Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Nat Commun ; 15(1): 3804, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714648

ABSTRACT

Messenger RNA (mRNA) therapeutics delivered via lipid nanoparticles hold the potential to treat metabolic diseases caused by protein deficiency, including propionic acidemia (PA), methylmalonic acidemia (MMA), and phenylketonuria (PKU). Herein we report results from multiple independent preclinical studies of mRNA-3927 (an investigational treatment for PA), mRNA-3705 (an investigational treatment for MMA), and mRNA-3210 (an investigational treatment for PKU) in murine models of each disease. All 3 mRNA therapeutics exhibited pharmacokinetic/pharmacodynamic (PK/PD) responses in their respective murine model by driving mRNA, protein, and/or protein activity responses, as well as by decreasing levels of the relevant biomarker(s) when compared to control-treated animals. These preclinical data were then used to develop translational PK/PD models, which were scaled allometrically to humans to predict starting doses for first-in-human clinical studies for each disease. The predicted first-in-human doses for mRNA-3927, mRNA-3705, and mRNA-3210 were determined to be 0.3, 0.1, and 0.4 mg/kg, respectively.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Disease Models, Animal , Phenylketonurias , Propionic Acidemia , RNA, Messenger , Propionic Acidemia/genetics , Propionic Acidemia/therapy , Propionic Acidemia/drug therapy , Animals , Phenylketonurias/genetics , Phenylketonurias/drug therapy , Phenylketonurias/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/drug therapy , Mice , Humans , Male , Female , Nanoparticles/chemistry , Mice, Inbred C57BL , Liposomes
2.
Nat Commun ; 11(1): 5339, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33087718

ABSTRACT

Propionic acidemia/aciduria (PA) is an ultra-rare, life-threatening, inherited metabolic disorder caused by deficiency of the mitochondrial enzyme, propionyl-CoA carboxylase (PCC) composed of six alpha (PCCA) and six beta (PCCB) subunits. We herein report an enzyme replacement approach to treat PA using a combination of two messenger RNAs (mRNAs) (dual mRNAs) encoding both human PCCA (hPCCA) and PCCB (hPCCB) encapsulated in biodegradable lipid nanoparticles (LNPs) to produce functional PCC enzyme in liver. In patient fibroblasts, dual mRNAs encoded proteins localize in mitochondria and produce higher PCC enzyme activity vs. single (PCCA or PCCB) mRNA alone. In a hypomorphic murine model of PA, dual mRNAs normalize ammonia similarly to carglumic acid, a drug approved in Europe for the treatment of hyperammonemia due to PA. Dual mRNAs additionally restore functional PCC enzyme in liver and thus reduce primary disease-associated toxins in a dose-dependent manner in long-term 3- and 6-month repeat-dose studies in PA mice. Dual mRNAs are well-tolerated in these studies with no adverse findings. These studies demonstrate the potential of mRNA technology to chronically administer multiple mRNAs to produce large complex enzymes, with applicability to other genetic disorders.


Subject(s)
Enzyme Replacement Therapy/methods , Propionic Acidemia/therapy , RNA, Messenger/therapeutic use , Animals , Disease Models, Animal , Glutamates/therapeutic use , Humans , Kinetics , Lipids/chemistry , Liver/enzymology , Methylmalonyl-CoA Decarboxylase/chemistry , Methylmalonyl-CoA Decarboxylase/genetics , Methylmalonyl-CoA Decarboxylase/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/enzymology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Propionic Acidemia/genetics , Propionic Acidemia/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics
3.
Br J Pharmacol ; 177(14): 3168-3182, 2020 07.
Article in English | MEDLINE | ID: mdl-32133631

ABSTRACT

BACKGROUND AND PURPOSE: Acute intermittent porphyria (AIP) results from haplo-insufficiency of the porphobilinogen deaminase (PBGD) gene encoding the third enzyme in the haem biosynthesis pathway. As liver is the main organ of pathology for AIP, emerging therapies that restore enzyme hepatic levels are appealing. The objective of this work was to develop a mechanistic-based computational framework to describe the effects of novel PBGD mRNA therapy on the accumulation of neurotoxic haem precursors in small and large animal models. EXPERIMENTAL APPROACH: Liver PBGD activity data and/or 24-hr urinary haem precursors were obtained from genetic AIP mice and wild-type mice, rats, rabbits, and macaques. To mimic acute attacks, porphyrogenic drugs were administered over one or multiple challenges, and animals were used as controls or treated with different PBGD mRNA products. Available experimental data were sequentially used to build and validate a semi-mechanistic mathematical model using non-linear mixed-effects approach. KEY RESULTS: The developed framework accounts for the different biological processes involved (i.e., mRNA sequence, release from lipid nanoparticle and degradation, mRNA translation, increased PBGD activity in liver, and haem precursor metabolism) in a simplified mechanistic fashion. The model, validated using external data, shows robustness in the extrapolation of PBGD activity data in rat, rabbit, and non-human primate species. CONCLUSION AND IMPLICATIONS: This quantitative framework provides a valuable tool to compare PBGD mRNA drug products during early preclinical stages, optimize the amount of experimental data required, and project results to humans, thus supporting drug development and clinical dose and dosing regimen selection.


Subject(s)
Porphyria, Acute Intermittent , Animals , Heme , Hydroxymethylbilane Synthase/genetics , Mice , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/genetics , RNA, Messenger , Rabbits , Rats
4.
EBioMedicine ; 45: 519-528, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31303505

ABSTRACT

BACKGROUND: Isolated methylmalonic acidemia/aciduria (MMA) is an ultra-rare, serious, inherited metabolic disorder with significant morbidity and mortality. Exogenously delivered mRNA encoding human methylmalonyl-CoA mutase (hMUT), the enzyme most frequently mutated in MMA, is a potential therapy to produce functional MUT enzyme in liver. METHODS: Two 12-week repeat-dose studies were conducted to evaluate the efficacy and safety of intravenously-administered hMUT mRNA encapsulated in lipid nanoparticles in two murine models of MMA. FINDINGS: In MMA hypomorphic mice, hMUT mRNA treatment resulted in dose-dependent and reproducible biomarker responses after each dose. Enzymatically-active MUT protein was produced in liver in a dose-dependent manner. hMUT mRNA was well-tolerated with no adverse effects, as indicated by the lack of clinical observations, minimal changes in clinical chemistry parameters, and histopathology examination across all tissues. In severe MMA mice, hMUT mRNA led to substantially improved survival and growth and ameliorated biochemical abnormalities, all of which are cardinal clinical manifestations in severely affected patients. INTERPRETATION: These data demonstrate durable functional benefit of hMUT mRNA and support development of this new class of therapy for a devastating, pediatric disorder. FUND: This work was funded by Moderna, Inc.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Liver/metabolism , Methylmalonyl-CoA Mutase/pharmacology , RNA, Messenger/pharmacology , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Child , Disease Models, Animal , Humans , Lipids/genetics , Liver/drug effects , Liver/pathology , Methylmalonyl-CoA Mutase/genetics , Mice , RNA, Messenger/genetics
5.
Hum Gene Ther ; 30(10): 1180-1189, 2019 10.
Article in English | MEDLINE | ID: mdl-31179759

ABSTRACT

Exogenous delivery of messenger RNA (mRNA) is emerging as a new class of medicine with broad applicability including the potential to treat rare monogenic disorders. Recent advances in mRNA technology, including modifications to the mRNA itself along with improvements to the delivery vehicle, have transformed the utility of mRNA as a potential therapy to restore or replace different types of therapeutic proteins. Preclinical proof-of-concept has been demonstrated for mRNA therapy for three different rare metabolic disorders: methylmalonic acidemia, acute intermittent porphyria, and Fabry disease. Herein, we review those preclinical efficacy and safety studies in multiple animal models. For all three disorders, mRNA therapy restored functional protein to therapeutically relevant levels in target organs, led to sustained and reproducible pharmacology following each dose administration of mRNA, and was well tolerated as supported by liver function tests evaluated in animal models including nonhuman primates. These data provide compelling support for the clinical development of mRNA therapy as a treatment for various rare metabolic disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Fabry Disease/therapy , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Porphyria, Acute Intermittent/therapy , RNA, Messenger/genetics , Rare Diseases/therapy , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Clinical Trials as Topic , Disease Models, Animal , Fabry Disease/genetics , Fabry Disease/metabolism , Gene Transfer Techniques , Genetic Diseases, Inborn/metabolism , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Haplorhini , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/metabolism , Proof of Concept Study , RNA, Messenger/administration & dosage , RNA, Messenger/metabolism , Rare Diseases/genetics , Rare Diseases/metabolism , Rodentia
6.
Mol Ther ; 27(7): 1242-1251, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31056400

ABSTRACT

Citrin deficiency is an autosomal recessive disorder caused by loss-of-function mutations in SLC25A13, encoding the liver-specific mitochondrial aspartate/glutamate transporter. It has a broad spectrum of clinical phenotypes, including life-threatening neurological complications. Conventional protein replacement therapy is not an option for these patients because of drug delivery hurdles, and current gene therapy approaches (e.g., AAV) have been hampered by immunogenicity and genotoxicity. Although dietary approaches have shown some benefits in managing citrin deficiency, the only curative treatment option for these patients is liver transplantation, which is high-risk and associated with long-term complications because of chronic immunosuppression. To develop a new class of therapy for citrin deficiency, codon-optimized mRNA encoding human citrin (hCitrin) was encapsulated in lipid nanoparticles (LNPs). We demonstrate the efficacy of hCitrin-mRNA-LNP therapy in cultured human cells and in a murine model of citrin deficiency that resembles the human condition. Of note, intravenous (i.v.) administration of the hCitrin-mRNA resulted in a significant reduction in (1) hepatic citrulline and blood ammonia levels following oral sucrose challenge and (2) sucrose aversion, hallmarks of hCitrin deficiency. In conclusion, mRNA-LNP therapy could have a significant therapeutic effect on the treatment of citrin deficiency and other mitochondrial enzymopathies with limited treatment options.


Subject(s)
Citrullinemia/drug therapy , Citrullinemia/metabolism , Drug Delivery Systems/methods , Genetic Therapy/methods , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , RNA, Messenger/therapeutic use , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Gene Knockout Techniques , Glucosephosphate Dehydrogenase/genetics , HeLa Cells , Hep G2 Cells , Humans , Lipids/chemistry , Loss of Function Mutation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Nanoparticles/chemistry , Open Reading Frames/genetics , RNA, Messenger/chemical synthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transfection , Treatment Outcome
7.
Orphanet J Rare Dis ; 14(1): 84, 2019 04 25.
Article in English | MEDLINE | ID: mdl-31023387

ABSTRACT

Methylmalonic acidemia/aciduria (MMA) is a genetically heterogeneous group of inherited metabolic disorders biochemically characterized by the accumulation of methylmalonic acid. Isolated MMA is primarily caused by the deficiency of methylmalonyl-CoA mutase (MMA mut; EC 5.4.99.2). A systematic literature review and a meta-analysis were undertaken to assess and compile published epidemiological data on MMA with a focus on the MMA mut subtype (OMIM #251000). Of the 1114 identified records, 227 papers were assessed for eligibility in full text, 48 articles reported on disease epidemiology, and 39 articles were included into the quantitative synthesis. Implementation of newborn screening in various countries has allowed for the estimation of birth prevalence of MMA and its isolated form. Meta-analysis pooled point estimates of MMA (all types) detection rates were 0.79, 1.12, 1.22 and 6.04 per 100,000 newborns in Asia-Pacific, Europe, North America and the Middle East and North Africa (MENA) regions, respectively. The detection rate of isolated MMA was < 1 per 100,000 newborns in all regions with the exception of MENA where it approached 6 per 100,000 newborns. Few studies published data on the epidemiology of MMA mut, therefore no meta-analysis could have been performed on this subtype. Most of the identified papers reported birth prevalence estimates below 1 per 100,000 newborns for MMA mut. The systematic literature review clearly demonstrates that MMA and its subtypes are ultra-rare disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Metabolism, Inborn Errors/epidemiology , Methylmalonyl-CoA Mutase/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Female , Humans , Infant, Newborn , Male , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/epidemiology , Metabolism, Inborn Errors/genetics , Methylmalonyl-CoA Mutase/deficiency , Neonatal Screening
8.
Am J Hum Genet ; 104(4): 625-637, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30879639

ABSTRACT

Fabry disease is an X-linked lysosomal storage disease caused by loss of alpha galactosidase A (α-Gal A) activity and is characterized by progressive accumulation of globotriaosylceramide and its analogs in all cells and tissues. Although enzyme replacement therapy (ERT) is considered standard of care, the long-term effects of ERT on renal and cardiac manifestations remain uncertain and thus novel therapies are desirable. We herein report preclinical studies evaluating systemic messenger RNA (mRNA) encoding human α-Gal A in wild-type (WT) mice, α-Gal A-deficient mice, and WT non-human primates (NHPs). The pharmacokinetics and distribution of h-α-Gal A mRNA encoded protein in WT mice demonstrated prolonged half-lives of α-Gal A in tissues and plasma. Single intravenous administration of h-α-Gal A mRNA to Gla-deficient mice showed dose-dependent protein activity and substrate reduction. Moreover, long duration (up to 6 weeks) of substrate reductions in tissues and plasma were observed after a single injection. Furthermore, repeat i.v. administration of h-α-Gal A mRNA showed a sustained pharmacodynamic response and efficacy in Fabry mice model. Lastly, multiple administrations to non-human primates confirmed safety and translatability. Taken together, these studies across species demonstrate preclinical proof-of-concept of systemic mRNA therapy for the treatment of Fabry disease and this approach may be useful for other lysosomal storage disorders.


Subject(s)
Fabry Disease/genetics , Fabry Disease/therapy , RNA, Messenger/therapeutic use , alpha-Galactosidase/genetics , Animals , Disease Models, Animal , Endocytosis , Enzyme Replacement Therapy , Genetic Therapy , Humans , Lipids/chemistry , Lysosomes/metabolism , Macaca fascicularis , Male , Mice , Mice, Knockout , RNA, Messenger/pharmacokinetics , Tissue Distribution , Trihexosylceramides/metabolism
9.
Orphanet J Rare Dis ; 14(1): 40, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760309

ABSTRACT

Propionic acidemia (PA, OMIM #606054) is a serious, life-threatening, inherited, metabolic disorder caused by the deficiency of the mitochondrial enzyme propionyl-coenzyme A (CoA) carboxylase (EC 6.4.1.3). The primary objective of this study was to conduct a systematic literature review and meta-analysis on the epidemiology of PA. The literature search was performed covering Medline, Embase, Cochrane Database of Systematic Reviews, CRD Database, Academic Search Complete, CINAHL and PROSPERO databases. Websites of rare disease organizations were also searched for eligible studies. Of the 2338 identified records, 188 articles were assessed for eligibility in full text, 43 articles reported on disease epidemiology, and 31 studies were included into the quantitative synthesis. Due to the rarity of PA, broadly targeted population-based prevalence studies are not available. Nonetheless, implementation of newborn screening programs has allowed the estimation of the birth prevalence data of PA across multiple geographic regions. The pooled point estimates indicated detection rates of 0.29; 0.33; 0.33 and 4.24 in the Asia-Pacific, Europe, North America and the Middle East and North Africa (MENA) regions, respectively. Our systematic literature review and meta-analysis confirm that PA is an ultra-rare disorder, with similar detection rates across all regions with the exception of the MENA region where the disease, similar to other inherited metabolic disorders, is more frequent.


Subject(s)
Propionic Acidemia/diagnosis , Propionic Acidemia/epidemiology , Humans , Infant, Newborn , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/epidemiology , Neonatal Screening/methods
10.
Nat Med ; 24(12): 1899-1909, 2018 12.
Article in English | MEDLINE | ID: mdl-30297912

ABSTRACT

Acute intermittent porphyria (AIP) results from haploinsufficiency of porphobilinogen deaminase (PBGD), the third enzyme in the heme biosynthesis pathway. Patients with AIP have neurovisceral attacks associated with increased hepatic heme demand. Phenobarbital-challenged mice with AIP recapitulate the biochemical and clinical characteristics of patients with AIP, including hepatic overproduction of the potentially neurotoxic porphyrin precursors. Here we show that intravenous administration of human PBGD (hPBGD) mRNA (encoded by the gene HMBS) encapsulated in lipid nanoparticles induces dose-dependent protein expression in mouse hepatocytes, rapidly normalizing urine porphyrin precursor excretion in ongoing attacks. Furthermore, hPBGD mRNA protected against mitochondrial dysfunction, hypertension, pain and motor impairment. Repeat dosing in AIP mice showed sustained efficacy and therapeutic improvement without evidence of hepatotoxicity. Finally, multiple administrations to nonhuman primates confirmed safety and translatability. These data provide proof-of-concept for systemic hPBGD mRNA as a potential therapy for AIP.


Subject(s)
Genetic Therapy , Hydroxymethylbilane Synthase/genetics , Porphyria, Acute Intermittent/therapy , RNA, Messenger/administration & dosage , Animals , Disease Models, Animal , Female , Haploinsufficiency/genetics , Heme/genetics , Heme/metabolism , Hepatocytes/drug effects , Humans , Hydroxymethylbilane Synthase/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/pathology , RNA, Messenger/genetics
12.
Cell Rep ; 21(12): 3548-3558, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29262333

ABSTRACT

Isolated methylmalonic acidemia/aciduria (MMA) is a devastating metabolic disorder with poor outcomes despite current medical treatments. Like other mitochondrial enzymopathies, enzyme replacement therapy (ERT) is not available, and although promising, AAV gene therapy can be limited by pre-existing immunity and has been associated with genotoxicity in mice. To develop a new class of therapy for MMA, we generated a pseudoU-modified codon-optimized mRNA encoding human methylmalonyl-CoA mutase (hMUT), the enzyme most frequently mutated in MMA, and encapsulated it into biodegradable lipid nanoparticles (LNPs). Intravenous (i.v.) administration of hMUT mRNA in two different mouse models of MMA resulted in a 75%-85% reduction in plasma methylmalonic acid and was associated with increased hMUT protein expression and activity in liver. Repeat dosing of hMUT mRNA reduced circulating metabolites and dramatically improved survival and weight gain. Additionally, repeat i.v. dosing did not increase markers of liver toxicity or inflammation in heterozygote MMA mice.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Genetic Therapy/methods , Methylmalonyl-CoA Mutase/genetics , Nanoparticles/administration & dosage , RNA, Messenger/genetics , Administration, Intravenous , Animals , Female , Humans , Lipids/chemistry , Liver/metabolism , Male , Methylmalonyl-CoA Mutase/metabolism , Mice , Nanoparticles/chemistry , RNA, Messenger/metabolism
13.
Alzheimers Dement (N Y) ; 3(4): 603-611, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29201994

ABSTRACT

INTRODUCTION: Cognitive training improves cognitive performance and delays functional impairment, but its effects on dementia are not known. We examined whether three different types of cognitive training lowered the risk of dementia across 10 years of follow-up relative to control and if greater number of training sessions attended was associated with lower dementia risk. METHODS: The Advanced Cognitive Training in Vital Elderly (NCT00298558) study was a randomized controlled trial (N = 2802) among initially healthy older adults, which examined the efficacy of three cognitive training programs (memory, reasoning, or speed of processing) relative to a no-contact control condition. Up to 10 training sessions were delivered over 6 weeks with up to four sessions of booster training delivered at 11 months and a second set of up to four booster sessions at 35 months. Outcome assessments were taken immediately after intervention and at intervals over 10 years. Dementia was defined using a combination of interview- and performance-based methods. RESULTS: A total of 260 cases of dementia were identified during the follow-up. Speed training resulted in reduced risk of dementia (hazard ratio [HR] 0.71, 95% confidence interval [CI] 0.50-0.998, P = .049) compared to control, but memory and reasoning training did not (HR 0.79, 95% CI 0.57-1.11, P = .177 and HR 0.79, 95% CI 0.56-1.10, P = .163, respectively). Each additional speed training session was associated with a 10% lower hazard for dementia (unadjusted HR, 0.90; 95% CI, 0.85-0.95, P < .001). DISCUSSION: Initially, healthy older adults randomized to speed of processing cognitive training had a 29% reduction in their risk of dementia after 10 years of follow-up compared to the untreated control group.

14.
J Invest Dermatol ; 136(6): 1116-1123, 2016 06.
Article in English | MEDLINE | ID: mdl-26899947

ABSTRACT

The tissue half-life of proteins largely determines treatment frequency of non-gene-editing-based therapies targeting the cause of genodermatoses. Surprisingly, such knowledge is missing for a vast number of proteins involved in pathologies. The dermal-epidermal junction zone is believed to be a rather static structure, but to our knowledge no detailed analysis of the stability of proteins within this zone has been performed. Here, we addressed the in vivo half-life of collagen type VII using genetic ablation of its expression and therapeutic introduction of exogenous collagen VII in a preclinical model. A similar in vivo stability of collagen VII was observed in the skin, tongue, and esophagus, with a half-life of about 1 month. Collagen VII expressed by intradermally injected mesenchymal stromal cells also exhibited a similar half-life. Our study provides key information needed for the development of protein replacement or cell-based therapies for dystrophic epidermolysis bullosa caused by genetic deficiency of collagen VII. Moreover, by showing what we define as an intermediate half-life of collagen VII, our study challenges the view of the dermal-epidermal junction zone as a static structure with very slow turnover.


Subject(s)
Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/therapy , Genetic Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Animals , Blotting, Western , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Dermis/metabolism , Disease Models, Animal , Epidermis/metabolism , Epidermolysis Bullosa Dystrophica/genetics , Fibroblasts/cytology , Half-Life , Humans , Male , Mice , Mice, Knockout , Random Allocation , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/therapy
15.
Clin Exp Dent Res ; 2(2): 85-95, 2016 Nov.
Article in English | MEDLINE | ID: mdl-29744154

ABSTRACT

The purpose of the present study was to evaluate the longitudinal survival rate of the treatment of teeth affected with periodontally hopeless prognosis and secondary occlusal traumatism (SOT) using intentional replantation (IR) and periodontal prosthesis. We collected data from 17 individuals who received IR and participated in the study during 1995 to 2014. Of the 17 teeth affected by periodontally extreme conditions with deep angular bone defects, severe alveolar bone loss extending to or beyond the apex, and SOT, was recognized as having hopeless prognosis. Those teeth were treated sequentially using procedures that included basic periodontal therapy, therapeutic provisional prosthesis, IR, fixed dental prosthesis, crown and sleeve-coping telescopic dentures (CSCTDs), or fixed prosthesis and CSCTD combined. Longitudinal assessments of clinical parameters and radiographic bone change before and after IR were evaluated. Clinical results showed that the overall cumulative survival rate of assayed teeth after IR therapy (5-12 years) was 88.2%. The mean (±SD) estimated radiographic alveolar bone loss was 12.7 ± 2.1 mm (88.5% ± 13.3%) of the root length, initially, and estimated radiographic alveolar bone gain was 4.0 ± 2.2 mm ultimately, in 17 replanted teeth with SOT. Only one tooth (5.9%) exhibited root resorption. Ankylosis was not observed during the study. Periapical radiographs demonstrated that satisfactory periodontal healing of lamina dura and bone fills occurred in all replanted teeth with SOT. Generally, tooth mobility and SOT were significantly improved after therapy. Most treated teeth functioned well and remained stable clinically throughout the periods of study. The present study documented a promising outcome for autogenous IR and periprosthetic therapy of 17 periodontally hopeless teeth for 5-12 years. The present study revealed good bone gain and elimination of SOT and prominent occlusal function. We concluded that the application of IR, minocycline-HCL and periodontal prosthetic procedures later elevated the prognosis of these otherwise hopeless teeth with SOT, which are valuable options for retaining teeth with periodontally extreme situations.

16.
J Invest Dermatol ; 135(12): 3060-3067, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26203639

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited disorder characterized by skin fragility, blistering, and multiple skin wounds with no currently approved or consistently effective treatment. It is due to mutations in the gene encoding type VII collagen (C7). Using recombinant human C7 (rhC7) purified from human dermal fibroblasts (FB-rhC7), we showed previously that intravenously injected rhC7 distributed to engrafted RDEB skin, incorporated into its dermal-epidermal junction (DEJ), and reversed the RDEB disease phenotype. Human dermal fibroblasts, however, are not used for commercial production of therapeutic proteins. Therefore, we generated rhC7 from Chinese hamster ovary (CHO) cells. The CHO-derived recombinant type VII collagen (CHO-rhC7), similar to FB-rhC7, was secreted as a correctly folded, disulfide-bonded, helical trimer resistant to protease degradation. CHO-rhC7 bound to fibronectin and promoted human keratinocyte migration in vitro. A single dose of CHO-rhC7, administered intravenously into new-born C7-null RDEB mice, incorporated into the DEJ of multiple skin sites, tongue and esophagus, restored anchoring fibrils, improved dermal-epidermal adherence, and increased the animals' life span. Furthermore, no circulating or tissue-bound anti-C7 antibodies were observed in the mice. These data demonstrate the efficacy of CHO-rhC7 in a preclinical murine model of RDEB.


Subject(s)
Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/drug therapy , Animals , Animals, Newborn , CHO Cells , Cell Movement/drug effects , Cells, Cultured , Collagen Type VII/administration & dosage , Collagen Type VII/chemistry , Collagen Type VII/immunology , Cricetulus , Humans , Injections, Intravenous , Phenotype , Recombinant Proteins/therapeutic use
17.
PLoS One ; 9(5): e89952, 2014.
Article in English | MEDLINE | ID: mdl-24818791

ABSTRACT

INTRODUCTION: Germline variants in TP63 have been consistently associated with several tumors, including bladder cancer, indicating the importance of TP53 pathway in cancer genetic susceptibility. However, variants in other related genes, including TP53 rs1042522 (Arg72Pro), still present controversial results. We carried out an in depth assessment of associations between common germline variants in the TP53 pathway and bladder cancer risk. MATERIAL AND METHODS: We investigated 184 tagSNPs from 18 genes in 1,058 cases and 1,138 controls from the Spanish Bladder Cancer/EPICURO Study. Cases were newly-diagnosed bladder cancer patients during 1998-2001. Hospital controls were age-gender, and area matched to cases. SNPs were genotyped in blood DNA using Illumina Golden Gate and TaqMan assays. Cases were subphenotyped according to stage/grade and tumor p53 expression. We applied classical tests to assess individual SNP associations and the Least Absolute Shrinkage and Selection Operator (LASSO)-penalized logistic regression analysis to assess multiple SNPs simultaneously. RESULTS: Based on classical analyses, SNPs in BAK1 (1), IGF1R (5), P53AIP1 (1), PMAIP1 (2), SERINPB5 (3), TP63 (3), and TP73 (1) showed significant associations at p-value≤0.05. However, no evidence of association, either with overall risk or with specific disease subtypes, was observed after correction for multiple testing (p-value≥0.8). LASSO selected the SNP rs6567355 in SERPINB5 with 83% of reproducibility. This SNP provided an OR = 1.21, 95%CI 1.05-1.38, p-value = 0.006, and a corrected p-value = 0.5 when controlling for over-estimation. DISCUSSION: We found no strong evidence that common variants in the TP53 pathway are associated with bladder cancer susceptibility. Our study suggests that it is unlikely that TP53 Arg72Pro is implicated in the UCB in white Europeans. SERPINB5 and TP63 variation deserve further exploration in extended studies.


Subject(s)
Tumor Suppressor Protein p53/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Female , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Young Adult
18.
J Am Geriatr Soc ; 62(1): 16-24, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24417410

ABSTRACT

OBJECTIVES: To determine the effects of cognitive training on cognitive abilities and everyday function over 10 years. DESIGN: Ten-year follow-up of a randomized, controlled single-blind trial (Advanced Cognitive Training for Independent and Vital Elderly (ACTIVE)) with three intervention groups and a no-contact control group. SETTING: Six U.S. cities. PARTICIPANTS: A volunteer sample of 2,832 persons (mean baseline age 73.6; 26% African American) living independently. INTERVENTION: Ten training sessions for memory, reasoning, or speed of processing; four sessions of booster training 11 and 35 months after initial training. MEASUREMENTS: Objectively measured cognitive abilities and self-reported and performance-based measures of everyday function. RESULTS: Participants in each intervention group reported less difficulty with instrumental activities of daily living (IADLs) (memory: effect size = 0.48, 99% confidence interval (CI) = 0.12-0.84; reasoning: effect size = 0.38, 99% CI = 0.02-0.74; speed of processing: effect size = 0.36, 99% CI = 0.01-0.72). At a mean age of 82, approximately 60% of trained participants, versus 50% of controls (P < .05), were at or above their baseline level of self-reported IADL function at 10 years. The reasoning and speed-of-processing interventions maintained their effects on their targeted cognitive abilities at 10 years (reasoning: effect size = 0.23, 99% CI = 0.09-0.38; speed of processing: effect size = 0.66, 99% CI = 0.43-0.88). Memory training effects were no longer maintained for memory performance. Booster training produced additional and durable improvement for the reasoning intervention for reasoning performance (effect size = 0.21, 99% CI = 0.01-0.41) and the speed-of-processing intervention for speed-of-processing performance (effect size = 0.62, 99% CI = 0.31-0.93). CONCLUSION: Each Advanced Cognitive Training for Independent and Vital Elderly cognitive intervention resulted in less decline in self-reported IADL compared with the control group. Reasoning and speed, but not memory, training resulted in improved targeted cognitive abilities for 10 years.


Subject(s)
Activities of Daily Living/psychology , Aging , Cognition Disorders/prevention & control , Cognitive Behavioral Therapy , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Independent Living , Male , Memory Disorders/prevention & control , Mental Processes , Single-Blind Method , United States
19.
J Thorac Cardiovasc Surg ; 148(4): 1467-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24332668

ABSTRACT

OBJECTIVE: To compare the interstage cardiac catheterization hemodynamic and angiographic findings between shunt types for the Pediatric Heart Network Single Ventricle Reconstruction trial. The trial, which randomized subjects to a modified Blalock-Taussig shunt (MBTS) or right ventricle-to-pulmonary artery shunt (RVPAS) for the Norwood procedure, demonstrated the RVPAS was associated with a smaller pulmonary artery diameter but superior 12-month transplant-free survival. METHODS: We analyzed the pre-stage II catheterization data for the trial subjects. The hemodynamic variables and shunt and pulmonary angiographic data were compared between shunt types; their association with 12-month transplant-free survival was also evaluated. RESULTS: Of 549 randomized subjects, 389 underwent pre-stage II catheterization. A smaller size, lower aortic and superior vena cava saturation, and higher ventricular end-diastolic pressure were associated with worse 12-month transplant-free survival. The MBTS group had a lower coronary perfusion pressure (27 vs 32 mm Hg; P<.001) and greater pulmonary blood flow/systemic blood flow ratio (1.1 vs 1.0, P=.009). A greater pulmonary blood flow/systemic blood flow ratio increased the risk of death or transplantation only in the RVPAS group (P=.01). The MBTS group had fewer shunt (14% vs 28%, P=.004) and severe left pulmonary artery (0.7% vs 9.2%, P=.003) stenoses, larger mid-main branch pulmonary artery diameters, and greater Nakata indexes (164 vs 134, P<.001). CONCLUSIONS: Compared with the RVPAS subjects, the MBTS subjects had more hemodynamic abnormalities related to shunt physiology, and the RVPAS subjects had more shunt or pulmonary obstruction of a severe degree and inferior pulmonary artery growth at pre-stage II catheterization. A lower body surface area, greater ventricular end-diastolic pressure, and lower superior vena cava saturation were associated with worse 12-month transplant-free survival.


Subject(s)
Blalock-Taussig Procedure , Heart Defects, Congenital/surgery , Heart Ventricles/abnormalities , Heart Ventricles/surgery , Norwood Procedures , Pulmonary Artery/abnormalities , Child, Preschool , Coronary Angiography , Female , Heart Defects, Congenital/diagnostic imaging , Heart Ventricles/diagnostic imaging , Hemodynamics , Humans , Male , North America , Treatment Outcome
20.
Circ Cardiovasc Imaging ; 6(6): 934-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24097422

ABSTRACT

BACKGROUND: The Pediatric Heart Network's Single Ventricle Reconstruction (SVR) trial randomized infants with single right ventricles (RVs) undergoing a Norwood procedure to a modified Blalock-Taussig or RV-to-pulmonary artery shunt. This report compares RV parameters in the 2 groups using 3-dimensional echocardiography. METHODS AND RESULTS: Three-dimensional echocardiography studies were obtained at 10 of 15 SVR centers. Of the 549 subjects, 314 underwent 3-dimensional echocardiography studies at 1 to 4 time points (pre-Norwood, post-Norwood, pre-stage II, and 14 months) for a total of 757 3-dimensional echocardiography studies. Of these, 565 (75%) were acceptable for analysis. RV volume, mass, mass:volume ratio, ejection fraction, and severity of tricuspid regurgitation did not differ by shunt type. RV volumes and mass did not change after the Norwood, but increased from pre-Norwood to pre-stage II (end-diastolic volume [milliliters]/body surface area [BSA](1.3), end-systolic volume [milliliters]/BSA(1.3), and mass [grams]/BSA(1.3) mean difference [95% confidence interval]=25.0 [8.7-41.3], 19.3 [8.3-30.4], and 17.9 [7.3-28.5], then decreased by 14 months (end-diastolic volume/BSA(1.3), end-systolic volume/BSA(1.3), and mass/BSA(1.3) mean difference [95% confidence interval]=-24.4 [-35.0 to -13.7], -9.8 [-17.9 to -1.7], and -15.3 [-22.0 to -8.6]. Ejection fraction decreased from pre-Norwood to pre-stage II (mean difference [95% confidence interval]=-3.7 [-6.9 to -0.5]), but did not decrease further by 14 months. CONCLUSIONS: We found no statistically significant differences between study groups in 3-dimensional echocardiography measures of RV size and function, or magnitude of tricuspid regurgitation. Volume unloading was seen after stage II, as expected, but ejection fraction did not improve. This study provides insights into the remodeling of the operated univentricular RV in infancy.


Subject(s)
Echocardiography, Three-Dimensional/methods , Heart Ventricles/abnormalities , Hypoplastic Left Heart Syndrome/diagnostic imaging , Norwood Procedures/methods , Pulmonary Artery/surgery , Child, Preschool , Female , Heart Ventricles/diagnostic imaging , Heart Ventricles/surgery , Humans , Hypoplastic Left Heart Syndrome/physiopathology , Hypoplastic Left Heart Syndrome/surgery , Male , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...