Abstract
Amyotrophic lateral sclerosis (ALS) is a slowly progressing neurodegenerative disease that affects motor neurons of the nervous system. Despite the identification of many potential therapeutics targeting pathogenic mechanisms in in vitro models, there has been limited progress in translating them into a successful pharmacotherapy in the animal model of ALS. Further, efforts to translate any promising results from preclinical trials to effective pharmacotherapies for patients have been unsuccessful, with the exception of riluzole, the only FDA-approved medication, which only modestly extends survival both in the animal model and in patients. Thus, it is essential to reconsider the strategies for developing ALS pharmacotherapies. Growing evidence suggests that problems identifying highly effective ALS treatments may result from an underestimated issue of drug bioavailability and disease-driven pharmacoresistance, mediated by the ATP-binding cassette (ABC) drug efflux transporters. ABC transporters are predominately localized to the lumen of endothelial cells of the blood-brain and blood-spinal cord barriers (BBB, BSCB) where they limit the entry into the central nervous system (CNS) of a wide range of neurotoxicants and xenobiotics, but also therapeutics. In ALS, expression and function of ABC transporters is increased at the BBB/BSCB and their expression has been detected on neurons and glia in the CNS parenchyma, which may further reduce therapeutic action in target cells. Understanding and accounting for the contribution of these transporters to ALS pharmacoresistance could both improve the modest effects of riluzole and set in motion a re-evaluation of previous ALS drug disappointments. In addition, identifying pathogenic mechanisms regulating ABC transporter expression and function in ALS may lead to the development of new therapeutic strategies. It is likely that novel pharmacological approaches require counteracting pharmacoresistance to improve therapeutic efficacy.
Keywords: Amyotrophic Lateral Sclerosis, Drug Efflux Transporters, Blood-brain barrier, Pharmacoresistance, Neurodegeneration
Amyotrophic Lateral Sclerosis (ALS)
Amyotrophic Lateral Sclerosis (ALS) is a slowly progressing, fatal neurodegenerative disease that selectively affects both upper and lower motor neurons of the nervous system. One of the biggest challenges in the study of ALS is its complex etiology, which combines both environmental and genetic factors (Siddique and Ajroud-Driss, 2011). For ~90% of ALS cases, there is no family history and no identified cause (sporadic ALS). In the remaining 10% of cases (familial ALS), the disease is caused by genetic factors with 2/3 of disease-causative genes identified to date (Al-Chalabi et al., 2012). Currently, there is only one FDA-approved drug, riluzole, available to treat ALS, which modestly extends patient survival by 2–4 months (Miller et al., 2012). The first genetic linkage identified to contribute to ALS was Cu2+/Zn2+ superoxide dismutase 1 (SOD1) (Rosen et al., 1993). Several other genes have recently been identified as contributors to familial ALS, including the genes encoding for DNA and RNA-binding proteins TDP-43 and FUS/TLS, TARDP and FUS, as well as UBQLN2, VCP, and C9ORF72 (DeJesus-Hernandez et al., 2011; Deng et al., 2011; Johnson et al., 2010; Kwiatkowski et al., 2009; Neumann et al., 2006; Renton et al., 2011; Vance et al., 2009).
Historically, research on the mechanisms and pathology of ALS has utilized transgenic mouse models, which overexpress the human SOD1 gene carrying one of three mutations (SOD1G93A, SOD1G37R, and SOD1G85R). These animal models provided insight on the toxicity of the mutated gene toward neuronal and non-neuronal cell types, which has led to the development of widespread theories of the underlying disease pathogenesis. Several lines of investigations now indicate that linking ALS to a single mechanism is unlikely as multiple pathways of toxicity were found to contribute to disease (Al-Chalabi et al., 2012; Ferraiuolo et al., 2011; Garbuzova-Davis et al., 2011).
Pharmacological Interventions in ALS
The entirety of preclinical ALS research is currently focused on the transgenic mouse lines expressing human SOD1 mutations, which mimic many aspects of human ALS pathology and for which extended survival is one of the main predictors of preclinical success (Ludolph et al., 2007). Behavioral and functional assessments also measure disease onset and progression (Knippenberg et al., 2010). Several compounds have been identified that provide some degree of improvement in these functional assessment tests and in survival, but none thus far has proved to be a substantial treatment option when translated in patients.
There are multiple issues that could account for this discrepancy, including the study design of preclinical trials, the lack of additional animal models available for research, and insufficient insight into pathological causes. Preclinical drug trials typically begin pharmacological intervention prior to the onset of symptoms (Benatar, 2007). This allows the researchers to examine differences in onset and progression of the disease, but hinders the practicality of translating the treatment to patients. On average, ALS patients are diagnosed 9 months after the onset of the first symptoms, at which point, treatment can begin. The disparity between initial treatment in preclinical animal studies and realistic treatment administration in patients appears to diminish efficacy in the clinic. Furthermore, studying the mutant SOD1 transgenic mouse model has identified multiple cell types and molecular mechanisms that are affected, hence single treatments that target one pathway at a time may not be enough. Recently, a number of investigators have begun to test combination therapies, which can potentially enhance the effect of single pharmacological agents (Kong et al., 2012).
While the mutant SOD1 mice mimic ALS to some degree, multiple problems prevent accurate prediction of clinical trial success. One of the major concerns is the lack of standardization in mutant SOD1 mouse strains. Depending upon the route of administration, dose, type and gender of mutant mouse, and onset of treatment, the effects of survival can be markedly different across studies and the interpretation and extrapolation of the actual benefits of a potential ALS-treating drug becomes difficult, if not unpredictable. As evidenced by the multitude of preclinical trials, a modest extension of lifespan in the mouse model of ALS has failed in predicting success in clinical trials (Table 1 and Table 2). Recently, a workshop led by Dr. Storey Landis, Director of the National Institute for Neurological Disorders and Stroke (NINDS), defined a set of guidelines designed to mitigate such problems. They concluded that in the preclinical research community, alleviating poor reporting and improving experimental design and reporting practices should lead to greater transparency and reproducibility (Landis et al., 2012).
Table 1.
Compound | Study ID | Study Design | Dosage | Route | Primary Outcome Measure | Prospective End Date | Location |
---|---|---|---|---|---|---|---|
VEGF165 | NCT01384162 | Phase 1/2 single arm | Not specified | ICV | Safety and tolerability | December 2013 | Belgium |
Rasagiline | NCT01232738 | Phase 2 RCT | 1mg/day | P.O. | Difference in rate of decline in function, ALSFRS-R | May 2013 | USA & Canada |
NP001 | NCT01281631 | Phase 2 RCT | Low/High | I.V. | Measures of clinical function | October 2012 | USA |
Creatine and Tamoxifen | NCT01257581 | Phase 2 RCT | 30g/40mg/day | P.O. | Decline in ALSFRS-R | December 2012 | USA |
Mecobalamin | NCT00444613 | Phase 2/3 RCT | 25mg or 50mg twice weekly | I.M. | Survival rate, functional rating scale | March 2014 | Japan |
Mecobalamin | NCT00445172 | Phase 2/3 NRSA | Not specified | I.M. | Safety and tolerability | January 2014 | Japan |
Tirasemtiv (CK-2017357) | NCT01709149 | Phase 2b RCT | 250–500mg twice daily | P.O. | Change from baseline to average of ALSFRS-R | October 2013 | USA |
Anakinra and Riluzole | NCT01277315 | Phase 2 | 100mg | SubQ | Safety and tolerability | June 2012 | Germany |
Arimoclomol | NCT00706147 | Phase 2/3 RCT | 200mg three times daily | P.O. | Decline of ALSFRS-R | December 2012 | USA |
MCI-186 | NCT01492686 | Phase 3 RCT | 60mg/day | I.V. | Decline of ALSFRS-R | March 2015 | Japan |
Table 2.
Compound | Mechanism of Action | Preclinical Impact | Preclinical Survival Effect | Clinical Trials | Reference |
---|---|---|---|---|---|
Arimoclomol | Protein Aggregation Inibition | Prolonged lifespan Increased motor neuron survival | ~9–22% | Ongoing | (Kalmar et al., 2008) (Kieran et al., 2004) |
AEOL-10150 | Antioxidant and inhibits lipid peroxidation | Preserved SC motor neurons, decreased astrogliosis | ~26% | No data | (Crow et al., 2005) |
AM-1241 | Anti-inflammatory (CB2 receptor agonist) | Delayed progression | ~10% | No data | (Shoemaker et al., 2007) |
Ammonium Tetrathiomolybdate | Copper chelation | Prolonged survival and delayed onset | ~25% | No data | (Tokuda et al., 2008) |
BDNF | TrkB receptor activation | Improved motor performance, decreased atrophy and loss of ventral root myelinated fibers | No effect | Failed | (Ikeda et al., 1995) (Mitsumoto, Ikeda, Klinkosz, et al., 1994) |
Ceftriaxone | Increased EAAT2 transcription | Prolonged survival Increased EAAT2 levels, delayed loss of motor neurons | 8% | Failed | (Rothstein et al., 2005) |
Celastrol | Anti-inflammatory, anti-oxidant, prevents protein aggregation | Delayed onset, Improved weight loss and motor performance | 9–13% | No data | (Kiaei et al., 2005) |
Celecoxib | COX-2 inhibitor | Prolonged survival and delayed onset preserved motor neurons and decreased gliosis | 25% | Failed | (Drachman et al., 2002) |
CGP 3466B | Anti-apoptotic | Conflicting results: No effect seen in G93A mice, Slowing of motor neuron loss in pmn/pmn mice | No effect in SOD1-G93A 57% in pmn/pmn |
Failed | (Sagot et al., 2000) (Groeneveld et al., 2004) (R. Miller et al., 2007) |
Ciliary Neurotrophic Factor | Promotes motor neuron survival | Improved motor performance, decreased muscle atrophy, reduced loss of ventral root myelinated fibers | No effect | Failed | (Mitsumoto, Ikeda, Holmlund, et al., 1994) |
Cobalamin | Anti-glutamatergic, anti-oxidant, anti-apoptic | Prolonged survival and delayed onset improved motor performance | ~3% | Ongoing | (X. Zhang et al., 2008) |
Coenzyme Q | Anti-oxidant | Prolonged survival | ~4% | Failed | (Matthews et al., 1998) |
Creatine | Anti-oxidant, mitochondrial cofactor | Prolonged survival and delayed onset | 14.6% | Failed | (Andreassen et al., 2001) (Shefner et al., 2004) |
Dexpramipexole | Anti-oxidant Anti-apoptotic |
No Data | No Data | Failed | (Pattee et al., 2003) (H. Wang et al., 2008) |
Edaravone | Anti-oxidant | Improved motor performance and decreased SOD1 deposits | No effect | Ongoing | (Hidefumi Ito et al., 2008) |
Erythropoietin | Anti-inflammatory, anti-apoptotic | Conflicting results: Some studies prolonged survival and delayed onset; while others see no effect | ~10% | Failed | (Koh et al., 2007) (Lauria et al., 2009) |
G-CSF | Neuroprotective Antiapoptotic |
Increased MN survival | ~7% | None | (Pitzer et al., 2008) |
Gabapentin | Anti-glutamatergic | Prolonged survival | ~5% | Failed | (Gurney et al., 1996) |
GDNF | Promotes motor neuron survival | Prolonged survival and delayed onset improved motor function, reduced motor neuron loss | ~12% | Failed | (Acsadi et al., 2002) (M. Suzuki et al., 2008) |
Glatiramer acetate | Anti-inflammatory Anti-glutamatergic |
Conflicting results: some studies prolonged survival, while others did not | No effect | Failed | (Gordon et al., 2006) (Haenggeli et al., 2007) (Meininger et al., 2009) |
HGF | Neuroprotective Anti-apoptotic Anti-glutamatergic |
Prevented MN cell death prolonged survival |
18.8% | No data | (Sun et al., 2002) (Kadoyama et al., 2007) |
IGF-I | Neuroprotective Anti-oxidant |
Suppression of reactive species, protection against glutamate induced toxicity | No effect | Failed/Inconclusive | (Lai et al., 1997) (Sakowski et al., 2009) (Saenger et al., 2012) |
L-Arginine | Anti-glutamatergic | Prolonged survival, delayed onset and slowed neuropathology | 9–20% | No data | (J. Lee et al., 2009) |
Lithium | Promotes autophagy | Delayed disease onset and progression | ~36% | Failed | (Aggarwal et al., 2010) (Fornai et al., 2008) |
Mechno-growth factor | Neuroprotective | Increased survival of motor neurons | No effect | No data | (Riddoch-Contreras et al., 2009) |
Memantine | Anti-glutamatergic | Prolonged survival and delayed progression | ~5–7% | No data | (R. Wang and D. Zhang, 2005) (Joo et al., 2007) |
Minocycline | Anti-apoptotic Anti-inflammatory |
Delayed disease progression and onset | ~6–15% | Failed | (Kriz et al., 2002) (Zhu et al., 2002) (Van Den Bosch et al., 2002) (Gordon et al., 2004) |
N-acetylated alpha linked acidic dipeptidase | Anti-glutamatergic | Prevented MN cell death | 15% | No data | (Ghadge et al., 2003) |
N-acetylcystine | Anti-oxidant | Prolonged survival and delayed onset | ~7% | Failed | (Louwerse et al., 1995) (Andreassen et al., 2000) |
Nordihydroguaiaretic acid | Anti-inflammatory Anti-glutamatergic |
No effect on survival Increased glutamate uptake |
No effect | No data | (Boston-howes et al., 2008) |
ONO-2506 | Prevents astrogliosis Anti-inflammatory Anti-glutamatergic |
See Valproate (parent compound) | No data | Failed | (dePaulis, 2003) |
Pentoxifylline | Anti-apoptotic Anti-inflammatory |
No preclinical data | No data | Failed | (Meininger et al., 2006) |
Pioglitazone | Neuroprotective Anti-inflammatory Anti-apoptotic |
Prolonged survival and delayed onset | ~8% | Failed | (Schütz et al., 2005) |
Rasagiline | Anti-apoptotic | Prolonged survival | ~14% | Ongoing | (Waibel et al., 2004) |
Riluzole | Anti-glutamatergic | Prolonged survival and delayed onset | ~10% | Only effective treatment known | (Gurney et al., 1996) (Gurney et al., 1998) |
RO-26-2853 | Anti-inflammatory (inhibition of MMPs) | Prolonged survival and improved motor performance | ~11% | No data | (Lorenzl et al., 2006) |
Sodium phenylbutyrate | Protein aggregation inhibitor | Prolonged survival with conflicting results | 21.9% | Phase I Completed | (Ryu et al., 2005) (Cudkowicz et al., 2009) |
Talampanel | Anti-glutamatergic | Reduces MN calcium | No effect | Phase II Completed | (Paizs et al., 2011) (Pascuzzi et al., 2010) |
Thalidomide | Anti-inflamatory | Prolonged survival and decreased motor neuron cell death | ~11% | Failed | (Kiaei et al., 2006) (Stommel et al., 2009) |
Topiramate | Anti-glutamatergic | Prevents MN degeneration in culture | No effect | Failed | (Maragakis et al., 2003) (Cudkowicz et al., 2003) |
TRO19622 | Anti-oxidant | Prolonged survival and delayed onset | 10% | No data | (Bordet et al., 2007) |
Valproate | Histone deacetylase inhibitor | Conflicting results; two studies found prolonged survival, while two others showed no effect | No effect - 10% | Failed | (Sugai et al., 2004) (Feng et al., 2008) (Rouaux et al., 2007) (Crochemore et al., 2009) (Piepers et al., 2009) |
VEGF | Neuroprotective | Decreased astrogliosis and increased neuromuscular junctions; Decreased motor neuron death | No effect | Ongoing | (Zheng et al., 2007) (Tovar-Y-Romo et al., 2007) |
Vitamin E | Anti-oxidant | Delayed onset and slowed progression; does not prolong survival | No effect | Failed | (Gurney et al., 1996) (Terro et al., 1996) (Desnuelle et al., 2001) (Graf et al., 2005) |
zVAD-fmk | Anti-apoptotic | Prolonged survival and delayed onset | 22% | No data | (M. Li et al., 2000) |
In addition to the many issues mentioned above, there is also a widely recognized need for more animal models that mimic ALS. Fortunately, the last couple of years have yielded a number of new genes and protein targets that are associated with ALS (see above). Developing models from these targets will likely lead to the discovery of novel pathogenic mechanisms and possibly more effective treatment options.
Combinatorial therapies
As ALS is a disease with various pathways contributing to it, combination therapies targeting more than one disease-related pathway have proved effective at the preclinical stage and may provide an effective clinical option (Turner and Talbot, 2008). Zhang et al. (2003) treated SOD1-G93A mutant mice with a combination of minocycline, an anti-inflammatory and anti-apoptotic compound, and creatine, an anti-oxidant, and found a beneficially additive effect on survival, progression and onset of disease. However, recent attempts to utilize this strategy in the clinic have proved difficult. A phase II trial in ALS patients tested riluzole in combination with Pioglitazone, a drug with anti-inflammatory properties. Pioglitazone did not provide additional benefits as compared to riluzole alone (Dupuis et al., 2012). Riluzole has also been used in combination with lithium in a clinical trial, which did not provide a benefit in slowing ALS disease progression (Aggarwal et al., 2010). This trial was based on a pilot study, which established significant effects of the addition of lithium to riluzole in ALS patients (Fornai et al., 2008). This example, in particular, highlights the importance of selecting the proper combination therapies to test in the clinic. However, directly targeting multiple pathways and systems that are involved in ALS could establish more efficacious treatment. For example, Kong et al. (2012) treated mutant SOD1-G93A mice beginning at symptom onset with minocycline and ceftriaxone alone or in combination. Minocycline inhibits microglial activation and the apoptotic cascade, while ceftriaxone reduces excitotoxicity by activating the glutamate transporter, EAAT2. The combination treatment, significantly extended survival; no change was seen with single treatment of either compound compared to vehicle-treated mice.
Regulation of ABC transporters in health and disease
ATP-binding cassette (ABC) transporters in excretory and barrier tissues contribute to the difficulty in treating many systemic and CNS diseases using pharmacotherapy (Loscher and Potschka, 2005). These transporters are ATP-driven efflux pumps with remarkably broad substrate specificity and are responsible for the high urine-to-plasma and bile-to-plasma concentration ratios seen for some xenobiotics (excretory function) and for the inability of many xenobiotics to enter the CNS (barrier function) (Neuwelt et al., 2011). The superfamily of ABC transporters includes P-glycoprotein (P-gp), breast cancer resistance protein (Bcrp), and the family of multidrug resistant proteins (Mrp1, Mrp2, Mrp3, Mrp4, Mrp5, and Mrp6). While ABC transporters are primarily localized to the luminal plasma membrane of brain and spinal cord capillary endothelial cells, they are also expressed in other cells in the nervous system, including astrocytes, microglia and neurons (Neuwelt et al., 2011). The mRNA and protein expression patterns for many of the ABC transporters are currently being established in the cells comprising the neurovascular unit (endothelial cells, pericytes, and perivascular astrocytes) and CNS parenchyma (Hartz and Bauer, 2011). For example, the luminal membrane of capillary endothelial cells expresses P-gp, Bcrp, Mrp1, Mrp2, Mrp4, and Mrp5 (Bauer et al., 2006; Cooray et al., 2002; Nies et al., 2004). There are also reports of P-gp, Mrp1, and Mrp4 localizing to the abluminal membrane of brain capillary endothelial cells, however, efflux transporters on that membrane would not contribute to barrier function (Bendayan et al., 2006; Kilic et al., 2008; Pardridge et al., 1997; Soontornmalai et al., 2006; Zhang et al., 2004). Moreover, protein expression of P-gp, Bcrp, Mrp1, Mrp3, Mrp4, and Mrp5 has also been detected on astrocytic endfeet and microglia (Table 3).
Table 3.
Endothelial Cells | Astrocytes | Pericytes | Neurons | Microglia | Oligodendrocytes | Choroid Plexus | |
---|---|---|---|---|---|---|---|
P gp | + | + | + | + | + | + | |
BCRP | + | + | + | + | + | ||
Mrp1 | + | + | + | + | + | + | |
Mrp2 | + | + | |||||
Mrp3 | + | + | |||||
Mrp4 | + | + | + | + | |||
Mrp5 | + | + | + | + |
By determining the expression and localization of ABC transporters in situ, disease-driven alterations can be identified. For example, P-gp expression in AIDS patients with HIV encephalitis demonstrated a shift from the typical dominant endothelial cell expression to expression in astrocytes and microglia, which resulted in an overall increase in P-gp levels (Langford et al., 2004). In ALS, immunofluorescent labeling of P-gp in the lumbar spinal cord of SOD1-G93A mice indicates robust P-gp expression in astrocytes of symptomatic mice compared to pre-symptomatic mice (Figure 1). This disease-driven increase in spinal cord parenchyma P-gp expression could provide a secondary barrier to drug penetration.
Because many potential therapeutic compounds are ABC transporter substrates, treatments to blunt the increases of ABC transporters have been effective towards increasing the brain:plasma ratios of these compounds. Multiple pathways are associated with alterations in drug efflux transporter expression and transport activity. Activation of these pathways with xenobiotics or endogenous ligands serves to alter the expression and function of various ABC transporters. For instance, multiple ligand-activated transcription factors are capable of increasing P-gp expression in mouse and rat brain capillaries and in primary cultured rat and porcine brain endothelial cells (Miller, 2010). More specifically, activation of the pregnane X receptor (PXR) and the constitutive androstane receptor (CAR) by their respective ligands leads to increases in P-gp mRNA and protein expression (Chan et al., 2011). Furthermore, PXR activation in porcine brain capillary endothelial cells leads to increased expression and activity of P-gp and Bcrp (Lemmen et al., 2012). Similar types of regulation are exemplified in other ABC transporters as well. Activation of the transcription factor, peroxisome proliferator activated receptor alpha (PPARα), leads to increased expression and activity of Bcrp (Hoque et al., 2012). With prolonged treatment of TNF-alpha and Endothelin-1, P-gp expression and activity is increased in rat brain capillaries through an NFkB-dependent mechanism (Bauer et al., 2007). Extrapolating the results to in vivo studies, chronic inflammatory diseases may lead to increased expression and function of P-gp, which would work in reducing the efficacy of therapeutic compounds.
Multidrug efflux transporters mediate pharmacoresistance in ALS and other CNS disorders
Alterations in ABC transporter expression have been identified in epilepsy, brain cancer, HIV, spinal cord injury, Alzheimer’s disease, and Parkinson’s disease. In normal aging in the human brain, there is a slight decrease in ABC transporter function based on PET imaging with [11C]-verapamil, a well-characterized P-gp substrate (Bartels et al., 2009). The same finding was reported in a later study, which also identified a greater decrease in elderly men compared to elderly women (van Assema et al., 2012b). These overall reductions in normal aging could account for the susceptibility of degeneration in the elderly.
In epilepsy, excess glutamate due to seizure induction increases P-gp expression at the blood-brain barrier (Bankstahl et al., 2008). In addition, treatment with anti-epileptic drugs can result in upregulation of ABC transporters (Potschka, 2012). Combination therapy with tariquidar, a P-gp inhibitor, and phenobarbital was effective in a rat model of drug-resistant epilepsy. Therapy with phenobarbital alone did not alter seizure activity in these rats (Brandt et al., 2006). To understand how these transporters are regulated in disease, the mechanisms leading to alterations in transporters are beginning to emerge. Recent studies suggest that during seizures excess glutamate drive increased P-gp expression by signaling through COX-2, PGE2 and NFkB (Bauer et al., 2008). Consistent with this, P-gp expression levels were decreased in kainate-induced epilepsy mice when treated with a COX-2 inhibitor or a NFkB inhibitor; however it is likely that this signaling pathway is not the only one that drives increases in P-gp expression (Yu et al., 2011).
Drug efflux transporter-mediated chemoresistance has been identified and studied extensively in certain cancers such as glioblastoma multiforme (GBM) and other metastatic and primary brain cancers (Agarwal et al., 2011). The importance of P-gp contribution to chemoresistance remains a perplexing issue and the challenges associated with chemoresistance have proven difficult to address in the clinic (Shaffer et al., 2012).
Inflammation, which accompanies all CNS diseases, also signals changes in ABC transporter expression. As P-gp is known to increase under conditions of inflammation, treatment with an anti-inflammatory drug, licofelone, improved the bioavailability of riluzole, as well as reduced levels of P-gp expression in vivo (Dulin et al., 2013). However, all conditions of inflammation do not consistently produce similar change. For example, P-gp expression was decreased in a rat brain capillary endothelial cell line after exposure to TNF-alpha (Theron et al., 2003). From available evidence, it is evident that whether P-gp increases or decreases in response to inflammation depends on the nature of the stimulus and the time of exposure. For example, in rat brain capillaries, Hartz et al. (2006, 2004) found a rapid and reversible decrease in P-gp transport function with activation of innate immune signaling through TNF-alpha and endothelin-1 (ET-1). However, at later time points, P-gp expression and activity was upregulated with TNF-alpha and ET-1 exposure of isolated rat brain capillaries (Bauer et al., 2007).
In contrast, two neurodegenerative diseases lead to decreases in expression and function of ABC transporters. One proposed cause of excess Aβ protein accumulation in Alzheimer’s disease (AD) is reduced efflux of the protein from the brain (Cirrito et al., 2005). Brains from patients with AD show decreased P-gp expression (van Assema et al., 2012a). Studies with animal models indicate that Aβ protein can be transported by both P-gp and Bcrp, interesting observations given the reduced Aβ protein clearance from the brain in AD models. In AD model mice, there is a significant accumulation of Aβ if P-gp is inhibited, indicating that clearance, rather than the increased production of Aβ is contributing to its removal and plaque formation (Cirrito et al., 2005). Consistent with that observation, increasing P-gp expression in those mice reduces brain Aβ accumulation (Hartz et al., 2010). However, it appears that not all transporters are modulated in the same direction as increases in Bcrp in AD patients and animal models have been found (Xiong et al., 2009). Bcrp-null mice accumulate more Aβ in brain parenchyma compared to wild-type mice, so it is not clear at present how changes in Bcrp expression contribute to AD pathology (Xiong et al., 2009).
One proposed contributing factor to the etiology of Parkinson’s disease is exposure to environmental toxins that may target dopaminergic neurons in the striatum. Some of these toxins are substrates for P-gp and a subset of human Parkinson’s disease patients exhibit reductions in ABCB1 (P-gp) gene expression in brain (Westerlund et al., 2008). Understanding ABC transporter alterations in neurodegeneration, therefore, may lead to improvements in current and future treatments.
Although ABC transporter expression levels at the blood-brain barrier have been shown to change in several neurodegenerative diseases, parallel studies for ALS are just beginning. Multiple reports have identified increases in P-gp expression in ALS mutant mice (Boston-howes et al., 2008; Jablonski et al., 2012; Milane et al., 2010). Furthermore, Bcrp has been implicated as another transporter with increased expression of mRNA and protein in ALS mutant mice (Jablonski et al., 2012), although another report did not detect mRNA and protein increases of Bcrp (Milane et al., 2010). The reason for this discrepancy could be that Milane et al. (2010) isolated mRNA and capillaries from whole brain, which is not the primary region of degeneration in the mouse, while Jablonski et al. (2012) isolated mRNA and capillaries from the spinal cord of ALS mutant mice. Examination of the BBB and BSCB at areas of degeneration in the mutant ALS mice reveals specific, disease-driven expression and functional increases of two ABC transporters, P-gp and Bcrp, in the spinal cord and cortex of SOD1-G93A mice (Jablonski et al., 2012). In isolated cortex and spinal cord capillaries from mutant SOD1-G93A mice, P-gp and Bcrp expression levels are increased as the disease progresses. These increases are specific to the CNS as protein expression of P-gp and Bcrp in the liver of these mice is unchanged. Furthermore, the functional activity of P-gp and Bcrp in the isolated capillaries is increased in symptomatic compared to presymptomatic mice, indicating the functional relevance in P-gp and Bcrp in extruding pharmacological agents from the CNS. Increased protein levels of P-gp and Bcrp have been detected in post-mortem human lumbar spinal cords of ALS patients, confirming the relevance of the mouse findings for the human disease (Jablonski et al., 2012).
Strategies to improve treatment for ALS
Improving the bioavailability of therapeutics: Overcoming Pharmacoresistance
One of the limiting factors associated with preclinical drug trials in ALS, as well as other CNS disorders, is the ability of therapeutic drugs to enter the CNS and remain there. For ALS, the issue of pharmacoresistance and the consequences of altered bioavailability have been largely ignored. One can combat limited drug movement across the blood-brain and blood-spinal cord barriers by modifying the route of drug delivery, e.g., systemic vs direct CNS delivery. Although not discussed in detail in this review, modifying or altering the delivery route of a compound requires the systematic analysis of the altered pharmacokinetics, dynamics, and penetration of that compound. Another way to increase the bioavailability and improve efficacy of compounds is to target mechanisms that limit the accumulation of therapeutics in the CNS. Many proposed ALS therapeutics are substrates for or interact with P-gp and/or Bcrp. These include, Riluzole, Minocycline, and Ceftriaxone (Boston-howes et al., 2008; Gosland et al., 1989; Milane et al., 2009, 2007). Thus, for drugs like Riluzole, disease-driven increases in transporter expression at the blood-brain and blood-spinal cord barriers limit penetration into the CNS (Milane et al., 2007). In addition, consider NDGA, a compound that increases glutamate uptake that was tested as a potential therapeutics for ALS (Boston-howes et al., 2008). In a preclinical trial with wild-type mice, NDGA increased glutamate uptake over 30 days of treatment. In the SOD1-G93A mice, the increase in glutamate uptake was only sustained for 10 days, before a return to lower levels. Concomitant with the decrease of glutamate uptake, was a steady increase in expression levels of the ABC transporter, P-gp. Further experiments identify NDGA as a substrate of P-gp (Figure 2). As an initial proof of principle to inhibiting P-gp in ALS mice, transgenic mutant SOD1-G93A/P-gp knockout mice were treated with a known P-gp substrate and protectant against mitochondrial-mediated apoptosis, cyclosporine A. Survival was extended by 12% in the SOD1-G93A/P-gp knockout mice compared to mutant SOD1-G93A mice given cyclosporine A (Kirkinezos et al., 2004). The combined effect would seem marginal, however, cyclosporine A was shown to be toxic to neuronal cells expressing mutant SOD1 (Maxwell et al., 2004) and perhaps was not an appropriate therapeutic agent.
While it is possible to inhibit efflux transporters with various pharmacological agents, these approaches tend to produce severe side-effects. This can be evidenced by the lack of efficacy in various types of clinical cancer trials (Agarwal et al., 2011; Shaffer et al., 2012). Inhibition does not necessarily target specific tissues, leading to issues of selectivity that could potentially exacerbate disease conditions with chronic treatment. However, subsequent generations of inhibitors are becoming more specific for individual transporters and much more effective at reducing side-effects. Various third generation inhibitors (i.e. tariquidar and elacridar) have more potency and specificity than earlier inhibitors. They are also tolerated well in human trials (Bauer et al., 2012). An alternative approach would be to target the signaling pathways that regulate basal transporter activity. In a recent study using rats, Cannon et al. (2012) decreased P-gp activity at the blood-brain barrier by targeting the sphingosine-1-phosphate (S1P) receptor 1. Using fingolimod, a S1P receptor agonist approved for use in the clinic, they demonstrated increased brain accumulation in vivo of several drugs that are P-gp substrates. A second recent publications shows that spinal cord capillaries possess the same S1P-based signaling pathway (Cartwright et al., 2012).
For ALS, the clinical relevance of pharmacoresistance is still somewhat uncertain. When we pay attention to lessons learned from other diseases, the arguments for induced pharmacoresistance in ALS are strengthened and potential mechanisms of induction are disclosed. For example, epilepsy and ALS are characterized by glutamate excitotoxicity, which can lead to up-regulation of P-gp. In epilepsy, excess glutamate released from neurons in the synaptic cleft activates the NMDA receptor on endothelial cells to release arachidonic acid, which is converted to PGE2 (via COX-2), activating the EP1 receptor and NFkB and increasing ABCB1 gene transcription and translation (Bauer et al., 2008). Identification of the pathways controlling P-gp and Bcrp up-regulation in ALS could lead to more targeted manipulation of the transporters.
Conclusion
ALS is a disease of the upper and lower motor neurons, however, these are not the only cell types affected. The non-cell autonomous nature of ALS results in a variety of pathogenic mechanisms contributing to disease. With only one moderately effective drug that can be used in the clinic, riluzole, there is a desperate need for the development of more efficacious treatments. The standard method of identifying a potential therapeutic agent, demonstrating an extension of lifespan in a mouse model, and moving to a clinical trial has not produced effective therapies. The complexity of disease and the disparity between effective preclinical trials and ineffective clinical trials requires further research on underlying ALS disease mechanisms and a re-thinking of clinical treatment strategies in light of new information. In recent years, some combination therapies have proven successful at the preclinical stage. Indeed, combination treatment can begin at the onset of symptoms in the ALS mice and produce a significant extension of lifespan, making this strategy particularly appealing. Translating this strategy to the clinic may prove more beneficial than the use of single therapeutic treatments initially tested in presymptomatic ALS mice.
Finally, given the recent discovery of a selective and disease-driven pharmacoresistance seen in ALS mice and the upregulation of ABC transporters in human patients with the disease, targeting pharmacoresistance appears to be one way to improve the efficacy of ALS therapeutics. The ability to control P-gp and BCRP transport activity in ALS patients could prove invaluable. Activities of P-gp and Bcrp could be controlled by use of pharmacologic inhibitors of the transporters or by targeting pathways that regulate activity or expression. However, first we must identify how ABC transporters are upregulated in the brain and spinal cord endothelial barriers and in brain parenchymal cells of the CNS. These studies will initially have to be done using ALS mouse models rather than healthy, wild-type controls. Moreover, the determination of bioavailability and proper dosage should be studied throughout the course of the disease so that changes in dosage of therapeutics can be tied to increases of P-gp and Bcrp expression. In this instance, the issues of pharmacokinetics and pharmacodynamics require persistent analysis throughout the ALS disease course and should be studied appropriately.
Highlights.
ABC drug efflux transporters are a potential cause for pharmacoresistance in ALS
Pre-clinical drug trials have failed to cure the ALS mouse model
Novel pharmacological approaches require combating pharmacoresistance in ALS
Acknowledgments
The authors thank Warren Anderson, Nathan Henderson, and Eric Kostuk for assembly of Table 1 and Supplementary Table 1. This work was supported by the National Institute of Health grant RO1-NS074886 (DT), DoD grant 08019250x13401 (PP), the Muscular Dystrophy Association Developmental Award (DAJ), F31-NS080539 (MRJ) and by the Intramural Research Program of the National Institute of Environmental Health Sciences, National Institutes of Health (DSM). The Weinberg Unit for ALS research is also supported by the Farber Family Foundation.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- Agarwal S, Hartz AMS, Elmquist WF, Bauer B. Breast cancer resistance protein and P-glycoprotein in brain cancer: two gatekeepers team up. Curr Pharm Des. 2011;17:2793–802. doi: 10.2174/138161211797440186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aggarwal SP, Zinman L, Simpson E, Mckinley J, Jackson KE, Pinto H, Conwit RA, Schoenfeld DA. Clinical Trial Testing Lithium in ALS Terminates Early for Futility. Lancet neurol. 2010;9:481–488. doi: 10.1016/S1474-4422(10)70068-5.Clinical. [DOI] [Google Scholar]
- Al-Chalabi A, Jones A, Troakes C, King A, Al-Sarraj S, van den Berg LH. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol. 2012;124:339–52. doi: 10.1007/s00401-012-1022-4. [DOI] [PubMed] [Google Scholar]
- Bankstahl JP, Hoffmann K, Bethmann K, Loscher W. Glutamate is critically involved in seizure-induced overexpression of P-glycoprotein in the brain. Neuropharmacol. 2008;54:1006–16. doi: 10.1016/j.neuropharm.2008.02.008. [DOI] [PubMed] [Google Scholar]
- Bartels AL, Kortekaas R, Bart J, Willemsen ATM, de Klerk OL, de Vries JJ, van Oostrom JCH, Leenders KL. Blood-brain barrier P-glycoprotein function decreases in specific brain regions with aging: a possible role in progressive neurodegeneration. Neurobiol Aging. 2009;30:1818–24. doi: 10.1016/j.neurobiolaging.2008.02.002. [DOI] [PubMed] [Google Scholar]
- Bauer B, Hartz AMS, Miller DS. Tumor Necrosis Factor-alpha and Endothelin-1 Increase P-Glycoprotein Expression and Transport Activity at the Blood-Brain Barrier. Molec Pharmacol. 2007;71:667–75. doi: 10.1124/mol.106.029512.with. [DOI] [PubMed] [Google Scholar]
- Bauer B, Hartz AMS, Pekcec A, Toellner K, Miller DS, Potschka H. Seizure-Induced Up-Regulation of P-Glycoprotein at the Blood-Brain Barrier through Glutamate and Cyclooxygenase-2 Signaling. Molec Pharmacol. 2008;73:1444–1453. doi: 10.1124/mol.107.041210.show. [DOI] [PubMed] [Google Scholar]
- Bauer B, Yang X, Hartz AMS, Olson ER, Zhao R, Kalvass JC, Pollack GM, Miller DS. In Vivo Activation of Human Pregnane X Receptor Tightens the Blood-Brain Barrier to Methadone through P-Glycoprotein. Molec Pharmacol. 2006;70:1212–19. doi: 10.1124/mol.106.023796.inhibitors. [DOI] [PubMed] [Google Scholar]
- Bauer M, Zeitlinger M, Karch R, Matzneller P, Stanek J, Jäger W, Böhmdorfer M, Wadsak W, Mitterhauser M, Bankstahl JP, Löscher W, Koepp M, Kuntner C, Müller M, Langer O. Pgp-mediated interaction between (R)-[11C]verapamil and tariquidar at the human blood-brain barrier: a comparison with rat data. Clin Pharmacol Ther. 2012;91:227–33. doi: 10.1038/clpt.2011.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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]
- Bendayan R, Ronaldson PT, Gingras D, Bendayan M. In situ localization of P-glycoprotein (ABCB1) in human and rat brain. J Histochem Cytochem. 2006;54:1159–67. doi: 10.1369/jhc.5A6870.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boston-howes W, Williams EO, Bogush A, Scolere M, Pasinelli P, Trotti D. Nordihydroguaiaretic acid increases glutamate uptake in vitro and in vivo: Therapeutic implications for amyotrophic lateral sclerosis. Exp Neurol. 2008;213:229–37. doi: 10.1016/j.expneurol.2008.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandt C, Bethmann K, Gastens AM, Löscher W. The multidrug transporter hypothesis of drug resistance in epilepsy: Proof-of-principle in a rat model of temporal lobe epilepsy. Neurobiol Dis. 2006;24:202–11. doi: 10.1016/j.nbd.2006.06.014. [DOI] [PubMed] [Google Scholar]
- Cannon RE, Peart JC, Hawkins BT, Campos CR, Miller DS. Targeting blood-brain barrier sphingolipid signaling reduces basal P-glycoprotein activity and improves drug delivery to the brain. Proc Natl Acad Sci U S A. 2012;109:15930–5. doi: 10.1073/pnas.1203534109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cartwright TA, Campos CR, Cannon RE, Miller DS. Mrp1 is essential for sphingolipid signaling to p-glycoprotein in mouse blood-brain and blood-spinal cord barriers. J Cereb Blood Flow Metab. 2012:1–8. doi: 10.1038/jcbfm.2012.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chan GNY, Hoque MT, Cummins CL, Bendayan R. Regulation of P-glycoprotein by orphan nuclear receptors in human brain microvessel endothelial cells. J Neurochem. 2011;118:163–75. doi: 10.1111/j.1471-4159.2011.07288.x. [DOI] [PubMed] [Google Scholar]
- Cirrito JR, Deane R, Fagan AM, Spinner ML, Parsadanian M, Finn MB, Jiang H, Prior JL, Sagare A, Bales KR, Paul SM, Zlokovic BV, Piwnica-worms D, Holtzman DM. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model. J Clin Invest. 2005;115:3285–90. doi: 10.1172/JCI25247.mdr1a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cooray HC, Blackmore CG, Maskell L, Barrand Ma. Localisation of breast cancer resistance protein in microvessel endothelium of human brain. Neuroreport. 2002;13:2059–63. doi: 10.1097/00001756-200211150-00014. [DOI] [PubMed] [Google Scholar]
- DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, Nicholson AM, Finch Na, Flynn H, Adamson J, Kouri N, Wojtas A, Sengdy P, Hsiung GYR, Karydas A, Seeley WW, Josephs Ka, Coppola G, Geschwind DH, Wszolek ZK, Feldman H, Knopman DS, Petersen RC, Miller BL, Dickson DW, Boylan KB, Graff-Radford NR, Rademakers R. 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]
- Deng HX, Chen W, Hong ST, Boycott KM, Gorrie GH, Siddique N, Yang Y, Fecto F, Shi Y, Zhai H, Jiang H, Hirano M, Rampersaud E, Jansen GH, Donkervoort S, Bigio EH, Brooks BR, Ajroud K, Sufit RL, Haines JL, Mugnaini E, Pericak-Vance Ma, Siddique T. Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia. Nature. 2011;477:211–5. doi: 10.1038/nature10353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dulin JN, Moore ML, Grill RJ. The dual cyclooxygenase/5-lipoxygenase inhibitor licofelone attenuates p-glycoprotein-mediated drug resistance in the injured spinal cord. J Neurotrauma. 2013;30:211–26. doi: 10.1089/neu.2012.2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dupuis L, Dengler R, Heneka MT, Meyer T, Zierz S, Kassubek J, Fischer W, Steiner F, Lindauer E, Otto M, Dreyhaupt J, Grehl T, Hermann A, Winkler AS, Bogdahn U, Benecke R, Schrank B, Wessig C, Grosskreutz J, Ludolph AC. A randomized, double blind, placebo-controlled trial of pioglitazone in combination with riluzole in amyotrophic lateral sclerosis. PLoS One. 2012;7:e37885. doi: 10.1371/journal.pone.0037885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol. 2011;7:616–630. doi: 10.1038/nrneurol.2011.152. [DOI] [PubMed] [Google Scholar]
- Fornai F, Longone P, Cafaro L, Kastsiuchenka O, Ferrucci M, Manca ML, Lazzeri G, Spalloni A, Bellio N, Lenzi P, Modugno N, Siciliano G, Isidoro C, Murri L, Ruggieri S, Paparelli A. Lithium delays progression of amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A. 2008;105:2052–7. doi: 10.1073/pnas.0708022105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garbuzova-Davis S, Rodrigues MCO, Hernandez-Ontiveros DG, Louis MK, Willing AE, Borlongan CV, Sanberg PR. Amyotrophic lateral sclerosis: A neurovascular disease. Brain Res. 2011;1398:113–25. doi: 10.1016/j.brainres.2011.04.049. [DOI] [PubMed] [Google Scholar]
- Gosland MP, Lum BL, Sikic BI. Reversal by Cefoperazone of Resistance to Etoposide, Doxorubicin, and Vinblastine in Multidrug Resistant Human Sarcoma Cells. Cancer Res. 1989;49:6901–05. [PubMed] [Google Scholar]
- Hartz AMS, Bauer B. ABC transporters in the CNS - an inventory. Curr Pharm Biotechnol. 2011;12:656–73. doi: 10.2174/138920111795164020. [DOI] [PubMed] [Google Scholar]
- Hartz AMS, Bauer B, Fricker G, Miller DS. Rapid Regulation of P-Glycoprotein at the Blood-Brain Barrier by Endothelin-1. Molec Pharmacol. 2004;66:387–94. doi: 10.1124/mol.104.001503.bition. [DOI] [PubMed] [Google Scholar]
- Hartz AMS, Bauer B, Pekcec A, Toellner K, Miller DS. Rapid Modulation of P-Glycoprotein-Mediated Transport at the Blood-Brain Barrier by Tumor Necrosis Factor-alpha and Lipopolysaccharide. Molec Pharmacol. 2006;69:462–70. doi: 10.1124/mol.105.017954.pumps. [DOI] [PubMed] [Google Scholar]
- Hartz AMS, Miller DS, Bauer B. Restoring Blood-Brain Barrier P-glycoprotein Reduces Brain Aβ in a Mouse Model of Alzheimer’s Disease. Molec Pharmacol. 2010;77:715–35. doi: 10.1124/mol.109.061754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoque MT, Robillard KR, Bendayan R. Regulation of breast cancer resistant protein by peroxisome proliferator-activated receptor α in human brain microvessel endothelial cells. Molec Pharmacol. 2012;81:598–609. doi: 10.1124/mol.111.076745. [DOI] [PubMed] [Google Scholar]
- Jablonski MR, Jacob DA, Campos CR, Miller DS, Maragakis NJ, Pasinelli P, Trotti D. Selective increase of two ABC drug efflux transporters at the blood-spinal cord barrier suggests induced pharmacoresistance in ALS. Neurobiol Dis. 2012;47:194–200. doi: 10.1016/j.nbd.2012.03.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson JO, Mandrioli J, Benatar M, Abramzon Y, Van Deerlin VM, Trojanowski JQ, Gibbs JR, Brunetti M, Gronka S, Wuu J, Ding J, McCluskey LF, Martinez-Lage M, Falcone D, Hernandez DG, Arepalli S, Chong S, Schymick JC, Rothstein JD, Landi F, Wang YD, Calvo A, Mora G, Sabatelli M, Monsurrò MR, Battistini S, Salvi F, Spataro R, Sola P, Borghero G, Galassi G, Scholz SW, Taylor JP, Restagno G, Chiò A, Traynor BJ. Exome sequencing reveals VCP mutations as a cause of familial ALS. Neuron. 2010;68:857–64. doi: 10.1016/j.neuron.2010.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kilic E, Spudich A, Kilic U, Rentsch KM, Vig R, Matter CM, Wunderli-Allenspach H, Fritschy JM, Bassetti CL, Hermann DM. ABCC1: a gateway for pharmacological compounds to the ischaemic brain. Brain. 2008;131:2679–89. doi: 10.1093/brain/awn222. [DOI] [PubMed] [Google Scholar]
- Kirkinezos IG, Hernandez D, Bradley WG, Moraes CT. An ALS mouse model with a permeable blood-brain barrier benefits from systemic cyclosporine A treatment. J Neurochem. 2004;88:821–26. doi: 10.1046/j.1471-4159.2003.02181.x. [DOI] [PubMed] [Google Scholar]
- Knippenberg S, Thau N, Dengler R, Petri S. Significance of behavioural tests in a transgenic mouse model of amyotrophic lateral sclerosis (ALS) Behav Brain Res. 2010;213:82–7. doi: 10.1016/j.bbr.2010.04.042. [DOI] [PubMed] [Google Scholar]
- Kong Q, Carothers S, Chang Y, Glenn Lin CL. The importance of preclinical trial timing - a potential reason for the disconnect between mouse studies and human clinical trials in ALS. CNS Neurosci Ther. 2012;18:791–3. doi: 10.1111/j.1755-5949.2012.00358.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwiatkowski TJ, Bosco Da, Leclerc aL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau Ga, Hosler Ba, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance Ma, Yan J, Ticozzi N, Siddique T, McKenna-Yasek D, Sapp PC, Horvitz HR, Landers JE, Brown RJ. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science (80-) 2009;323:1205–8. doi: 10.1126/science.1166066. [DOI] [PubMed] [Google Scholar]
- Landis SC, Amara SG, Asadullah K, Austin CP, Blumenstein R, Bradley EW, Crystal RG, Darnell RB, Ferrante RJ, Fillit H, Finkelstein R, Fisher M, Gendelman HE, Golub RM, Goudreau JL, Gross Ra, Gubitz AK, Hesterlee SE, Howells DW, Huguenard J, Kelner K, Koroshetz W, Krainc D, Lazic SE, Levine MS, Macleod MR, McCall JM, Moxley RT, Narasimhan K, Noble LJ, Perrin S, Porter JD, Steward O, Unger E, Utz U, Silberberg SD. A call for transparent reporting to optimize the predictive value of preclinical research. Nature. 2012;490:187–91. doi: 10.1038/nature11556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langford D, Grigorian A, Hurford R, Adame A, Ellis RJ, Hansen L, Masliah E. Altered P-glycoprotein expression in AIDS patients with HIV encephalitis. J Neuropathol Exp Neurol. 2004;63:1038–47. doi: 10.1093/jnen/63.10.1038. [DOI] [PubMed] [Google Scholar]
- Lemmen J, Tozakidis IEP, Galla H-J. Pregnane X receptor upregulates ABC-transporter Abcg2 and Abcb1 at the blood-brain barrier. Brain Res. 2012 doi: 10.1016/j.brainres.2012.10.060. [DOI] [PubMed] [Google Scholar]
- Loscher W, Potschka H. Drug resistance in brain diseases and the role of drug efflux transporters. Nat Rev Neurosci. 2005;6:591–602. doi: 10.1038/nrn1728. [DOI] [PubMed] [Google Scholar]
- Ludolph AC, Bendotti C, Blaugrund E, Hengerer B, Löffler JP, Martin J, Meininger V, Meyer T, Moussaoui S, Robberecht W, Scott S, Silani V, Van Den Berg LH. 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]
- Maxwell MM, Pasinelli P, Kazantsev AG, Brown RJ. RNA interference-mediated silencing of mutant superoxide dismutase rescues cyclosporin A-induced death in cultured neuroblastoma cells. Proc Natl Acad Sci U S A. 2004;101:3178–83. doi: 10.1073/pnas.0308726100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Milane A, Fernandez C, Dupuis L, Buyse M, Loeffler J, Farinotti R, Meininger V, Bensimon G. Neuroscience Letters P-glycoprotein expression and function are increased in an animal model of amyotrophic lateral sclerosis. Neurosci Lett. 2010;472:166–70. doi: 10.1016/j.neulet.2010.01.078. [DOI] [PubMed] [Google Scholar]
- Milane A, Fernandez C, Vautier S, Bensimon G, Meininger V, Farinotti R. Minocycline and riluzole brain disposition: interactions with p-glycoprotein at the blood-brain barrier. J Neurochem. 2007;103:164–73. doi: 10.1111/j.1471-4159.2007.04772.x. [DOI] [PubMed] [Google Scholar]
- Milane A, Vautier S, Chacun H, Meininger V, Bensimon G, Farinotti R, Fernandez C. Interactions between riluzole and ABCG2/BCRP transporter. Neurosci Lett. 2009;452:12–6. doi: 10.1016/j.neulet.2008.12.061. [DOI] [PubMed] [Google Scholar]
- Miller DS. Regulation of P-glycoprotein and other ABC drug transporters at the blood-brain barrier. Trends Pharmacol Sci. 2010;31:246–54. doi: 10.1016/j.tips.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller DS, Mitchell J, Moore D. Riluzole for amyotrophic lateral sclerosis (ALS)/motor neuron disease (MND) Cochrane Database 2012 [Google Scholar]
- Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar Ha, Trojanowski JQ, Lee VM-Y. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science (80-) 2006;314:130–3. doi: 10.1126/science.1134108. [DOI] [PubMed] [Google Scholar]
- Neuwelt Ea, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, Leybaert L, Molnár Z, O’Donnell ME, Povlishock JT, Saunders NR, Sharp F, Stanimirovic D, Watts RJ, Drewes LR. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci. 2011;12:169–82. doi: 10.1038/nrn2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nies A, Jedlitschky G, König J, Herold-Mende C, Steiner HH, Schmitt HP, Keppler D. Expression and immunolocalization of the multidrug resistance proteins, MRP1-MRP6 (ABCC1-ABCC6), in human brain. Neuroscience. 2004;129:349–60. doi: 10.1016/j.neuroscience.2004.07.051. [DOI] [PubMed] [Google Scholar]
- Pardridge WM, Golden PL, Kang YS, Bickel U. Brain microvascular and astrocyte localization of P-glycoprotein. J Neurochem. 1997;68:1278–85. doi: 10.1046/j.1471-4159.1997.68031278.x. [DOI] [PubMed] [Google Scholar]
- Potschka H. Role of CNS efflux drug transporters in antiepileptic drug delivery: overcoming CNS efflux drug transport. Adv Drug Deliv Rev. 2012;64:943–52. doi: 10.1016/j.addr.2011.12.007. [DOI] [PubMed] [Google Scholar]
- Renton AE, Majounie E, Waite AJ, Simón-Sánchez J, Rollinson S, Gibbs JR, Schymick JC, Laaksovirta H, van Swieten JC, Myllykangas L, Kalimo H, Paetau A, Abramzon Y, Remes AM, Kaganovich A, Scholz SW, Duckworth J, Ding J, Harmer DW, Hernandez DG, Johnson JO, Mok K, Ryten M, Trabzuni D, Guerreiro RJ, Orrell RW, Neal JW, Murray A, Pearson J, Jansen IE, Sondervan D, Seelaar H, Blake DJ, Young K, Halliwell N, Callister JB, Toulson G, Richardson A, Gerhard A, Snowden J, Mann D, Neary D, Nalls Ma, Peuralinna T, Jansson L, Isoviita VM, Kaivorinne AL, Hölttä-Vuori M, Ikonen E, Sulkava R, Benatar M, Wuu J, Chiò A, Restagno G, Borghero G, Sabatelli M, Heckerman D, Rogaeva E, Zinman L, Rothstein JD, Sendtner M, Drepper C, Eichler EE, Alkan C, Abdullaev Z, Pack SD, Dutra A, Pak E, Hardy J, Singleton A, Williams NM, Heutink P, Pickering-Brown S, Morris HR, Tienari PJ, Traynor BJ. 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]
- Rosen D, Siddique T, Patterson D, Figlewicz D, Sapp PC, Hentati A. 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]
- Shaffer BC, Gillet JP, Patel C, Baer MR, Bates SE, Gottesman MM. Drug resistance: still a daunting challenge to the successful treatment of AML. Drug Disc Today. 2012;15:62–9. doi: 10.1016/j.drup.2012.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siddique T, Ajroud-Driss S. Familial amyotrophic lateral sclerosis, a historical perspective. Acta Myol. 2011;30:117–20. [PMC free article] [PubMed] [Google Scholar]
- Soontornmalai a, Vlaming MLH, Fritschy JM. Differential, strain-specific cellular and subcellular distribution of multidrug transporters in murine choroid plexus and blood-brain barrier. Neuroscience. 2006;138:159–69. doi: 10.1016/j.neuroscience.2005.11.011. [DOI] [PubMed] [Google Scholar]
- Theron D, de Lagerie SB, Tardivel S, Pelerin H, Demeuse P, Mercier C, Mabondzo A, Farinotti R, Lacour B, Roux F, Gimenez F. Influence of tumor necrosis factor-alpha on the expression and function of P-glycoprotein in an immortalised rat brain capillary endothelial cell line. Biochem Pharmacol. 2003;66:579–587. doi: 10.1016/s0006-2952(03)00340-x. [DOI] [PubMed] [Google Scholar]
- Turner BJ, Talbot K. Transgenics, toxicity and therapeutics in rodent models of mutant SOD1-mediated familial ALS. Prog Neurobiol. 2008;85:94–134. doi: 10.1016/j.pneurobio.2008.01.001. [DOI] [PubMed] [Google Scholar]
- Van Assema DME, Lubberink M, Bauer M, van der Flier WM, Schuit RC, Windhorst AD, Comans EFI, Hoetjes NJ, Tolboom N, Langer O, Müller M, Scheltens P, Lammertsma Aa, van Berckel BNM. Blood-brain barrier P-glycoprotein function in Alzheimer’s disease. Brain. 2012a;135:181–9. doi: 10.1093/brain/awr298. [DOI] [PubMed] [Google Scholar]
- Van Assema DME, Lubberink M, Boellaard R, Schuit RC, Windhorst AD, Scheltens P, Lammertsma Aa, van Berckel BNM. P-glycoprotein function at the blood-brain barrier: effects of age and gender. Mol Imaging Biol. 2012b;14:771–6. doi: 10.1007/s11307-012-0556-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vance C, Rogelj B, Hortobagyi T, Shaw CE. Mutations in FUS, an RNA Processing Protein, Cause Familial Amyotrophic Lateral Sclerosis Type 6. Science (80-) 2009;323:1208–1211. doi: 10.1126/science.1165942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Westerlund M, Belin AC, Olson L, Galter D. Expression of multi-drug resistance 1 mRNA in human and rodent tissues: reduced levels in Parkinson patients. Cell Tissue Res. 2008;334:179–85. doi: 10.1007/s00441-008-0686-5. [DOI] [PubMed] [Google Scholar]
- Xiong H, Callaghan D, Jones A, Bai J, Rasquinha I, Smith C, Pei K, Walker D, Lue LF, Stanimirovic D, Zhang W. ABCG2 is upregulated in Alzheimer’s brain with cerebral amyloid angiopathy and may act as a gatekeeper at the blood-brain barrier for Abeta(1-40) peptides. J Neurosci. 2009;29:5463–75. doi: 10.1523/JNEUROSCI.5103-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yu N, Di Q, Liu H, Hu Y, Jiang Y, Yan Y, Zhang Y, Zhang Y. Nuclear factor-kappa B activity regulates brain expression of P-glycoprotein in the kainic acid-induced seizure rats. Mediat Inflamm 670613. 2011 doi: 10.1155/2011/670613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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]
- Zhang Y, Schuetz JD, Elmquist WF, Miller DW. Plasma Membrane Localization of Multidrug Resistance-Associated Protein Homologs in Brain Capillary Endothelial Cells. J Pharmacol Exp Ther. 2004;311:449–55. doi: 10.1124/jpet.104.068528.pathological. [DOI] [PubMed] [Google Scholar]