheless this drug did not show clinical efficacy in a Phase III clinical trial [63]. Significantly more adverse gastrointestinal and hepatobiliary events were detected in ALS patients treated with ceftriaxone than in the placebo group.

Also, memantine, a noncompetitive NMDA receptor antagonist and a neuroprotective agent against glutamate-induced toxicity, initially demonstrated therapeutic efficacy in G93A SOD 1 mice by significantly delaying disease progression and increasing lifespan of mice [64], even when administered into symptomatic animals [65]. When this drug was evaluated in a Phase II/III double-blind, randomized clinical trial for 12 months, there were no adverse events but efficacy of memantine was not observed [66]. Interestingly, another multi-center double-blind, placebo controlled pilot study investigating the effect of memantine in ALS patients who took riluzole by examining changes in CSF biomarkers demonstrated that the drug was well tolerated with declines in CSF tau levels, even to healthy control levels in some patients [67]. Thus, combined treatment of memantine with riluzole might be efficacious for ALS patients. However, the effect of this combinatory treatment in an animal model of disease was not determined.

Since riluzole, an antiglutamaterdic agent, is the only FDA approved drug to treat ALS patients [16], combining riluzole with new compound(s) might be an effective therapeutic strategy. Early studies by Gurney et al. [68,69] showed that riluzole significantly maintained motor function and prolonged survival in a transgenic mouse model of ALS, but disease onset was not delayed. The effect of riluzole on motor function in mice was better in early-stage disease than later [69], suggesting that early drug intervention in ALS patients might be beneficial [70]. Supporting this suggestion, reduced effectiveness of riluzole in enhancing glutamate uptake was noted at end-stage of disease in a transgenic rat model of ALS [71]. Although riluzole modestly prolongs survival of ALS patients by 4–6 months [72,73], there is an urgent need to develop therapy benefiting both survival and quality of life for ALS patients,

Recently, a randomized, placebo-controlled, double-blind Phase IIb trial evaluated the safety and efficacy of tirasemtiv, a fast skeletal troponin activator, for 12 weeks in ALS patients from 73 centers in eight countries [74]. The application for this clinical trial was based on beneficial results demonstrating muscle and motor function improvement in the G93A SOD1 mouse model [75]. However, no drug effect was shown in the changes in ALSFRS-R from baseline but losses of vital capacity and muscle strength were significantly slowed in tirasemtiv vs. placebo group. Yet, serious adverse events were determined more frequently in the tirasemtiv group. Besides the negative study results, the authors suggest “a potentially important effect of tirasemtiv warranting further evaluation over a longer period in ALS” [74].

The above descriptions of drug therapies in animal studies and ALS clinical trials are illustrative, rather than comprehensive. However, the obvious discrepancy between results of pre-clinical animal studies and following clinical trials is disappointing. Is the dismal record of ALS clinical trials due to inadequacies in the animal models or are weaknesses endemic in the designs of these clinical trials?

Transgenic animal models of ALS carrying SOD-1gene mutations are widely used in translational treatment research. Although ALS pathogenesis in these animal models seems to mimic human disease, the rapid course of disease progression is only one of several challenges to accurate evaluation of experimental drug efficacy. To improve pre-clinical study research, guidelines for standardized drug testing methods in rodent models of ALS have been provided [76]. Importantly, consistent methodologies, including those for toxicity and dose-response effects, should be applied in pre-clinical investigations. In agreement with Scott et al. [77], confounding biological variables should be also considered in designing and interpreting drug efficacy studies, particularly investigations using G93A SOD-1 mice, which carry approximately 23 copies of the human SOD-1 G93A transgene. Interestingly, after identifying the most critical biological variables and retesting the efficacy of various compounds in G93A mice using an optimized study design, the authors found no survival benefit in mice for minocycline, creatine or other tested drugs. They stated that the majority of studies showing previous beneficial effects were “most likely measurements of noise in the distribution of survival means as opposed to actual drug effect” due to the presence of uncontrolled confounding variables [77]. The authors provided recommendations on improved pre-clinical survival study design for therapeutic drug testing in the G93A mouse. Also, pharmacodynamic and pharmacokinetic data on drugs tested in animal disease models might help identify useful agents for future ALS clinical trials [31]. Additionally, the majority of ALS studies using animal models began treatment prior to symptom onset, so investigations treating animals at late symptomatic disease stage should have increased translational value [48,78,79]. However, some reviews [7981] have noted that the current FALS animal models may lack utility for identifying therapeutic agents and/or evaluating drug efficacy for clinical trials of SALS patients.

Numerous comprehensive reviews have deliberated the limitations and future directions of pre-clinical studies and clinical trials for ALS [2931,48,49,78,79,8284]. In remarks on ALS clinical drug trials, the failures were largely attributed to methodological pitfalls including: insufficient numbers of enrolled participants, heterogeneous patient populations, short duration of treatment, discrepant timing of patient drug treatments accordingly to their disease stages (vs. pre-clinical studies), absence of placebo-controlled patient groups, lack of clinically implemented specific biomarkers in evaluation of drug efficacy, and others. However, there is also ongoing debate regarding the optimal cellular target for ALS therapy. Ludolph and Jesse [82] noted that rational pharmacological intervention in disease-modifying strategies aims to prevent further deterioration of motor neurons throughout the neural axis. In this view, a current Phase II randomized, double-blind pharmacodynamic trial (NCT02450552) is evaluating the effect of oral ezogabine (retigabine) treatment on upper and lower motor neuron excitability in ALS patients. The effect of two doses of retigabine, a potassium channel opener, vs. placebo is determined by transcranial magnetic stimulation and threshold tracking nerve conduction studies. This clinical trial could be beneficial since pre-trial in vitro study results showed blocking of hyperexcitability and improving motor neuron survival using induced pluripotent stem cell-derived motor neurons from ALS patients harboring mutant SOD1 [85].

However, as the disease progresses, motor neurons become subject to neuroinflammation via reactive astrocytes and activated microglia. Hence, glial cells might be a potential therapeutic target and initiation of this targeted drug treatment could depend on disease stage. Hereafter, one consideration for drug intervention studies should be selection of the appropriate cellular target due to evolving dynamic interactions between motor neurons and glial cells [78]. Also, as Kiernan et al. [1] note: “There is a crucial need to formulate therapies that not only slow disease progression, but also deal with the secondary consequences of malnutrition and respiratory failure.” Therefore, the development of effective drug therapies for ALS has a major challenge due to multiple and variable targets for therapeutic interventions. A combined drug treatment, “cocktail therapy”, is a likely and promising next step for clinical trials in ALS [29,79,83]. Even combinations of approved drugs using in treatment of other diseases might serve as potential therapeutic agents for ALS [86]. Also, gene therapy might have a significant role in treatment development for ALS by delivering various therapeutic molecules using viral/non-viral vectors across the blood-brain and blood-spinal cord barriers.

Although major problems have been identified in both pre-clinical research and clinical trials in ALS, solid animal drug-screening studies are necessary to select drugs for future human studies. Since 2010, a zebrafish genetic model of ALS has been widely used for drug testing [87,88]. This simple animal model for ALS might facilitate screening of chemical agents and even drug discovery via various genetic manipulations revealing new therapeutic targets [89]. Although the zebrafish model of ALS has numerous advantages for pre-clinical drug screening, some neuroanatomical differences were noted in the motor system vs. humans [90]. Also, it has been shown that zebrafish display a mature blood-brain barrier (BBB) in cerebral microvessels between 3 and 10 days post-fertilization and both structural and functional characteristics of BBB are similar to that of mammals [9193]. However, whether the BBB in zebrafish accurately reflects BBB status in humans and mimics specific BBB pathological conditions related to ALS, as discussed below, needs future investigation. Thus, improved translational investigations as well as clinical trials with proper methodological design are hallmarks for successful drug treatment in ALS. However, one aspect of any effective drug therapy for ALS needs further discussion. Since therapeutic compounds must reach their target within the CNS, they need the ability to cross the blood-CNS-barrier. However, some potent agents considered for ALS therapy such as GDNF do not cross the barrier [94] while others such as VEGF can cross this barrier [95,96]. This review considers the blood-CNS-barrier as an integrated system allowing delivery of therapeutics to the CNS rather than focuses on particular barrier transport mechanisms.

3. Is blood-CNS barrier integrity critical for drug delivery to the CNS?

The CNS is protected from entry of hazardous serum proteins by the blood-brain barrier (BBB), blood-spinal cord barrier (BSCB), and blood-cerebrospinal fluid barrier (BCSFB). These CNS barriers control cerebral/spinal cord homeostasis by selective transport of various substances [97101]. This control is possible due to the unique structure of the microvasculature – capillaries formed by endothelial cells (BBB and BSCB) and epithelial cells of the choroids plexus (BCSFB). Brain and spinal cord capillary endothelial cells (ECs) are connected via adherens and tight junctions [102105]. The basement membrane (i.e. basal lamina) surrounding the ECs and pericytes supports the abluminal surface of the endothelium [106,107]. Astrocyte perivascular end-feet, ensheathing approximately 95% of the vessel wall, appear to have an important role for maintenance of the BBB/BSCB [108110]. Integrity of all BBB/BSCB elements is critical for protection of the CNS. Impairment of this cellular machinery may cause blood-CNS-barrier (B-CNS-B) breakdown.

In ALS, one possible pathogenic mechanism is impairment of the B-CNS-B. Although some early reports showed altered BCSFB permeability in ALS [111,112], later studies were inconclusive [113,114]. Relatively recently, however, compelling evidence demonstrated structural and functional alterations in the BBB and BSCB in both ALS patients and animal models [115122]. Major findings were degeneration of ECs and astrocyte end-feet processes surrounding microvessels, extra- and intracellular edema, impairment of endothelial transport system, and dysfunction of tight junction proteins compromising BBB/BSCB integrity, resulting in vascular leakage, microhemorrhages, and hemosiderin deposits in CNS parenchyma. This vascular pathology, demonstrating impairment of all neurovascular unit components in the brain and spinal cord, identifies ALS as a neurovascular disease [123]. It is possible that the initiating pathological trigger for ALS is a dysfunctional B-CNS-B, allowing detrimental factors from the systemic circulation to penetrate the CNS and rendering motor neurons susceptible to degeneration [124]. Supporting this suggestion, BSCB breakdown has been demonstrated in pre-symptomatic SOD1 mutant mice prior to motor neuron degeneration and inflammatory changes in the spinal cord [97]. Although B-CNS-B damage might not be a causative event in ALS, it is still a significant, and possibly critical, pathologic effector.

However, the majority of findings on ALS microvascular pathology have been determined in mutant SOD-1 rodent models, which might similarly occur in FALS patients carrying the SOD-1 mutations. To determine B-CNS-B competence without involvement of mutant SOD-1, our group recently analyzed post-mortem gray and white matter microvessels of medulla and spinal cord tissue from SALS patients and showed pervasive barrier damage in these CNS areas [117]. Furthermore, similarities and differences in barrier pathology between SALS patients and an FALS animal model were noted. Numerous signs of barrier impairment (EC degeneration, capillary leakage, perivascular edema, downregulation of tight junction proteins, and microhemorrhages) are common in both mutant SOD1 animal models of ALS and SALS patients, although other pathogenic barrier alterations such as pericyte degeneration and perivascular collagen IV expansion are still unique to SALS patients [125]. Our thoughts are that compromised BBB/BSCB integrity in animal models of ALS might be due to toxic effects of mutant SOD-1 upon ECs, but the cause(s) of barrier damage in SALS patients are still unclear. Pericyte degeneration, one noted difference between animal models and patients, might lead to the reduced capillary blood flow shown in brains of ALS patients and correlating with disease severity [126]. For instance, extensive vascular basement membrane collagen IV accumulation, 2–2.5 times higher than controls, and even collagen fiber calcifications were demonstrated in the majority of brain and spinal cord vessels from SALS patients [117]. It is possible that buildup of perivascular collagen IV occurs as a compensatory mechanism for maintenance of vascular integrity or/and due to defective regulation of the matrix metalloproteinase (MMP) pathway by damaged endothelial cells leading to a possible imbalance between MMPs and tissue inhibitors of metalloproteinases (TIMPs) [127]. It seems highly likely that abnormal perivascular collagen accumulation in SALS patients takes place over a long period of time and could present a significant obstacle to effective drug influx to the CNS. Also, thickening of the basement membrane by collagen IV accumulation would slow diffusion of various substances across the barrier [128]. Additionally, given the high oxygen consumption of motor neurons, the higher frequency of hypertension in ALS patients may reflect a compensatory increase in blood pressure in response to a thickened vascular basement membrane [129]. A population based study found that individuals whom had taken a high dose of angiotensin-converting enzyme inhibitor (a type of blood pressure medication) for at least four years had a 57% lower risk of developing ALS than the general population [130]. Thus, there is an obvious link between thickness of the vascular basement membrane and diffusion rates of various substances across the B-CNS-B.

Additionally, similarly to ALS, Alzheimer’s clinical drug trials have shown a disappointingly high rate of failures – 99.6% [131]. Identified trial weaknesses include a poor understanding of mechanisms underlying most late onset dementias and difficulties recruiting and retaining study members [132]. Interestingly, biopsied brain tissue from Alzheimer’s patients revealed collagen accumulation in cerebral vascular basement membranes in addition to focal necrotic changes in endothelial cells [133,134]. Recent animal studies employing the triple transgenic Alzheimer’s mouse model (3xTg-AD) reported reduced cerebrovascular volume [135], increased thickening of the epithelial basal membrane, and greater collagen-IV deposition around capillaries in choroid plexus [136]. Moreover, Mehta et al. [137] noted a significant reduction in the brain uptake of [3H] diazepam and [3H] propranolol, and the anti-AD drug, memantine, in aged 3×TG-AD mice versus wild type mice of the same age (up to 57% in the cortex and up to 62% in the hippocampus). A slower rate of drug diffusion in this animal model is consistent with the thickening of the perivascular basement membrane confirmed by collagen IV immunohistochemistry in cortical slices from these aged transgenic mice suggesting that the reduced uptake of the drugs was likely due to impaired perivascular basement membrane. Perhaps perivascular collagen IV buildup is hindering delivery of therapeutics to the brain in both ALS and AD.

Henriques et al. [48] noted that “we have to be certain that the protein in question passes the BBB, and reaches motoneurons in sufficient concentration, and over a reasonable time period. The delivery route and formulation need to be fitted to that purpose.” Although B-CNS-B related pathologies in SALS patients have been discussed in detail [125], it is important to emphasize that differences, especially perivascular collagen IV expansion, in B-CNS-B damage between ALS patients and animal models may explain ineffective drug therapy in clinical trials for ALS. Also, pericyte degeneration, leading to capillary blood flow changes, might hinder drug delivery for treatment of ALS. Although pericyte degeneration may be difficult to combat, collagen IV accumulation can be targeted, perhaps by administration of fenofibric acid [138], ramipril [139], anti-collagen IV antibodies or alpha, alpha’9-dipyridyl [140]. Reduction in collagen IV may aid new therapeutic strategies for ALS. Thus, drug deliveries to the CNS might be impeded, or even thwarted, by pervasive B-CNS-B damage in ALS, and this barrier should be considered the initial primary therapeutic target in any treatment approach for this disease.

4. Conclusion

ALS is a complex multifactorial disease and the development of effective drug therapies is challenged by multiple therapeutic targets. Disease-modifying strategies for ALS primarily focus on pharmacological interventions to prevent and/or delay further motor neuron deterioration and thereby, slow and/or arrest disease progression. However, none of the potent drugs in human studies significantly prolonged survival or improved patient quality of life, although these agents showed beneficial results in ALS animal models. This discrepancy between pre-clinical and clinical trials in drug efficacy for ALS is disappointing. Numerous reports have addressed the limitations in both animal studies and clinical trials for ALS. Mainly, the utility of current animal models of ALS for drug-screening studies has been questioned as these animals have imperfectly represented human disease outcomes. The primary shortcomings of ALS clinical trials have been identified as methodological inadequacies and/or inconsistencies between the various trials. The disconnect between effective drug therapies in ALS mice and unsuccessful drug treatments in ALS patients might be also point to more complex mechanisms underlying sporadic disease. Supporting this possibility, differences in blood-CNS barrier alterations have been shown between sporadic ALS patients and animal models of disease. Significant perivascular collagen IV accumulation, determined only in ALS patients, seems likely to restrict drug influx to the CNS, thereby attenuating therapeutic strategies in ALS clinical trials. Similarly to ALS, collagen IV accumulation may impact AD therapies. Thus, the blood-CNS barrier should be considered as a primary therapeutic target prior to development of any treatment approach for ALS as well as AD.

5. Expert Commentary

The mismatch between results of pre-clinical studies and ALS drug clinical trials is challenging for development of effective treatments. Although weaknesses have been identified in both animal studies and clinical trials, the key weakness is the failure to translate research findings into viable clinical benefits. Also, methodological pitfalls are indicated in numerous clinical trials. To address this point, therapeutic compounds rely upon their ability to cross the B-CNS-B. The pervasive B-CNS-B impairment found in ALS likely impedes drug delivery to the CNS. Perivascular collagen IV accumulation in ALS patients, one distinct difference from animal models of ALS, could be hindering transport of therapeutics to the CNS. Similarly, drug therapy failures in AD clinical trials might be attributed to basement membrane collagen IV expansion. If pharmacokinetic issues are confirmed, i.e. insufficient drug delivery to the CNS, then repair of the B-CNS-B might be an essential prerequisite to development of an effective therapy and collagen IV should be a primary therapeutic target. In summary, restoration of B-CNS-B integrity would foster delivery of therapeutics to the CNS as well as aid in removal of waste products from the CNS, providing an improved environment for motor neuron survival.

6. Five-year view

Effective therapy for ALS is an urgent need, yet developing treatment strategies for a multifactorial disease has proven extremely challenging. Despite intensive therapeutic development efforts, riluzole remains the only FDA approved ALS therapy and modestly extends patient lifespan.

The mismatch between effective drug therapies in animal model of ALS and unsuccessful drug treatments in ALS patients is disappointing and cause(s) of this discrepancy need to be identified and defeated. Animal models remain an essential tool for disease research but cannot be expected to provide a perfect window into human disease. Perhaps, an improved animal model of ALS, possibly primate, might facilitate effective therapy development for ALS in the near future. Also, increasing our knowledge regarding etiopathology of sporadic ALS should be addressed for identifying disease factors towards therapy development for all ALS cases.

Also, it is important to more deeply investigate pathogenic differences between animal models and human ALS. For example, differences in blood-CNS barrier alterations have been shown between sporadic ALS patients and animal models of disease. The specific barrier pathology found only in ALS patients might explain failed clinical trials in ALS. Blood-CNS barrier damage, a relatively recently discovered ALS pathology, may be a limiting factor in CNS drug delivery. The optimistic view is that: technological advances in the near future will allow non-invasive evaluation of barrier status in early stage ALS patients; effective therapies will be developed targeting barrier damage; and improved barrier status will promote the effective use of a variety of drug therapies for ALS.

7. Key issues.

  • There is a significant mismatch between results of pre-clinical studies (positive) and ALS clinical trials (negative)

  • B-CNS-B is pervasively damaged in ALS patients

  • Similarities and differences in B-CNS-B alterations have been shown in animal models of ALS and ALS patients

  • Perivascular Collagen IV accumulation in ALS patients is one distinct difference from ALS animal models

  • Basement membrane collagen IV buildup could hinder drug delivery across the blood-CNS-barrier

  • Repair of the B-CNS-B might be an essential prerequisite to development of an effective therapy for ALS

Acknowledgments

Funding

This review was supported by the Center for Excellence and Brain Repair, Department of Neurosurgery and Brain Repair at the University of South Florida. S Garbuzova-Davis is funded by NIH (1R01 NS090962-01).

Footnotes

Declaration of interests

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

References

Reference annotations

* Of interest

** Of considerable interest

  • 1.Kiernan MC, Vucic S, Cheah BC, et al. Amyotrophic lateral sclerosis. Lancet. 2011;377:942–55. doi: 10.1016/S0140-6736(10)61156-7. [DOI] [PubMed] [Google Scholar]
  • 2.Sorarù G, Ermani M, Logroscino G, et al. Natural history of upper motor neuron-dominant ALS. Amyotroph Lateral Scler. 2010;11:424–9. doi: 10.3109/17482960903300867. [DOI] [PubMed] [Google Scholar]
  • 3.Talbot K. Motor neuron disease: the bare essentials. Pract Neurol. 2009;9:303–9. doi: 10.1136/jnnp.2009.188151. [DOI] [PubMed] [Google Scholar]
  • 4.Mehta P, Antao V, Kaye W, et al. Prevalence of amyotrophic lateral sclerosis - United States, 2010–2011. MMWR Supplements. 2014;63:1–14. [PubMed] [Google Scholar]
  • 5.Chiò A, Logroscino G, Traynor BJ, et al. Global epidemiology of amyotrophic lateral sclerosis: a systematic review of the published literature. Neuroepidemiology. 2013;41:118–30. doi: 10.1159/000351153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hirtz D, Thurman DJ, Gwinn-Hardy K, et al. How common are the “common” neurologic disorders? Neurology. 2007;68:326–37. doi: 10.1212/01.wnl.0000252807.38124.a3. [DOI] [PubMed] [Google Scholar]
  • 7.Logroscino G, Traynor BJ, Hardiman O, et al. Incidence of amyotrophic lateral sclerosis in Europe. J Neurol Neurosurg Psychiatr. 2010;81:385–90. doi: 10.1136/jnnp.2009.183525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rosen DR, Siddique T, Patterson D, et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature. 1993;362:59–62. doi: 10.1038/362059a0. [DOI] [PubMed] [Google Scholar]
  • 9.Pasinelli P, Brown RH. Molecular biology of amyotrophic lateral sclerosis: insights from genetics. Nat Rev Neurosci. 2006;7:710–23. doi: 10.1038/nrn1971. [DOI] [PubMed] [Google Scholar]
  • 10.Yokoseki A, Shiga A, Tan C-F, et al. TDP-43 mutation in familial amyotrophic lateral sclerosis. Ann Neurol. 2008;63:538–42. doi: 10.1002/ana.21392. [DOI] [PubMed] [Google Scholar]
  • 11.Kwiatkowski TJ, Jr, Bosco DA, Leclerc AL, et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science. 2009;323:1205–8. doi: 10.1126/science.1166066. [DOI] [PubMed] [Google Scholar]
  • 12.Vance C, Rogelj B, Hortobágyi T, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science. 2009;323:1208–11. doi: 10.1126/science.1165942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Greenway MJ, Andersen PM, Russ C, et al. ANG mutations segregate with familial and “sporadic” amyotrophic lateral sclerosis. Nat Genet. 2006;38:411–3. doi: 10.1038/ng1742. [DOI] [PubMed] [Google Scholar]
  • 14.DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72:245–56. doi: 10.1016/j.neuron.2011.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 2011;72:257–68. doi: 10.1016/j.neuron.2011.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Miller RG, Mitchell JD, Lyon M, Moore DH. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND) Amyotroph Lateral Scler Other Motor Neuron Disord. 2003;4:191–206. [PubMed] [Google Scholar]
  • 17.Kumar DR, Aslinia F, Yale SH, Mazza JJ. Jean-Martin Charcot: the father of neurology. Clin Med Res. 2011;9:46–9. doi: 10.3121/cmr.2009.883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rothstein JD. Current hypotheses for the underlying biology of amyotrophic lateral sclerosis. Ann Neurol. 2009;65(Suppl 1):S3–9. doi: 10.1002/ana.21543. [DOI] [PubMed] [Google Scholar]
  • 19.Bruijn LI. Amyotrophic lateral sclerosis: from disease mechanisms to therapies. BioTechniques. 2002;32:1112, 1114, 1116. doi: 10.2144/02325dd01. passim. [DOI] [PubMed] [Google Scholar]
  • 20.Strong MJ, Kesavapany S, Pant HC. The pathobiology of amyotrophic lateral sclerosis: a proteinopathy? J Neuropathol Exp Neurol. 2005;64:649–664. doi: 10.1097/01.jnen.0000173889.71434.ea. [DOI] [PubMed] [Google Scholar]
  • 21.Wijesekera LC, Leigh PN. Amyotrophic lateral sclerosis. Orphanet J Rare Dis. 2009;4:3. doi: 10.1186/1750-1172-4-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Martin LJ, Price AC, Kaiser A, et al. Mechanisms for neuronal degeneration in amyotrophic lateral sclerosis and in models of motor neuron death (Review) Int J Mol Med. 2000;5:3–13. doi: 10.3892/ijmm.5.1.3. [DOI] [PubMed] [Google Scholar]
  • 23.Rodrigues MCO, Voltarelli JC, Sanberg PR, et al. Immunological aspects in amyotrophic lateral sclerosis. Transl Stroke Res. 2012;3:331–40. doi: 10.1007/s12975-012-0177-6. [DOI] [PubMed] [Google Scholar]
  • 24.Rodrigues MCO, Sanberg PR, Cruz LE, Garbuzova-Davis S. The innate and adaptive immunological aspects in neurodegenerative diseases. J Neuroimmunol. 2014;269:1–8. doi: 10.1016/j.jneuroim.2013.09.020. [DOI] [PubMed] [Google Scholar]
  • 25.McCombe PA, Henderson RD. The role of immune and inflammatory mechanisms in ALS. Curr Mol Med. 2011;11:246–54. doi: 10.2174/156652411795243450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.D’Amico E, Factor-Litvak P, Santella RM, Mitsumoto H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic Biol Med. 2013;65:509–27. doi: 10.1016/j.freeradbiomed.2013.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Papadimitriou D, Le Verche V, Jacquier A, et al. Inflammation in ALS and SMA: sorting out the good from the evil. Neurobiol Dis. 2010;37:493–502. doi: 10.1016/j.nbd.2009.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhao W, Beers DR, Appel SH. Immune-mediated mechanisms in the pathoprogression of amyotrophic lateral sclerosis. J Neuroimmune Pharmacol. 2013;8:888–99. doi: 10.1007/s11481-013-9489-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Calvo AC, Manzano R, Mendonça DMF, et al. Amyotrophic lateral sclerosis: a focus on disease progression. Biomed Res Int. 2014;2014:925101. doi: 10.1155/2014/925101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30*.Aggarwal S, Cudkowicz M. ALS drug development: reflections from the past and a way forward. Neurotherapeutics. 2008;5:516–27. doi: 10.1016/j.nurt.2008.08.002. Comprehensive review of recent clinical drug trials for ALS (most failures) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31*.Cudkowicz ME, Katz J, Moore DH, et al. Toward more efficient clinical trials for amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11:259–65. doi: 10.3109/17482960903358865. Comprehensive review identifying methodological deficiencies in pre-clinical studies and clinical drug trials for ALS. [DOI] [PubMed] [Google Scholar]
  • 32.Kalra S, Genge A, Arnold DL. A prospective, randomized, placebo-controlled evaluation of corticoneuronal response to intrathecal BDNF therapy in ALS using magnetic resonance spectroscopy: feasibility and results. Amyotroph Lateral Scler Other Motor Neuron Disord. 2003;4:22–6. doi: 10.1080/14660820310006689. [DOI] [PubMed] [Google Scholar]
  • 33.Beck M, Flachenecker P, Magnus T, et al. Autonomic dysfunction in ALS: a preliminary study on the effects of intrathecal BDNF. Amyotroph Lateral Scler Other Motor Neuron Disord. 2005;6:100–3. doi: 10.1080/14660820510028412. [DOI] [PubMed] [Google Scholar]
  • 34.Miller RG, Petajan JH, Bryan WW, et al. A placebo-controlled trial of recombinant human ciliary neurotrophic (rhCNTF) factor in amyotrophic lateral sclerosis. rhCNTF ALS Study Group. Ann Neurol. 1996;39:256–60. doi: 10.1002/ana.410390215. [DOI] [PubMed] [Google Scholar]
  • 35.Giménez y Ribotta M, Revah F, Pradier L, et al. Prevention of motoneuron death by adenovirus-mediated neurotrophic factors. J Neurosci Res. 1997;48:281–5. doi: 10.1002/(sici)1097-4547(19970501)48:3<281::aid-jnr11>3.3.co;2-i. [DOI] [PubMed] [Google Scholar]
  • 36.Ikeda K, Klinkosz B, Greene T, et al. Effects of brain-derived neurotrophic factor on motor dysfunction in wobbler mouse motor neuron disease. Ann Neurol. 1995;37:505–11. doi: 10.1002/ana.410370413. [DOI] [PubMed] [Google Scholar]
  • 37.Sendtner M, Kreutzberg GW, Thoenen H. Ciliary neurotrophic factor prevents the degeneration of motor neurons after axotomy. Nature. 1990;345:440–441. doi: 10.1038/345440a0. [DOI] [PubMed] [Google Scholar]
  • 38.Sendtner M, Schmalbruch H, Stöckli KA, et al. Ciliary neurotrophic factor prevents degeneration of motor neurons in mouse mutant progressive motor neuronopathy. Nature. 1992;358:502–4. doi: 10.1038/358502a0. [DOI] [PubMed] [Google Scholar]
  • 39.Kaspar BK, Lladó J, Sherkat N, et al. Retrograde viral delivery of IGF-1 prolongs survival in a mouse ALS model. Science. 2003;301:839–42. doi: 10.1126/science.1086137. [DOI] [PubMed] [Google Scholar]
  • 40.Nagano I, Ilieva H, Shiote M, et al. Therapeutic benefit of intrathecal injection of insulin-like growth factor-1 in a mouse model of Amyotrophic Lateral Sclerosis. J Neurol Sci. 2005;235:61–8. doi: 10.1016/j.jns.2005.04.011. [DOI] [PubMed] [Google Scholar]
  • 41.Lepore AC, Haenggeli C, Gasmi M, et al. Intraparenchymal spinal cord delivery of adeno-associated virus IGF-1 is protective in the SOD1G93A model of ALS. Brain Res. 2007;1185:256–65. doi: 10.1016/j.brainres.2007.09.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dodge JC, Haidet AM, Yang W, et al. Delivery of AAV-IGF-1 to the CNS extends survival in ALS mice through modification of aberrant glial cell activity. Mol Ther. 2008;16:1056–64. doi: 10.1038/mt.2008.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Messi ML, Clark HM, Prevette DM, et al. The lack of effect of specific overexpression of IGF-1 in the central nervous system or skeletal muscle on pathophysiology in the G93A SOD-1 mouse model of ALS. Exp Neurol. 2007;207:52–63. doi: 10.1016/j.expneurol.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Franz CK, Federici T, Yang J, et al. Intraspinal cord delivery of IGF-I mediated by adeno-associated virus 2 is neuroprotective in a rat model of familial ALS. Neurobiol Dis. 2009;33:473–81. doi: 10.1016/j.nbd.2008.12.003. [DOI] [PubMed] [Google Scholar]
  • 45.Nagano I, Shiote M, Murakami T, et al. Beneficial effects of intrathecal IGF-1 administration in patients with amyotrophic lateral sclerosis. Neurol Res. 2005;27:768–72. doi: 10.1179/016164105X39860. [DOI] [PubMed] [Google Scholar]
  • 46.Sorenson EJ, Windbank AJ, Mandrekar JN, et al. Subcutaneous IGF-1 is not beneficial in 2-year ALS trial. Neurology. 2008;71:1770–5. doi: 10.1212/01.wnl.0000335970.78664.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sakowski SA, Schuyler AD, Feldman EL. Insulin-like growth factor-I for the treatment of amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2009;10:63–73. doi: 10.1080/17482960802160370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48**.Henriques A, Pitzer C, Schneider A. Neurotrophic growth factors for the treatment of amyotrophic lateral sclerosis: where do we stand? Front Neurosci. 2010;4:32. doi: 10.3389/fnins.2010.00032. Comprehensive review of neurotrophic growth factors as potential drug therapies for ALS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yacila G, Sari Y. Potential therapeutic drugs and methods for the treatment of amyotrophic lateral sclerosis. Curr Med Chem. 2014;21:3583–93. doi: 10.2174/0929867321666140601162710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kriz J, Nguyen MD, Julien J-P. Minocycline slows disease progression in a mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2002;10:268–78. doi: 10.1006/nbdi.2002.0487. [DOI] [PubMed] [Google Scholar]
  • 51.Zhang W, Narayanan M, Friedlander RM. Additive neuroprotective effects of minocycline with creatine in a mouse model of ALS. Ann Neurol. 2003;53:267–70. doi: 10.1002/ana.10476. [DOI] [PubMed] [Google Scholar]
  • 52.Van Den Bosch L, Tilkin P, Lemmens G, Robberecht W. Minocycline delays disease onset and mortality in a transgenic model of ALS. Neuroreport. 2002;13:1067–70. doi: 10.1097/00001756-200206120-00018. [DOI] [PubMed] [Google Scholar]
  • 53.Gordon PH, Moore DH, Miller RG, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet Neurol. 2007;6:1045–53. doi: 10.1016/S1474-4422(07)70270-3. [DOI] [PubMed] [Google Scholar]
  • 54.Gámez J. Minocycline for the treatment of amyotrophic lateral sclerosis: neuroprotector or neurotoxin? Reflections on another failure of translational medicine. Neurologia. 2008;23:484–93. [PubMed] [Google Scholar]
  • 55.Keller AF, Gravel M, Kriz J. Treatment with minocycline after disease onset alters astrocyte reactivity and increases microgliosis in SOD1 mutant mice. Exp Neurol. 2011;228:69–79. doi: 10.1016/j.expneurol.2010.12.010. [DOI] [PubMed] [Google Scholar]
  • 56.Groeneveld GJ, Veldink JH, van der Tweel I, et al. A randomized sequential trial of creatine in amyotrophic lateral sclerosis. Ann Neurol. 2003;53:437–45. doi: 10.1002/ana.10554. [DOI] [PubMed] [Google Scholar]
  • 57.Shefner JM, Cudkowicz ME, Schoenfeld D, et al. A clinical trial of creatine in ALS. Neurology. 2004;63:1656–61. doi: 10.1212/01.wnl.0000142992.81995.f0. [DOI] [PubMed] [Google Scholar]
  • 58.Rosenfeld J, King RM, Jackson CE, et al. Creatine monohydrate in ALS: effects on strength, fatigue, respiratory status and ALSFRS. Amyotroph Lateral Scler. 2008;9:266–72. doi: 10.1080/17482960802028890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Klivenyi P, Ferrante RJ, Matthews RT, et al. Neuroprotective effects of creatine in a transgenic animal model of amyotrophic lateral sclerosis. Nat Med. 1999;5:347–50. doi: 10.1038/6568. [DOI] [PubMed] [Google Scholar]
  • 60.Drachman DB, Frank K, Dykes-Hoberg M, et al. Cyclooxygenase 2 inhibition protects motor neurons and prolongs survival in a transgenic mouse model of ALS. Ann Neurol. 2002;52:771–778. doi: 10.1002/ana.10374. [DOI] [PubMed] [Google Scholar]
  • 61.Cudkowicz ME, Shefner JM, Schoenfeld DA, et al. Trial of celecoxib in amyotrophic lateral sclerosis. Ann Neurol. 2006;60:22–31. doi: 10.1002/ana.20903. [DOI] [PubMed] [Google Scholar]
  • 62.Rothstein JD, Patel S, Regan MR, et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature. 2005;433:73–7. doi: 10.1038/nature03180. [DOI] [PubMed] [Google Scholar]
  • 63.Cudkowicz ME, Titus S, Kearney M, et al. Safety and efficacy of ceftriaxone for amyotrophic lateral sclerosis: a multi-stage, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2014;13:1083–91. doi: 10.1016/S1474-4422(14)70222-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wang R, Zhang D. Memantine prolongs survival in an amyotrophic lateral sclerosis mouse model. Eur J Neurosci. 2005;22:2376–80. doi: 10.1111/j.1460-9568.2005.04431.x. [DOI] [PubMed] [Google Scholar]
  • 65.Joo I-S, Hwang D-H, Seok J-I, et al. Oral administration of memantine prolongs survival in a transgenic mouse model of amyotrophic lateral sclerosis. J Clin Neurol. 2007;3:181–6. doi: 10.3988/jcn.2007.3.4.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.de Carvalho M, Pinto S, Costa J, et al. A randomized, placebo-controlled trial of memantine for functional disability in amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2010;11:456–60. doi: 10.3109/17482968.2010.498521. [DOI] [PubMed] [Google Scholar]
  • 67.Levine TD, Bowser R, Hank N, Saperstein D. A pilot trial of memantine and riluzole in ALS: correlation to CSF biomarkers. Amyotroph Lateral Scler. 2010;11:514–9. doi: 10.3109/17482968.2010.513052. [DOI] [PubMed] [Google Scholar]
  • 68.Gurney ME, Cutting FB, Zhai P, et al. Benefit of vitamin E, riluzole, and gabapentin in a transgenic model of familial amyotrophic lateral sclerosis. Ann Neurol. 1996;39:147–57. doi: 10.1002/ana.410390203. [DOI] [PubMed] [Google Scholar]
  • 69.Gurney ME, Fleck TJ, Himes CS, Hall ED. Riluzole preserves motor function in a transgenic model of familial amyotrophic lateral sclerosis. Neurology. 1998;50:62–66. doi: 10.1212/wnl.50.1.62. [DOI] [PubMed] [Google Scholar]
  • 70.Riviere M, Meininger V, Zeisser P, Munsat T. An analysis of extended survival in patients with amyotrophic lateral sclerosis treated with riluzole. Arch Neurol. 1998;55:526–8. doi: 10.1001/archneur.55.4.526. [DOI] [PubMed] [Google Scholar]
  • 71.Dunlop J, Beal McIlvain H, She Y, Howland DS. Impaired spinal cord glutamate transport capacity and reduced sensitivity to riluzole in a transgenic superoxide dismutase mutant rat model of amyotrophic lateral sclerosis. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience. 2003;23:1688–1696. doi: 10.1523/JNEUROSCI.23-05-01688.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Traynor BJ, Alexander M, Corr B, et al. Effect of a multidisciplinary amyotrophic lateral sclerosis (ALS) clinic on ALS survival: a population based study, 1996–2000. J Neurol Neurosurg Psychiatr. 2003;74:1258–61. doi: 10.1136/jnnp.74.9.1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zoccolella S, Beghi E, Palagano G, et al. Riluzole and amyotrophic lateral sclerosis survival: a population-based study in southern Italy. Eur J Neurol. 2007;14:262–8. doi: 10.1111/j.1468-1331.2006.01575.x. [DOI] [PubMed] [Google Scholar]
  • 74.Shefner JM, Wolff AA, Meng L, et al. A randomized, placebo-controlled, double-blind phase IIb trial evaluating the safety and efficacy of tirasemtiv in patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener. 2016:1–10. doi: 10.3109/21678421.2016.1148169. [DOI] [PubMed] [Google Scholar]
  • 75.Hwee DT, Kennedy A, Ryans J, et al. Fast skeletal muscle troponin activator tirasemtiv increases muscle function and performance in the B6SJL-SOD1G93A ALS mouse model. PLoS ONE. 2014;9:e96921. doi: 10.1371/journal.pone.0096921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ludolph AC, Bendotti C, Blaugrund E, et al. Guidelines for the preclinical in vivo evaluation of pharmacological active drugs for ALS/MND: report on the 142nd ENMC international workshop. Amyotroph Lateral Scler. 2007;8:217–23. doi: 10.1080/17482960701292837. [DOI] [PubMed] [Google Scholar]
  • 77**.Scott S, Kranz JE, Cole J, et al. Design, power, and interpretation of studies in the standard murine model of ALS. Amyotroph Lateral Scler. 2008;9:4–15. doi: 10.1080/17482960701856300. Comprehensive review on proper design and statistical methodologies for use of G93A SOD1 mouse model for ALS. [DOI] [PubMed] [Google Scholar]
  • 78.Vincent AM, Sakowski SA, Schuyler A, Feldman EL. Strategic approaches to developing drug treatments for ALS. Drug Discov Today. 2008;13:67–72. doi: 10.1016/j.drudis.2007.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zoccolella S, Santamato A, Lamberti P. Current and emerging treatments for amyotrophic lateral sclerosis. Neuropsychiatr Dis Treat. 2009;5:577–95. doi: 10.2147/ndt.s7788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bedlack RS, Traynor BJ, Cudkowicz ME. Emerging disease-modifying therapies for the treatment of motor neuron disease/amyotropic lateral sclerosis. Expert Opin Emerg Drugs. 2007;12:229–52. doi: 10.1517/14728214.12.2.229. [DOI] [PubMed] [Google Scholar]
  • 81.Benatar M. Lost in translation: treatment trials in the SOD1 mouse and in human ALS. Neurobiol Dis. 2007;26:1–13. doi: 10.1016/j.nbd.2006.12.015. [DOI] [PubMed] [Google Scholar]
  • 82*.Ludolph AC, Jesse S. Evidence-based drug treatment in amyotrophic lateral sclerosis and upcoming clinical trials. Ther Adv Neurol Disord. 2009;2:319–26. doi: 10.1177/1756285609336399. Comprehensive review on therapeutic strategies for ALS clinical trials. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Carter GT, Krivickas LS, Weydt P, et al. Drug therapy for amyotrophic lateral sclerosis: Where are we now? IDrugs. 2003;6:147–53. [PubMed] [Google Scholar]
  • 84.Siciliano G, Carlesi C, Pasquali L, et al. Clinical trials for neuroprotection in ALS. CNS Neurol Disord Drug Targets. 2010;9:305–13. doi: 10.2174/187152710791292648. [DOI] [PubMed] [Google Scholar]
  • 85.Wainger BJ, Kiskinis E, Mellin C, et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 2014;7:1–11. doi: 10.1016/j.celrep.2014.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Herrando-Grabulosa M, Mulet R, Pujol A, et al. Novel neuroprotective multicomponent therapy for amyotrophic lateral sclerosis designed by networked systems. PLoS ONE. 2016;11:e0147626. doi: 10.1371/journal.pone.0147626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ramesh T, Lyon AN, Pineda RH, et al. A genetic model of amyotrophic lateral sclerosis in zebrafish displays phenotypic hallmarks of motoneuron disease. Dis Model Mech. 2010;3:652–62. doi: 10.1242/dmm.005538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ciura S, Lattante S, Le Ber I, et al. Loss of function of C9orf72 causes motor deficits in a zebrafish model of amyotrophic lateral sclerosis. Ann Neurol. 2013;74:180–7. doi: 10.1002/ana.23946. [DOI] [PubMed] [Google Scholar]
  • 89.Patten SA, Parker JA, Wen X-Y, Drapeau P. Simple animal models for amyotrophic lateral sclerosis drug discovery. Expert Opin Drug Discov. 2016:1–8. doi: 10.1080/17460441.2016.1196183. [DOI] [PubMed] [Google Scholar]
  • 90.Babin PJ, Goizet C, Raldúa D. Zebrafish models of human motor neuron diseases: advantages and limitations. Prog Neurobiol. 2014;118:36–58. doi: 10.1016/j.pneurobio.2014.03.001. [DOI] [PubMed] [Google Scholar]
  • 91.Jeong J-Y, Kwon H-B, Ahn J-C, et al. Functional and developmental analysis of the blood-brain barrier in zebrafish. Brain Res Bull. 2008;75:619–28. doi: 10.1016/j.brainresbull.2007.10.043. [DOI] [PubMed] [Google Scholar]
  • 92.Eliceiri BP, Gonzalez AM, Baird A. Zebrafish model of the blood-brain barrier: morphological and permeability studies. Methods Mol Biol. 2011;686:371–8. doi: 10.1007/978-1-60761-938-3_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Fleming A, Diekmann H, Goldsmith P. Functional characterisation of the maturation of the blood-brain barrier in larval zebrafish. PLoS ONE. 2013;8:e77548. doi: 10.1371/journal.pone.0077548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kastin AJ, Akerstrom V, Pan W. Glial cell line-derived neurotrophic factor does not enter normal mouse brain. Neurosci Lett. 2003;340:239–41. doi: 10.1016/s0304-3940(03)00007-7. [DOI] [PubMed] [Google Scholar]
  • 95.Dobrogowska DH, Lossinsky AS, Tarnawski M, Vorbrodt AW. Increased blood-brain barrier permeability and endothelial abnormalities induced by vascular endothelial growth factor. J Neurocytol. 1998;27:163–73. doi: 10.1023/a:1006907608230. [DOI] [PubMed] [Google Scholar]
  • 96.Yang J-P, Liu H-J, Cheng S-M, et al. Direct transport of VEGF from the nasal cavity to brain. Neurosci Lett. 2009;449:108–11. doi: 10.1016/j.neulet.2008.10.090. [DOI] [PubMed] [Google Scholar]
  • 97.Pardridge WM. Blood-brain barrier biology and methodology. J Neurovirol. 1999;5:556–569. doi: 10.3109/13550289909021285. [DOI] [PubMed] [Google Scholar]
  • 98.Vorbrodt AW, Dobrogowska DH. Molecular anatomy of intercellular junctions in brain endothelial and epithelial barriers: electron microscopist’s view. Brain Res Rev. 2003;42:221–42. doi: 10.1016/s0165-0173(03)00177-2. [DOI] [PubMed] [Google Scholar]
  • 99.Ballabh P, Braun A, Nedergaard M. The blood-brain barrier: an overview: structure, regulation, and clinical implications. Neurobiol Dis. 2004;16:1–13. doi: 10.1016/j.nbd.2003.12.016. [DOI] [PubMed] [Google Scholar]
  • 100.Engelhardt B. Regulation of immune cell entry into the central nervous system. Results Probl Cell Differ. 2006;43:259–280. doi: 10.1007/400_020. [DOI] [PubMed] [Google Scholar]
  • 101.Abbott NJ, Patabendige AAK, Dolman DEM, et al. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010;37:13–25. doi: 10.1016/j.nbd.2009.07.030. [DOI] [PubMed] [Google Scholar]
  • 102.Hirase T, Staddon JM, Saitou M, et al. Occludin as a possible determinant of tight junction permeability in endothelial cells. J Cell Sci. 1997;110(Pt 14):1603–1613. doi: 10.1242/jcs.110.14.1603. [DOI] [PubMed] [Google Scholar]
  • 103.Furuse M, Hirase T, Itoh M, et al. Occludin: a novel integral membrane protein localizing at tight junctions. J Cell Biol. 1993;123:1777–1788. doi: 10.1083/jcb.123.6.1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Furuse M, Fujita K, Hiiragi T, et al. Claudin-1 and -2: novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J Cell Biol. 1998;141:1539–1550. doi: 10.1083/jcb.141.7.1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tsukita S, Furuse M. Overcoming barriers in the study of tight junction functions: from occludin to claudin. Genes Cells. 1998;3:569–573. doi: 10.1046/j.1365-2443.1998.00212.x. [DOI] [PubMed] [Google Scholar]
  • 106.Rutka JT, Apodaca G, Stern R, Rosenblum M. The extracellular matrix of the central and peripheral nervous systems: structure and function. J Neurosurg. 1988;69:155–170. doi: 10.3171/jns.1988.69.2.0155. [DOI] [PubMed] [Google Scholar]
  • 107.Dermietzel R, Krause D. Molecular anatomy of the blood-brain barrier as defined by immunocytochemistry. Int Rev Cytol. 1991;127:57–109. doi: 10.1016/s0074-7696(08)60692-0. [DOI] [PubMed] [Google Scholar]
  • 108.Prat A, Biernacki K, Wosik K, Antel JP. Glial cell influence on the human blood-brain barrier. Glia. 2001;36:145–55. doi: 10.1002/glia.1104. [DOI] [PubMed] [Google Scholar]
  • 109.Abbott NJ. Astrocyte-endothelial interactions and blood-brain barrier permeability. J Anat. 2002;200:629–38. doi: 10.1046/j.1469-7580.2002.00064.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006;7:41–53. doi: 10.1038/nrn1824. [DOI] [PubMed] [Google Scholar]
  • 111.Leonardi A, Abbruzzese G, Arata L, et al. Cerebrospinal fluid (CSF) findings in amyotrophic lateral sclerosis. J Neurol. 1984;231:75–78. doi: 10.1007/BF00313720. [DOI] [PubMed] [Google Scholar]
  • 112.Apostolski S, Nikolić J, Bugarski-Prokopljević C, et al. Serum and CSF immunological findings in ALS. Acta Neurol Scand. 1991;83:96–98. doi: 10.1111/j.1600-0404.1991.tb04656.x. [DOI] [PubMed] [Google Scholar]
  • 113.Bilic E, Rudan I, Kusec V, et al. Comparison of the growth hormone, IGF-1 and insulin in cerebrospinal fluid and serum between patients with motor neuron disease and healthy controls. Eur J Neurol. 2006;13:1340–1345. doi: 10.1111/j.1468-1331.2006.01503.x. [DOI] [PubMed] [Google Scholar]
  • 114.Pirttilä T, Vanhatalo S, Turpeinen U, Riikonen R. Cerebrospinal fluid insulin-like growth factor-1, insulin growth factor binding protein-2 or nitric oxide are not increased in MS or ALS. Acta Neurol Scand. 2004;109:337–341. doi: 10.1111/j.1600-0404.2004.00223.x. [DOI] [PubMed] [Google Scholar]
  • 115.Garbuzova-Davis S, Haller E, Saporta S, et al. Ultrastructure of blood-brain barrier and blood-spinal cord barrier in SOD1 mice modeling ALS. Brain Res. 2007;1157:126–37. doi: 10.1016/j.brainres.2007.04.044. [DOI] [PubMed] [Google Scholar]
  • 116.Garbuzova-Davis S, Saporta S, Haller E, et al. Evidence of compromised blood-spinal cord barrier in early and late symptomatic SOD1 mice modeling ALS. PLoS ONE. 2007;2:e1205. doi: 10.1371/journal.pone.0001205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117**.Garbuzova-Davis S, Hernandez-Ontiveros DG, Rodrigues MCO, et al. Impaired blood-brain/spinal cord barrier in ALS patients. Brain Res. 2012;1469:114–28. doi: 10.1016/j.brainres.2012.05.056. Research article determining pervasive B-CNS-B damage in sporadic ALS patients. [DOI] [PubMed] [Google Scholar]
  • 118.Nicaise C, Mitrecic D, Demetter P, et al. Impaired blood-brain and blood-spinal cord barriers in mutant SOD1-linked ALS rat. Brain Res. 2009;1301:152–62. doi: 10.1016/j.brainres.2009.09.018. [DOI] [PubMed] [Google Scholar]
  • 119.Zhong Z, Deane R, Ali Z, et al. ALS-causing SOD1 mutants generate vascular changes prior to motor neuron degeneration. Nat Neurosci. 2008;11:420–422. doi: 10.1038/nn2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Miyazaki K, Ohta Y, Nagai M, et al. Disruption of neurovascular unit prior to motor neuron degeneration in amyotrophic lateral sclerosis. J Neurosci Res. 2011;89:718–728. doi: 10.1002/jnr.22594. [DOI] [PubMed] [Google Scholar]
  • 121.Henkel JS, Beers DR, Wen S, et al. Decreased mRNA expression of tight junction proteins in lumbar spinal cords of patients with ALS. Neurology. 2009;72:1614–1616. doi: 10.1212/WNL.0b013e3181a41228. [DOI] [PubMed] [Google Scholar]
  • 122.Winkler EA, Sengillo JD, Sullivan JS, et al. Blood-spinal cord barrier breakdown and pericyte reductions in amyotrophic lateral sclerosis. Acta Neuropathol. 2013;125:111–20. doi: 10.1007/s00401-012-1039-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123*.Garbuzova-Davis S, Rodrigues MCO, Hernandez-Ontiveros DG, et al. Amyotrophic lateral sclerosis: a neurovascular disease. Brain Res. 2011;1398:113–25. doi: 10.1016/j.brainres.2011.04.049. Comprehensive review detailing accumulated evidence identifying ALS as a neurovascular disease. [DOI] [PubMed] [Google Scholar]
  • 124.Garbuzova-Davis S, Saporta S, Sanberg PR. Implications of blood-brain barrier disruption in ALS. Amyotroph Lateral Scler. 2008;9:375–376. doi: 10.1080/17482960802160990. [DOI] [PubMed] [Google Scholar]
  • 125**.Garbuzova-Davis S, Sanberg PR. Blood-CNS barrier impairment in ALS patients versus an animal model. Front Cell Neurosci. 2014;8:21. doi: 10.3389/fncel.2014.00021. Comprehensive review discussing similarities and differences in B-CNS-B alterations between animal model and ALS patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rule RR, Schuff N, Miller RG, Weiner MW. Gray matter perfusion correlates with disease severity in ALS. Neurology. 2010;74:821–7. doi: 10.1212/WNL.0b013e3181d3e2dd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Vlasenko A, Garbuzova-Davis S. The role of matrix metalloproteinases in neurovascular unit integrity in amyotrophic lateral sclerosis. Neurol Res Ther Open Access. 2015;2:112–29. [Google Scholar]
  • 128.Zwolinski BJ, Eyring H, Reese CE. Diffusion and membrane permeability. J Phys Chem. 1949;53:1426–53. [Google Scholar]
  • 129.Moreau C, Brunaud-Danel V, Dallongeville J, et al. Modifying effect of arterial hypertension on amyotrophic lateral sclerosis. Amyotroph Lateral Scler. 2012;13:194–201. doi: 10.3109/17482968.2011.610110. [DOI] [PubMed] [Google Scholar]
  • 130.Lin F-C, Tsai C-P, Kuang-Wu Lee J, et al. Angiotensin-converting enzyme inhibitors and amyotrophic lateral sclerosis risk: a total population-based case-control study. JAMA Neurol. 2015;72:40–8. doi: 10.1001/jamaneurol.2014.3367. [DOI] [PubMed] [Google Scholar]
  • 131.Burke M. Scientific American. 2014. Why Alzheimer’s drugs keep failing. [Google Scholar]
  • 132*.Gauthier S, Albert M, Fox N, et al. Why has therapy development for dementia failed in the last two decades? Alzheimers Dement. 2016;12:60–4. doi: 10.1016/j.jalz.2015.12.003. Comprehensive review discussing clinical drug trial failures in AD. [DOI] [PubMed] [Google Scholar]
  • 133.Claudio L. Ultrastructural features of the blood-brain barrier in biopsy tissue from Alzheimer’s disease patients. Acta Neuropathol. 1996;91:6–14. doi: 10.1007/s004010050386. [DOI] [PubMed] [Google Scholar]
  • 134.Kalaria RN, Pax AB. Increased collagen content of cerebral microvessels in Alzheimer’s disease. Brain Res. 1995;705:349–52. doi: 10.1016/0006-8993(95)01250-8. [DOI] [PubMed] [Google Scholar]
  • 135.Bourasset F, Ouellet M, Tremblay C, et al. Reduction of the cerebrovascular volume in a transgenic mouse model of Alzheimer’s disease. Neuropharmacology. 2009;56:808–13. doi: 10.1016/j.neuropharm.2009.01.006. [DOI] [PubMed] [Google Scholar]
  • 136.González-Marrero I, Giménez-Llort L, Johanson CE, et al. Choroid plexus dysfunction impairs beta-amyloid clearance in a triple transgenic mouse model of Alzheimer’s disease. Front Cell Neurosci. 2015;9:17. doi: 10.3389/fncel.2015.00017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Mehta DC, Short JL, Nicolazzo JA. Altered brain uptake of therapeutics in a triple transgenic mouse model of Alzheimer’s disease. Pharm Res. 2013;30:2868–79. doi: 10.1007/s11095-013-1116-2. [DOI] [PubMed] [Google Scholar]
  • 138.Trudeau K, Roy S, Guo W, et al. Fenofibric acid reduces fibronectin and collagen type IV overexpression in human retinal pigment epithelial cells grown in conditions mimicking the diabetic milieu: Functional implications in retinal permeability. Invest Ophthalmol Vis Sci. 2011;52:6348–54. doi: 10.1167/iovs.11-7282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Liebetrau M, Burggraf D, Büscher C, et al. Ramipril prevents extracellular matrix accumulation in cerebral microvessels. Neurol Res. 2005;27:477–82. doi: 10.1179/016164105X49256. [DOI] [PubMed] [Google Scholar]
  • 140.Stichel CC, Hermanns S, Luhmann HJ, et al. Inhibition of collagen IV deposition promotes regeneration of injured CNS axons. Eur J Neurosci. 1999;11:632–46. doi: 10.1046/j.1460-9568.1999.00466.x. [DOI] [PubMed] [Google Scholar]

RESOURCES