The present disclosure is directed to methods of gene therapy for treating a subject with Allan-Herndon-Dudley syndrome, and to methods of combining administration of 3,5-diiodothyropropionic acid (DITPA) to a subject with Allan-Herndon-Dudley syndrome with gene therapy administered to the subject.
Allan-Herndon-Dudley Syndrome (“ARDS”) is an X-linked recessive developmental disorder causing intellectual disability and movement issues in males. Specifically, patients with ARDS have a mutant SLC16A2 gene resulting in a malformed monocarboxylate transporter 8 (“MCT8”) protein. MCT8 is a thyroid hormone cell membrane transporter.
In 2004 it was demonstrated that a severe neuropsychomotor defect observed in males in 1944 by Allan Herndon and Dudley1 was caused by defects of MCT8 gene (also known as SLC16A2)2. These individuals, mostly children, cannot walk or talk because of deficiency in the active thyroid hormone T3 in brain. However, at the same time, these children have increased metabolism and failure to gain weight because the excess of T3 present in blood entered other body tissues though alternative transporters.
Symptoms of ARDS are caused by a lack of cellular uptake of the thyroid hormone triiodothyronine (“T3”), which is normally transported across the cell membrane by MCT8. This MCT8 deficiency leads to a lack of T3 in tissues that need T3 to function properly contributing to an accumulation of T3 in the blood serum. The other thyroid hormone thyroxine (“T4”) usually remains at normal serum levels in ARDS patients but may also be slightly reduced from a normal level. Thyroid stimulating hormone (“TSH”) is normal to slightly elevated in ARDS patients.
The thyroid hormone analogue, diiodothyropropionic acid (DITPA), given to mice deficient in MCT8 had equal suppressive effect on thyroid stimulating hormone (TSH) as that observed in normal mice3. This suggested that DITPA can enter cells in the absence of MCT8. Administration of DITPA to 4 MCT8 deficient children, on compassionate basis, showed reduction and normalization of the high blood T3, thus ameliorating the hypermetabolism and nutrition but not the neuropsychomotor deficit4.
Introduction of the normal human MCT8 into cells by the means of the viral vector AAV9 to newborn Mct8 deficient mice was able to increase T3 in their brain and induce a T3-mediated effect5. However, these mice had no neurological deficit owing to an alternative transporter. Recently the same gene therapy was given to peripubertal Mct8 deficient-dKO mice that have neurocognitive abnormalities. It corrected the neurological abnormalities, learning and recall abilities but not the high blood T3 causing the increased metabolism6.
Currently, no treatment for ARDS has been approved by the United States Food and Drug Administration. Clinical trials have been completed for the drug, triiodothyroacetic acid (“TRIAC”), for use in the treatment of ARDS. However, TRIAC shares a close structural similarity to T3, which makes it difficult to accurately assess T3 serum levels. Further, TRIAC has been shown to significantly reduce T4 serum levels. See, Groeneweg et al. Lancet Diabetes Endocrinol. 2019 September; 7(9); 695-706.
A combined gene and DITPA treatment should correct both neuropsychomotor and metabolic defects that each treatment alone could not achieve, providing full rescue of the genetic defect.
Allan-Herndon-Dudley Syndrome (“ARDS”) is an X-linked recessive developmental disorder causing intellectual disability and movement issues in males. Specifically, patients with ARDS have a mutant SLC16A2 gene resulting in a malformed monocarboxylate transporter 8 (“MCT8”) protein. Symptoms of ARDS are caused by a lack of cellular uptake of the thyroid hormone triiodothyronine (“T3”), which is normally transported across the cell membrane by MCT8. This MCT8 deficiency leads to a lack of T3 in tissues that need T3 to function properly contributing to an accumulation of T3 in the blood serum. The other thyroid hormone thyroxine (“T4”) usually remains at normal serum levels in ARDS patients but may also be slightly reduced from a normal level. Thyroid stimulating hormone (“TSH”) is normal to slightly elevated in ARDS patients.
3,5-diiodothyropropionic acid (“DITPA”) is another thyroid hormone analog that has been studied for treatment of ARDS. However, as mentioned above, DITPA has not yet been approved for use in the treatment of ARDS. This lack of approval may be due to a lack of effective dosing regimens, stable and effective compositions, and extensive pharmacological assessments. While WO/2012/171065, published Dec. 20, 2012, attempts to establish DITPA dosing regimens for ARDS patients, this publication offers only theoretical examples.
Administration of DITPA to 4 MCT8 deficient children, on compassionate basis, showed reduction and normalization of the high blood T3, thus ameliorating the hypermetabolism and nutrition but not the neuropsychomotor deficit4.
Introduction of the normal human MCT8 into cells by the means of the viral vector AAV9 to newborn Mct8 deficient mice was able to increase T3 in their brain and induce a T3-mediated effect5. However, these mice had no neurological deficit owing to an alternative transporter. Recently the same gene therapy was given to peripubertal Mct8 deficient-dKO mice that have neurocognitive abnormalities. It corrected the neurological abnormalities, learning and recall abilities but not the high blood T3 causing the increased metabolism6.
A combined gene and DITPA treatment should correct both neuropsychomotor and metabolic defects that each treatment alone could not achieve, providing full rescue of the genetic defect. Thus, there is a need for gene therapy, and especially gene therapy combined with dosing regimens of DITPA, that are effective at treating AHDS and symptoms of ARDS.
The present subject matter is directed to methods of gene therapy for treating a subject with Allan-Herndon-Dudley syndrome, and to methods of combining such gene therapy with administration of 3,5-diiodothyropropionic acid (DITPA) for treating a subject with Allan-Herndon-Dudley syndrome.
Background: Allan-Herndon-Dudley syndrome (AHDS) is a severe psychomotor disability disorder that also manifests characteristic abnormal thyroid hormone (TH) levels. AHDS is caused by inactivating mutations in monocarboxylate transporter 8 (MCT8), a specific TH plasma membrane transporter widely expressed in the central nervous system (CNS). MCT8 mutations cause impaired transport of TH across brain barriers, leading to insufficient neural TH supply. There is currently no successful therapy for the neurological symptoms. Earlier work has shown that intravenous, but not intracerebroventricular adeno-associated virus serotype 9 (AAV9)-based gene therapy given to newborn Mct8 knockout (Mct8−/y) male mice increased T3 brain content and partially rescued TH-dependent gene expression, suggesting a promising approach to treat this neurological disorder.
Methods: The potential of intravenous delivery of AAV9 carrying human MCT8 was tested in the well-established Mct8−/y/Organic anion-transporting polypeptide 1c1 (Oatp1c1)−/− double knockout (dKO) mouse model of ARDS, which unlike MCT8−/y mice, displays both neurological and TH phenotype. Further, as the condition is usually diagnosed during childhood, treatment was given intravenously to P30 mice and psychomotor tests were carried out blindly at P120-P140 after which tissues were collected and analyzed.
Results: Systemic intravenous delivery of AAV9-MCT8 at a juvenile stage led to improved locomotor and cognitive function at P120-P140, which was accompanied by a near normalization of T3 content and an increased response of positively regulated TH-dependent gene expression in different brain regions examined (thalamus, hippocampus, and parietal cortex). Effects on serum TH concentrations and peripheral tissues were less pronounced, showing only improvement in the serum T3/rT3 ratio and in liver deiodinase 1 expression.
Conclusion: Intravenous administration of AAV9, carrying the human MCT8, to juvenile dKO mice manifesting ARDS has long-term beneficial effect predominantly on the CNS. This preclinical study indicates that this gene therapy ameliorates the devastating neurological symptoms in patients with ARDS.
In one embodiment, the present technology is directed to methods of using gene therapy to treat and correct neurological abnormalities associated with ARDS in a subject in need of such treatment, wherein the gene therapy comprises administration of AAV9-MCT8 to the subject, wherein the administration preferably is intravenous.
Thyroid hormones (THs) are essential for the development and metabolic homeostasis of most organs and tissues (1). The major form of TH released in the blood from the thyroid gland is thyroxine (T4), which acts as a prohormone. T4 conversion to the active hormone, triiodothyronine (T3), or to the inactive form, reverse T3 (rT3), takes place intracellularly by iodothyronine deiodinases enzymes (2). The main mechanism of T3 action is achieved through binding to specific nuclear receptors, which in turn operate as regulators of gene transcription (3). Since TH metabolism and action are intracellular events, they require the presence of TH specific transporters mediating cellular TH uptake and efflux (4). The solute carrier family 16, member 2 (SLC16A2) gene, located on the X-chromosome, encodes for the monocarboxylate transporter 8 (MCT8) protein (5). MCT8 is well conserved throughout vertebrate evolution and is widely expressed in the body and central nervous system (CNS) (6). A key function of MCT8 is to facilitate TH transport across plasma membranes (5).
Inactivating MCT8 gene mutations in males cause a severe form of psychomotor disability (7-9), clinically described by Allan Herndon Dudley Syndrome (ARDS) (10). Patients exhibit neurological impairments including severe intellectual disability, truncal hypotonia, dystonia and movement disorders. MCT8-deficiency also causes a TH phenotype, including elevated serum T3 levels, low rT3 and T4 with normal or slightly elevated thyroid stimulating hormone (TSH) resulting in markedly elevated free T3/T4 and T3/rT3 ratios (11).
Two independently generated Mct8-KO mouse models (12, 13) closely recapitulate the TH phenotype observed in patients with ARDS, but do not display expected neurological or behavioral phenotypes. This is due to a milder TH deprivation in mouse brains owing to a T4-specific transporter not present in the human blood brain barrier (BBB). Specifically, the Organic anion-transporting polypeptide 1c1 (Oatp1c1), encoded by the slco1c1 gene, was identified in mice, but not human, brain capillaries (14-16). Double knockout mct8−/y; oatp1c1−/− mice (dKO) display disease-relevant phenotypes including an impaired TH transport into the CNS and consequently a significantly decreased number of cortical parvalbumin-positive GABAergic interneurons, reduced myelination and pronounced locomotor abnormalities (17). These results indicate that in mice Mct8 (together with Oatp1c1) plays a crucial role in the transport of THs into the CNS and, importantly, provides a robust disease model for human MCT8-deficiency (18, 19). To progress from an animal model to a human-based model, induced pluripotent stem cells (iPSCs) were derived from ARDS patients and differentiated into brain microvascular endothelial-cells, which showed MCT8-dependent transport of THs across the human BBB (16, 20). However, MCT8 is not restricted to the brain endothelium, and it also affects TH transport across neural cell plasma membranes (21).
Gene therapy offers a promising approach to treat monogenic disorders. Spinal muscular atrophy type 1 patients, carrying deleterious mutations in the SMN1 gene, were treated by a single intravenous infusion of adeno-associated virus serotype 9 (AAV9) containing DNA coding for SMN, resulting in improved survival, as well as achievement of motor milestones and motor functions (22, 23), with subsequent FDA approval of the ZOLGENSMA gene therapy product. Additional examples for beneficial gene therapy approach for monogenic disorders have been reported recently (24-26).
While intracerebroventricular (ICV)-delivery directly targets the brain ventricles, thereby circumventing the blood brain barrier (BBB), intravenous (IV)-delivery offers systemic delivery, transducing primarily tissues outside the CNS including blood vessels. Importantly, it has been shown that AAV9 IV-delivery can cross the BBB and efficiently infect CNS cells (27). In a recent proof of concept study, an AAV9-MCT8 construct was delivered by ICV or IV injections into neonatal Mct8-KO (MCT8−/y) mice (28), with an increase in brain TH signaling upon IV, but not ICV, delivery. However, since the Mct8-KO mice do not display neurological impairments, it is unclear whether this approach results in rescue of the neurological symptoms.
Here, we tested the potential of IV delivery of AAV9-MCT8 in dKO mice. We chose to treat juvenile male mice at postnatal day 30 (P30) and tested the potential rescue of neurological and behavioral parameters at adulthood. This approach was espoused as the diagnosis of MCT8 deficiency is usually made in childhood.
Materials and Methods
All procedures were approved by Cedars-Sinai Medical Center's Institutional Animal Care and Use Committee (IACUC #009128). mct8−/−/oatp1c1−/− females and mct8−/y/oatp1c1−/− males with C57BL/6 background were paired to generate dKO pups. WT C57BL/6 were used as controls. Only males were selected for all treatments. AAV9-MCT8 was administered to P30 juvenile dKO mice and controls by tail vein injection containing 50×1010 viral particles vp/g in a volume of 20 μl/g. Behavioral as well as biochemical and molecular measurements were all performed on tissues and serum at P120-P140 and analyzed double blinded without the knowledge to which group the mice belonged. Mice identities were blinded by the person administering the AAAV9-MCT8 and were unknown to the technicians while performing the behavioral assays, dissection, tissue collection and biochemical analysis. This was unblinded when results were assembled and for the statistical analysis. Animals were subjected to behavioral analysis before being sacrificed for tissue collection. Thus, results from the same mice are presented in all figures.
Statistical Analysis
All scatter plots were first tested for their normal distribution using Kolmogorov-Smirnov test, with the Dallal-Wilkinson-Lillie for corrected P value. Data sets that were normally distributed were tested using one-way ANOVA with Tukey's post-hoc test for multiple comparisons. Data sets that were not normally distributed were compared using Mann-Whitney non-parametric test for independent samples. Behavior data collected over time including the rotarod test and Barnes maze were analyzed with mixed model regression with random intercept and the fixed-factors of time (as a continuous variable), treatment group, and the interaction term of group with time. To compare learning curves between groups, the β coefficients were compared and differences were considered significant at the step-down Bonferroni-corrected alpha level of <0.05. Residuals were inspected to confirm the fit of the modeling. Data analysis was performed with GraphPad Prism v8.0.0 and SAS Enterprise Guide v8.2. All data are presented as mean±SEM. p<0.05 was considered statistically significant. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Results
IV delivery of AAV9-MCT8 at P30 improves locomotor performance of dKO mice.
Given that patients with AHDS are often diagnosed during childhood, treatment feasibility should optimally be tested at a juvenile stage. Previous experience (28) indicated that IV delivery is more feasible compared to ICV delivery, as IV is a simpler route and would target not only the brain but other body regions that express MCT8 such as the liver. Therefore, we tested IV delivery at the previously tested dose of 50×1010 vp/g of AAV9-MCT8 (28) to peri-pubertal P30 dKO mice (
In order to confirm that the viral delivery resulted in expression of human MCT8, liver and brain were collected at P140. As expected, human MCT8 mRNA expression was not detected in wild type (WT) or dKO untreated mice (data not shown). MCT8 expression was observed in the liver and various brain regions of AAV9-MCT8 IV-treated dKO mice, with the highest levels seen in the liver. Pituitary and choroid plexus had higher MCT8 levels compared to the BBB-protected regions of the thalamus, parietal cortex, and hippocampus (
Mice underwent various behavioral analyses to assess locomotor function, which were performed at P120, when differences between WT and dKO were previously reported (29). Assessment by a rotarod test showed that untreated dKO mice had an overall reduced latency to fall and decreased learning curve compared to the WT group (
IV delivery of AAV9-MCT8 at P30 improves cognitive performance in dKO mice.
Learning, spatial memory and memory recall were assessed using a Barnes maze (30). During the 4-day training period, treatment did not significantly improve the learning curve (
A spontaneous alternation maze (Y-maze) test was used to further examine spatial memory. Untreated dKO mice demonstrated a significant decrease in the percent of spontaneous alterations compared to untreated WT mice, which was not significantly improved in the AAV9-MCT8-treated dKO mice (
IV delivery of AAV9-MCT8 at P30 partially restores brain T3-content in dKO mice.
In order to assess effects on pathophysiology, the serum, liver and brain were collected at P140 (
IV delivery of AAV9-MCT8 at P30 corrects T3-inducible gene expression.
To assess the T3-effect in the different brain regions, we next studied T3-inducible gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). These genes were selected based on their known response to T3 (31). Hairless (Hr) levels were significantly improved in the thalamus (
IV delivery of AAV9-MCT8 at P30 partially rescues phenotypes in the liver and minimally in serum of dKO mice.
The significantly higher level of MCT8 expression in the liver (
Patients with AHDS have abnormal serum TH levels, including elevated T3, low rT3 and T4 with normal or slightly elevated TSH, resulting in low T3/T4 and T3/rT3 ratios. The increased liver deiodinase 1 enzymatic activity is one of the mechanisms responsible for these serum thyroid tests and a decrease in its expression is needed to ameliorate this phenotype (33). In order to test the effect of IV delivery of AAV9-MCT8 at P30 on the serum TH phenotype in dKO mice, blood was collected from P140 mice and serum TSH and TH levels were quantified. Serum levels of TSH, T4, T3, and rT3 as well as the T3/rT3 and the T3/T4 ratios were all significantly altered in dKO mice compared to their WT littermates (
In this study, we tested the potential of IV delivery of AAV9-MCT8 to juvenile dKO mice. Our analysis shows long-term expression of human MCT8 within the CNS and in peripheral tissue, suggesting that AAV9 can efficiently transduce cells with MCT8 in peripheral tissue and within the CNS. Re-expression of MCT8 resulted in improved locomotor and cognitive behavior as well as substantial rescue of T3 content and associated gene expression in different areas of the brain.
MCT8-deficient patients suffer from a severe neuro-psychomotor phenotype and TH excess in peripheral tissues. Thus, an effective therapeutic strategy should account for deficient transport of THs across brain barriers and neural plasma membranes as well as the excess of TH in peripheral tissues. Being a rare disorder, ARDS is often misdiagnosed resulting in later identification of the disease. Moreover, there are currently about 300 diagnosed cases, which mostly involve older children (11, 34). Thus, it is important to develop therapeutics that are effective at juvenile ages. We therefore tested the effect of tail vein IV delivery at P30, which is peri-pubertal when pathophysiological symptoms are apparent in both dKO mice and patients (17, 35, 36).
Endogenously, MCT8 is ubiquitously expressed and is prominently localized in the thyroid, liver, kidneys and CNS (6, 12, 13). In the current study, we showed that IV-administration of AAV9-MCT8 to P30 dKO mice led to long-term MCT8 expression in the CNS and liver.
In the brain, MCT8 expression was observed in various regions, confirming the ability of AAV9 vectors to cross brain barriers and efficiently transduce brain cells during this period of development (27). Interestingly, higher expression was observed in brain regions that are not protected by the BBB such as the choroid plexus and pituitary (37, 38) compared to the thalamus, hippocampus and cortex. Strikingly, in the current study, this long-term brain expression resulted in a nearly complete normalization of T3 brain concentrations and associated gene expressions in the thalamus, hippocampus and parietal cortex. MCT8-deficiency was previously suggested to be caused both, by reduced transport of T3 across brain barriers and across neural cell membranes (16, 21, 29, 39). These results therefore suggest that a rescue of the T3 brain content was achieved. However, future work is required to distinguish whether the improved brain content is caused by MCT8 expression in brain blood vessels or the choroid plexus, which can both serve as gateways to the brain.
The improved brain content was accompanied by improved performance in the rotarod test and in gait analyses. While it remains unclear why other locomotor functions did not change significantly, the observed improvements can be attributed to ameliorations in psychomotor functions. Treated animals also showed an improvement in the learning curve using the Rotarod test, suggesting that treatment may have beneficial effects on cognitive and motor functions. Exploratory behavior, learning and memory are thought to originate in the hippocampus (40). Notably, the mild rescue of hippocampus-dependent learning and memory in the Barnes maze test in response to treatment was observed to be correlated with the significant increase of T3 levels and T3-induced gene expression in the hippocampus.
MCT8 expression was significantly higher in the liver than in the brain. However, no significant rescue was observed in T3 levels in the liver. Nevertheless, a reduction was observed in liver Diol expression, a TH-regulated enzyme that generates T3 from T4 and is responsible in part for the T3 excess in serum (33). This effect on Diol expression resulted in a partial amelioration in serum T3/rT3 ratio, while other parameters were not significantly improved. These results suggest a mild beneficial effect of systemic (IV) delivery of MCT8 on the peripheral tissues. Additional mechanisms contribute to the characteristic serum thyroid tests of AHDS, including decreased thyroidal secretion and altered negative feedback to the hypothalamus and pituitary, and thus have different TH availability (41-43). Thus, to augment the partial rescue, additional TH-normalizing treatments should be considered in conjunction with gene therapy.
There are several current treatment strategies that focus on TH analogs. These thyromimetic compounds are required to activate TH-induced transcriptional pathways via thyroid nuclear receptors, and need to penetrate plasma membranes independent of MCT8. In animal models, the TH analogs DITPA (44, 45), TRIAC (18), TETRAC (19) and Sobetirome (46) were able to restore some of the peripheral and central abnormalities; however, their effects on neurological symptoms remained limited or unknown due to the use of mice that were not neurologically affected. In patients with ARDS, DITPA (47) and TRIAC (48) reduced the high serum T3 concentration, however, there was no evidence for improvement in neurological symptoms. Thus, TH analogs can be considered to be used in combination with gene therapy.
Chemical and pharmaceutical chaperons have also been suggested as an alternative approach. These chaperons can restore the ability of some MCT8 gene mutations to transport TH across plasma membranes in animal models (49, 50); however, their clinical effect has not been assessed to date. Moreover, the use of chemical chaperons is limited only to a handful of missense mutations, and is therefore not applicable to the majority of patients.
Gene therapy, which emerged as a promising approach to treat monogenic developmental neurological disorders (23, 25, 26, 51-53), can overcome these limitations and potentially target all mutations and patients. Moreover, restoration of a functional MCT8 has the potential to resolve TH transport, as well as unidentified MCT8 roles, such as the transport of additional potential substrates.
Although this study shows a promising therapeutic direction, it has limitations. While the dKO mice provide a useful model for ARDS, the symptoms are less severe than in patients. Furthermore, additional disease relevant features such as lack of language or the predisposition to death could not be tackled in this study due to limitations of the model.
Overall, this study shows that IV administration of AAV9-MCT8 to dKO mice provides substantial rescue of molecular and biochemical parameters in the brain, as well as amelioration of the TH excess effect in peripheral tissues. In addition, this treatment improves locomotor and behavioral performance. These findings support future clinical examination of AAV-based MCT8 gene therapy in patients with ARDS.
The Applicant has discovered dosing timings of 3,5-diiodothyropropionic acid (DITPA) that are surprisingly effective for the treatment of Allan-Herndon-Dudley Syndrome (ARDS).
In one embodiment, the present technology is directed to methods of treating ARDS comprising administering DITPA orally twice daily to a pregnant mother who elects to retain an affected male embryo, or for a subject with MCT8 deficiency in need thereof to reduce and normalize high blood T3 ameliorate the hypermetabolism and nutrition given orally three times daily. Wherein administration to a pregnant woman begins no later than 11 weeks after conception of the subject, but preferably between 8 and 10 weeks after conception. The dose of DITPA for pregnant women to treat their affected male fetuses is 0.5±0.2 mg per day and postpartum for affected infants and children of any age, 1.5±0.5 mg per day.
In another embodiment, the present technology is directed to methods of treating Allan-Herndon-Dudley syndrome comprising the following steps:
In a preferred embodiment, the first dosage is about 1 milligram per kilogram of body weight of the subject per day (“mg/kg/day”).
In another preferred embodiment, the second dosage is about 2 mg/kg/day.
In another preferred embodiment, the third dosage is about 2.5 mg/kg/day.
In another preferred embodiment, the fourth dosage is about 1.5 mg/kg/day.
As used herein the term “too high” refers to a T3 serum level that is more than about 15% over T3 serum levels considered normal for the age of the subject.
As used herein the term “too low” refers to a T3 serum level that is more than about 15% under T3 serum levels considered normal for the age of the subject.
As used herein “normal” T3 serum levels by age of the subject is based on levels disclosed in Lem et al., Serum thyroid hormone levels in healthy children from birth to adulthood and in short children born small for gestational age, J Clin Endocrinol Metab, 2012 September, 97(9), 3170-8, doi: 10.1210/jc.2012-1759, Epub 2012 Jun. 26.
In another embodiment, the present disclosure is directed to methods of treating ARDS comprising the following steps:
In a preferred embodiment, the daily dosage of DITPA is administered to a subject in need thereof once a day, more preferably the daily dosage of DITPA is divided in two parts and each part is administered every 12 hours and most preferably the daily dosage of DITPA is divided into three parts and each part is administered every 8 hours.
In a preferred embodiment, administration of DITPA occurs via the oral route.
In one embodiment, DITPA may be formulated in a composition comprising DITPA, or a salt thereof, and one or more pharmaceutically acceptable excipients.
In a preferred embodiment, DITPA, or a salt thereof, may present in the pharmaceutical compositions of the present subject matter at a concentration from about 0.001% to about 10% w/w or w/v.
In a preferred embodiment, the one or more pharmaceutically acceptable excipients may be present in the pharmaceutical compositions of the present disclosure at a concentration from about 90% to about 99.999% w/w or w/v.
Pharmaceutically acceptable excipients suitable for use in the present subject matter include, but are not limited to, disintegrants, binders, fillers, plasticizers, lubricants, permeation enhancers, surfactants, sweeteners, sweetness enhancers, flavoring agents and pH adjusting agents.
The term “disintegrants” as used herein refers to pharmaceutically acceptable excipients that facilitate the disintegration of the tablet once the tablet contacts water or other liquids. Disintegrants suitable for use in the present technology include, but are not limited to, natural starches, such as maize starch, potato starch etc., directly compressible starches such as starch 1500, modified starches such as carboxymethyl starches, sodium hydroxymethyl starches and sodium starch glycolate and starch derivatives such as amylose, cross-linked polyvinylpyrrolidones such as crospovidones, modified celluloses such as cross-linked sodium carboxymethyl celluloses, sodium hydroxymethyl cellulose, calcium hydroxymethyl cellulose, croscarmellose sodium, low-substituted hydroxypropyl cellulose, alginic acid, sodium alginate, microcrystalline cellulose, methacrylic acid-divinylbenzene copolymer salts and combinations thereof.
Binders suitable for use in the present technology include, but are not limited to, polyethylene glycols, soluble hydroxyalkyl celluloses, polyvinylpyrrolidone, gelatins, natural gums and combinations thereof.
Fillers suitable for use in the present technology include, but are not limited to, dibasic calcium phosphate, calcium phosphate tribasic, calcium hydrogen phosphate anhydrous, calcium sulfate and dicalcium sulfate, lactose, sucrose, amylose, dextrose, mannitol, inositol and combinations thereof.
Plasticizers suitable for use in the present subject matter include, but are not limited to, microcrystalline cellulose, triethyl citrate, poly-hexanediol, acetylated monoglyceride, glyceryl triacetate, castor oil, and combinations thereof.
Lubricants suitable for use in the present technology include, but are not limited to, magnesium stearate, sodium stearyl fumarate, stearic acid, glyceryl behenate, micronized polyoxyethylene glycol, talc, silica colloidal anhydrous and combinations thereof.
Permeation enhancers suitable for use in the present subject matter include, but are not limited to, precipitated silicas, maltodextrins, β-cyclodextrins menthol, limonene, carvone, methyl chitosan, polysorbates, sodium lauryl sulfate, glyceryl oleate, caproic acid, enanthic acid, pelargonic acid, capric acid, undecylenic acid, lauric acid, myristic acid, palmitic acid, oleic acid, stearic acid, linolenic acid, arachidonic acid, benzethonium chloride, benzethonium bromide, benzalkonium chloride, cetylpyridium chloride, edetate disodium dihydrate, sodium desoxycholate, sodium deoxyglycolate, sodium glycocholate, sodium caprate, sodium taurocholate, sodium hydroxybenzoyal amino caprylate, dodecyl dimethyl aminopropionate, L-lysine, glycerol oleate, glyceryl monostearate, citric acid, peppermint oil and combinations thereof.
Surfactants suitable for use in the present subject matter include, but are not limited to, sorbitan esters, docusate sodium, sodium lauryl sulphate, cetriride and combinations thereof.
Sweeteners suitable for use in the present technology include, but are not limited to, aspartame, saccharine, potassium acesulfame, sodium saccharinate, neohesperidin dihydrochalcone, sucralose, sucrose, dextrose, mannitol, glycerin, xylitol and combinations thereof.
Sweetness enhancers suitable for use in the present technology include, but are not limited to, ammonium salt forms of crude and refined glycyrrhizic acid.
Flavoring agents suitable for use in the present subject matter include, but are not limited to, peppermint oil, menthol, spearmint oil, citrus oil, cinnamon oil, strawberry flavor, cherry flavor, raspberry flavor, orange oil, tutti frutti flavor and combinations thereof.
pH adjusting agents suitable for use in the present formulation include, but are not limited to, hydrochloric acid, citric acid, fumaric acid, lactic acid, sodium hydroxide, sodium citrate, sodium bicarbonate, sodium carbonate, ammonium carbonate, sodium acetate and combinations thereof.
In another preferred embodiment, the pharmaceutical compositions of the present technology do not contain a preservative.
Pharmaceutical compositions of the present technology may be formulated in any dosage form including but not limited to aerosol including metered, powder and spray, chewable bar, bead, capsule including coated, film coated, gel coated, liquid filled and coated pellets, cellular sheet, chewable gel, concentrate, elixir, emulsion, film including soluble, film for solution and film for suspension, gel including metered gel, globule, granule including granule for solution, granule for suspension, chewing gum, inhalant, injectable including foam, liposomal, emulsion, lipid complex, powder, lyophilized powder and liposomal suspension, liquid, lozenge, ointment, patch, electrically controlled patch, pellet, implantable pellet, pill, powder, powder, metered powder, solution, metered solution, solution concentrate, gel forming solution/solution drops, spray, metered spray, suspension, suspension, syrup, tablet, chewable tablet, coated tablet, coated particles in a tablet, film coated tablet, tablet for solution, tablet for suspension, orally disintegrating tablet, soluble tablet, sugar coated tablet, dispersible tablet, tablet with sensor, tape, troche and wafer and extended release and delayed release forms thereof.
In a preferred embodiment, the pharmaceutical compositions of the present technology are in tablet form. In a more preferred embodiment, the pharmaceutical compositions of the present formulation are in a dispersible tablet form. In an even more preferred embodiment, the pharmaceutical compositions of the present formulation are in a water-dispersible tablet form. In a most preferred embodiment, the pharmaceutical compositions of the present formulation are in a water-dispersible tablet form wherein the tablet is scored such that the tablet is dividable into four equal parts.
In a preferred embodiment, when the pharmaceutical compositions of the present technology are in a water-dispersible tablet form the tablet dispersion time is about 70 seconds or less, more preferably about 60 seconds or less and even more preferably about 40 seconds or less, even more preferably about 30 seconds or less, even more preferably about 20 seconds or less, even more preferably about 10 seconds or less and even more preferably about 5 seconds or less.
As used herein the term “pharmaceutically acceptable” refers to ingredients that are not biologically or otherwise undesirable in an oral application.
As used herein, all numerical values relating to amounts, weights, and the like, are defined as “about” each particular value, that is, plus or minus 10%. For example, the phrase “10% w/w” is to be understood as “9% to 11% w/w.” Therefore, amounts within 10% of the claimed value are encompassed by the scope of the claims.
As used herein “% w/w” refers to the weight percent by weight of the total formulation.
As used herein “% w/v” refers to the weight percent by volume of the total formulation.
As used herein the term “effective amount” refers to the amount necessary to treat a subject in need thereof.
As used herein the term “treatment” or “treating” refers to alleviating or ameliorating ARDS or symptoms of ARDS.
As used herein, the term “stable” includes, but is not limited to, physical and chemical stability.
Pharmaceutically acceptable salts of that can be used in accordance with the current subject matter include but are not limited to hydrochloride, dihydrate hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, mesylate, maleate, gentisinate, fumarate, tannate, sulphate, tosylate, esylate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzensulfonate, p-toluenesulfonate and pamoate (i.e., 1,1′-methylene-bis-(2-hydroxy-3-naphthoate)) salts.
Throughout the application, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise.
The disclosed embodiments are simply exemplary embodiments of the inventive concepts disclosed herein and should not be considered as limiting unless the claims expressly state otherwise.
Administration of DITPA may be used to reduce and normalize high blood T3, ameliorating the hypermetabolism and nutrition but not the neuropsychomotor deficit associated with ARDS. Use of gene therapy helps correct the neurological abnormalities, learning and recall abilities, but not the high blood T3 causing the increased metabolism. A combination of DITPA treatment and gene therapy corrects both neuropsychomotor and metabolic defects that each treatment alone could not achieve, providing full rescue of the genetic defect.
In one embodiment, the present technology is directed to methods of using a combination of DITPA administration to a subject, along with gene therapy, to treat and correct neurological and metabolic abnormalities associated with ARDS in a subject in need of such treatment, wherein the gene therapy comprises administration of AAV9-MCT8 to the subject, and wherein the DITPA is administered daily, to reduce and normalize high blood T3 and ameliorate the hypermetabolism and nutrition in the subject, and preferably where the AAV9-MCT8 is administered intravenously to the subject.
The following examples are intended to illustrate the present technology and to teach one of ordinary skill in the art how to use the formulations of this new technology. They are not intended to be limiting in any way.
DITPA was administered to a pregnant mother of a male pre-natal subject that had previously tested positive for the SLC16A2 allele correlated with Allan-Herndon-Dudley syndrome at a daily dosage of 1 mg/kg/day divided over three administration spaced 8 hours apart starting at 4 weeks after conception and ending at birth of the subject.
The dosing regimen successfully reduced symptoms of ARDS in the newborn subject as compared to affected newborns whose mothers were not treated with DITPA.
DITPA reduces the activity of deiodinase-1 in vivo and in vitro in liver (see figure shown Dose response of DITPA added to liver in vitro and measurement of D1 enzymatic activity, i.e., conversion of T4 to T3). This is the main mechanism of reduction in T3 and increase in T4 by reducing its consumption. It was shown to occur in humans with MCT8 deficiency.
These are the consequences of the normalization of serum T3 levels, a critical endocrine biomarker and parameter for therapeutic efficacy, as they measure the anticipated metabolic changes resulting from the normalization of the thyroid tests.
More specifically, the reduction of T3, which acts on peripheral tissue to accelerate the metabolism, is expected to improve nutrition and increase the ability to gain weight.
Important measurements such as weight gain (corrected for age) and metabolic parameters (cholesterol, creatine kinase, SHBG) are secondary endpoints.
Annotated observations by the parents such as sleep, food record, motor activity are of immense value.
Dose response of DITPA added to liver in vitro and measurement of D1 enzymatic activity (conversion of T4 to T3)
Research centered on regulation of gene expression and gene therapy, with three aims.
We recently showed that gene therapy in peripubertal dKO mice with adeno associated virus 9 (AAV9) containing the human MCT8 cDNA improved the locomotor and cognitive function by near normalization of brain T3 content but failed to correct the serum thyroid tests. Adding the TH analogues diiodothyropropionic acid (DITPA) or triiodothyroacetic acid (TRIAC), known to correct the thyrotoxicosis of ARDS in peripheral tissues but not the neuropsychomotor manifestations, is intended to achieve rescue of this incapacitating disease.
Rationale & Impact: Deficiency of the X-linked MCT8 results in a complex phenotype in boys that includes a severe neuropsychomotor defect, caused by deficiency of TH transport in the brain, and systemic thyrotoxicosis caused by excess of circulating T3. Treatment with the TH analogues diiodothyropropionic acid (DITPA) or triiodothyroacetic acid (TRIAC) corrects the thyrotoxicosis of peripheral tissues but not the neuropsychomotor defect. Double knockout (dKO) mice, lacking Mct8 and the TH transporter Oatp1c1, recapitulate the findings of ARDS. We recently showed that gene therapy in peripubertal dKO mice with adeno associated virus 9 (AAV9), containing the human MCT8 cDNA, improved the locomotor and cognitive function by near normalization of brain T3 content but failed to correct the serum TFTs. Combined treatment with AAV9 and TH analogues has the potential to rescue MCT8 deficiency and serve as a preclinical model for treatment in humans.
We Will:
In addition to locomotor function, the metabolic and cardiac effects will be assesses using metabolic cages.
We have discovered defects in the TH transmembrane transporter monocarboxylate 8 (MCT8)6 causing the severe neuro-psychomotor syndrome [Allan-Herndon-Dudley syndrome (AHDS)] and defects in TH metabolism, SBP27 and DIO18.
We investigate highly significant areas, namely (i) expand the study of newly identified genetic defects that affect thyroid physiology including the deiodinases (ii) develop a potentially effective treatment for MIDS. Preliminary data make this proposal both clinically and scientifically relevant, emphasizing its translational strength.
Another significant aspect is the development of a treatment for AHDS. Those who have seen a child affected by AIMS will readily understand the urgent need for an effective treatment. While the current focus is on the use of TH analogues that correct only the hypermetabolism, we recently developed a promising gene therapy strategy. Our compelling new data show that using adeno-associated virus 9 (AAV9) expressing human MCT8 cDNA ameliorates the neuro-psychomotor component of ARDS even when administered in peripubertal life15. Combined with TH analogues, gene therapy holds great promise for long-term improvement of the global phenotype of ARDS.
Treatment of patients with ARDS' has remained an important challenge in thyroidology. We propose a pioneering approach for the thyroid field using AAV9 transduction of normal human MCT8 cDNA to deliver functional hMCT8 at juvenile stage to a mouse model of ARDS (mice deficient in Mct8 and Oatp1c1) which partially rescues the neuromotor abnormality15, with concomitant use of TH analogue treatments to correct the hypermetabolism18,19. This combined approach targets the complex pathophysiology of the MCT8 deficiency with the potential to rescue or attenuate the severe phenotype of this defect and serve as a preclinical model for treatment in humans.
Since the discovery of the MCT8 deficiency6,24 resulting in the severe neurological manifestations due to impaired TH transport in the brain, and thyrotoxicosis in other tissues, multiple patients have been identified. However, treatment remains challenging. The use of TH analogues improved the hypermetabolic state but failed to improve the neuropsychomotor manifestations. We showed that intravenous (IV) administration of adeno-associated virus serotype 9 vector expressing the human MCT8 cDNA (AAV9-hMCT8) to 1-day old Mct8KO mice, populated their choroid plexus with the hMCT8 protein. Their brain T3 content increased, as well as expression of genes regulated by TH25. When injected to 30-day old Mct8 and Oatp1c1 deficient (dKO) mice, manifesting thyroid and neurological abnormalities, improvement was observed in locomotor performance, learning and recall ability15. This was accompanied by an increase in brain T3 content and TH-regulated gene expression15. However, little to no changes were observed in serum and peripheral tissues. We now propose a combination of gene therapy together with TH analogues to improve both hypermetabolism and the neuropsychomotor deficitt26.
Background: MCT8 deficiency manifests a syndrome with two components: severe neurodevelopmental delay with gait disturbance, dystonia, poor head control and mental retardation as well as a characteristic pattern of thyroid tests abnormalities including increased serum T3 and TSH, and decreased T4 and rT36,24. Previously described in 1944 by Allan, Herndon and Dudley60 as X-linked mental retardation in males, and now referred to as AHDS, the syndrome was subsequently found to be caused by mutations in the MCT8 gene61.
Mouse models of Mct8 deficiency replicate the thyroid tests abnormalities in humans62,63. However, they do not manifest neurological abnormalities owing to the expression in brain of Oatp1c1, another TH cell membrane transporter, present in very small amounts in human brain64. Indeed, a mouse deficient in both Mct8 and Oatp1, or double KO (dKO), manifests both thyroid and neuromotor defects65.
It is now clear that a selective brain TH deficiency is produced by severe reduction in hormone transport at the level of vascular endothelial cells or the blood brain barrier (BBB)66,67. Thus, MCT8 deficiency produces TH deficiency in brain leading to the neuromotor abnormalities, and TH excess from increased circulating T3 that reaches tissues by alternative transporters, causes cardiotoxicity and hypermetabolism. Thus, contrary to TH deficiency caused by congenital absence of a thyroid gland or due to a defect in TH synthesis, the combined selective tissue deficiency and excess of MCT8 deficiency cannot be treated with TH replacement68.
Devising a new treatment that influences both components of the defect is proposed in this aim. This will require a combination of treatments to improve both hypermetabolism and the neuro psychomotor deficit, with the goal of altering the natural history of the defect26 through improved mobility, reduction in complications, increased survival, and facilitating long term care.
Rationale: Studies in Mct8 deficient mice showed that the TH analogues diiodothyropropionic acid (DITPA) or triiodothyroacetic acid (TRIAC) enter cells independent of MCT869,70. Trials using these analogues in children with MCT8 deficiency reduce the high serum T3 levels and diminish the hypermetabolism, the heart rate and improve some parameters that reflect TH action on peripheral tissues. However, they produced no significant effect on the neuropsychomotor deficit19,71 with no documented change in neurodevelopment even with prolonged TRIAC use18.
We resorted to gene therapy using an adeno-associated virus serotype 9 (AAV9) vector shown to rescue spinal muscular atrophy72 and that is FDA approved. We showed that AAV9 carrying human MCT8 cDNA, when given IV to 1-day old Mct8 deficient mice, increased brain T3 content and expression of genes positively regulated by TH, through populating the choroid plexus with the hMCT8 protein25. When injected to 30-day old dKO mice that manifest thyroid and neurological abnormalities, improvement was observed in locomotor performance, learning and recall ability15. This was accompanied by an increase in T3 content and TH-regulated gene expression in all three brain areas examined: thalamus, hippocampus and parietal cortex in males (
However, little to no changes were observed in liver markers despite important expression of hMCT8 in this tissue, nor in serum TH concentration except for a minimal decrease in the T3/rT3 ratio15. These early results of gene therapy are very encouraging as a single treatment has a lasting effect (more than 100 days) on brain function in a mouse model with close resemblance to the AHDS in humans. More importantly, it is still effective when applied at P30. Thus, to further improve the potential gene therapy, we propose to combine it with a treatment that is able to reduce the serum T3 concentration.
Experimental Approach: The proposed protocol is schematically presented in
AAV9-hMCT8 containing 50×1010 viral particles/g will be injected in the tail vein of P30 dKO mice. WT mice will receive the same amount of empty viral vector (AAV9 without hMCT8). Three days later, groups of 10 animals will be treated with doses of DITPA and TRIAC that normalize the serum T3 concentration of dKO mice. Given by daily intraperitoneal injections, these are per 100 g BW, 0.3 mg DITPA and 6 μg TRIAC. (1) Baseline measurements of serum T4, T3, rT3, TSH, cholesterol, alkaline phosphatase (AP) and creatine kinase (CK) will be obtained a day before. The same tests will be repeated 10 days later (P40) and the doses of DITPA and TRIAC will be adjusted according to the serum levels of T3, while T4 will be kept at a normal level of 3.5-5.0 μg/dL for adult C57B1/6J male mice.
Another blood sample will be obtained at P60 and again at P120. (2) Behavioral and locomotor tests (rotarod, open field, gate analysis, Barnes maze, Y maze) will be carried out over a period of 20 days. During the same interval of time, and while not undergoing behavioral and locomotor tests, metabolic studies will be performed using indirect calorimetry in metabolic cages as previously carried out in the laboratory73, to assess the metabolic and cardiac effects of the combination treatment. Total energy expenditure (TEE), Respiratory Exchange Ratio (RER), total activity, food and water intake and heart rate will be measured. Physical activity will be continuously measured, as well as O2 uptake and CO2 production at 30 min intervals. RER, TEE, glucose, and lipid oxidation will be calculated from the 02 consumption (VO2) and CO2 production (VCO2) relative to body weight.
On P140, after collection of a terminal blood sample, animals will be perfused under anesthesia, prior to tissue collection. Tissues from two animals from each group will be used for immunohistochemistry and confocal microscopy to localize the hMCT8 protein. The following tissues will be collected and immediately frozen: whole brain, anterior pituitary, thyroid, a fragment of liver, kidney, heart, and muscle (gastrocnemius). Subsequently, the frozen brain will be dissected to recover frontal, parietal and occipital cortices, hippocampus, thalamus, hypothalamus, striatum, choroid plexus, and cerebellum. Tissues will be analyzed for T3, T4 and respective analogues (DITPA and TRIAC) content as well as expression of tissue specific TH-regulated genes and enzymatic activities of the three deiodinases.
The expression of the following genes will be measured by qPCR. In brain (Hr, Dio2, Dio3, Aldh1a1, Cbr2); in pituitary (Tshb, Gh, Trhr, Dio2); in thyroid (Tshr, Tg, Tpo, Duox2, Nis, Ttf1); in liver (Dio1, Usp2, Me1); in heart (Serca2, Myh6, Myh7), in muscle (Hr, Myh1, 2 and 7, Mct4, Pkm); Kidney (Dio1, NaPi2, Clc2). The effect on other transporters (Mct10, Lat1, Lat2) and the injected human MCT8 will be also measured. It should be noted that while some monoclonal antibodies can measure T3 without interference from DITPA74, this is not the case with TRIAC75. Thus, T3 together with DITPA and TRIAC, when appropriate, will be measured by liquid cromatography-tandem mass spectrometry (LC-MS/MS)45.
Based on this information the following effects are anticipated. (a) DITPA should normalize all three iodothyronines as observed in vivo19 by decreasing the D1 activity73, an effect demonstrated in vitro (
Different from DITPA, TRIAC increases D176 and adding L-T4 further increases the T371. However, the resulting reduction of serum T4 which acts as obligatory precursor of T3 in the brain77, may dampen the effect of the gene therapy. As all treatments have been used separately in the past, it is anticipated that gene therapy in combination with both DITPA and TRIAC will produce a combination of the effects observed with the individual treatments as described above. In addition, unbiased approaches such as RNAseq can be used to assess for transcriptional changes in tissues in the setting of combined treatment with AAV9 mediated gene therapy and TH analogue.
It is to be understood that the subject matter herein is not limited to the specific embodiments described above but encompasses any and all embodiments within the scope of the generic language of the following claims enabled by the embodiments described herein, or otherwise described above in terms sufficient to enable one of ordinary skill in the art to make and use the claimed subject matter.
This application claims the benefit of U.S. Provisional Patent Application No. 63/388,235, filed on Jul. 11, 2022, the content of which is incorporated herein by reference in its 10 entirety.
This invention was made with government support under Grant No. DK15070 awarded by the National Institutes of Health (NIH). The government has certain rights in the invention.
Number | Date | Country | |
---|---|---|---|
63388235 | Jul 2022 | US |