Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Gene Ther ; 30(3-4): 386-397, 2023 04.
Article in English | MEDLINE | ID: mdl-36258038

ABSTRACT

Gene editing for the cure of inborn errors of metabolism (IEMs) has been limited by inefficiency of adult hepatocyte targeting. Here, we demonstrate that in utero CRISPR/Cas9-mediated gene editing in a mouse model of hereditary tyrosinemia type 1 provides stable cure of the disease. Following this, we performed an extensive gene expression analysis to explore the inherent characteristics of fetal/neonatal hepatocytes that make them more susceptible to efficient gene editing than adult hepatocytes. We showed that fetal and neonatal livers are comprised of proliferative hepatocytes with abundant expression of genes involved in homology-directed repair (HDR) of DNA double-strand breaks (DSBs), key for efficient gene editing by CRISPR/Cas9. We demonstrated the same is true of hepatocytes after undergoing a regenerative stimulus (partial hepatectomy), where post-hepatectomy cells show a higher efficiency of HDR and correction. Specifically, we demonstrated that HDR-related genome correction is most effective in the replicative phase, or S-phase, of an actively proliferating cell. In conclusion, this study shows that taking advantage of or triggering cell proliferation, specifically DNA replication in S-phase, may serve as an important tool to improve efficiency of CRISPR/Cas9-mediated genome editing in the liver and provide a curative therapy for IEMs in both children and adults.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Mice , Recombinational DNA Repair , DNA Breaks, Double-Stranded , DNA , DNA Repair
2.
Nat Commun ; 13(1): 5012, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008405

ABSTRACT

Conventional therapy for hereditary tyrosinemia type-1 (HT1) with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) delays and in some cases fails to prevent disease progression to liver fibrosis, liver failure, and activation of tumorigenic pathways. Here we demonstrate cure of HT1 by direct, in vivo administration of a therapeutic lentiviral vector targeting the expression of a human fumarylacetoacetate hydrolase (FAH) transgene in the porcine model of HT1. This therapy is well tolerated and provides stable long-term expression of FAH in pigs with HT1. Genomic integration displays a benign profile, with subsequent fibrosis and tumorigenicity gene expression patterns similar to wild-type animals as compared to NTBC-treated or diseased untreated animals. Indeed, the phenotypic and genomic data following in vivo lentiviral vector administration demonstrate comparative superiority over other therapies including ex vivo cell therapy and therefore support clinical application of this approach.


Subject(s)
Precancerous Conditions , Tyrosinemias , Animals , Disease Models, Animal , Genetic Therapy , Humans , Hydrolases/genetics , Hydrolases/metabolism , Liver Cirrhosis/therapy , Nitrobenzoates/pharmacology , Nitrobenzoates/therapeutic use , Swine , Tyrosinemias/genetics , Tyrosinemias/therapy
3.
Biochemistry ; 2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35436085

ABSTRACT

CRISPR gene editing and control systems continue to emerge and inspire novel research and clinical applications. Advances in CRISPR performance such as optimizing the duration of activity in cells, tissues, and organisms, as well as limiting off-target activities, have been extremely important for expanding the utility of CRISPR-based systems. By investigating the effects of various chemical modifications in guide RNAs (gRNAs) at defined positions and combinations, we find that 2'-O-methyl-3'-phosphonoacetate (MP) modifications can be substantially more effective than 2'-O-methyl-3'-phosphorothioate (MS) modifications at the 3' ends of single-guide RNAs (sgRNAs) to promote high editing yields, in some instances showing an order of magnitude higher editing yield in human cells. MP-modified 3' ends are especially effective at promoting the activity of guide RNAs cotransfected with Cas messenger RNA (mRNA), as the gRNA must persist in cells until the Cas protein is expressed. We demonstrate such an MP enhancement for sgRNAs cotransfected with a BE4 mRNA for cytidine base editing and also demonstrate that MP at the 3' ends of prime editing guide RNAs (pegRNAs) cotransfected with PE2 mRNA can promote maximal prime editing yields. In the presence of serum, sgRNAs with MP-modified 3' ends showed marked improvements in editing efficiency over sgRNAs with MS-modified 3' ends codelivered with Cas9 mRNA and showed more modest improvements at enhancing the activity of transfected ribonucleoprotein (RNP) complexes. Our results suggest that MP should be considered as a performance-enhancing modification for the 3' ends of synthetic gRNAs, especially in situations where the guide RNAs may be susceptible to exonuclease-mediated degradation.

5.
J Inherit Metab Dis ; 44(6): 1369-1381, 2021 11.
Article in English | MEDLINE | ID: mdl-33896013

ABSTRACT

Phenylketonuria (PKU) is the most common inborn error of metabolism of the liver, and results from mutations of both alleles of the phenylalanine hydroxylase gene (PAH). As such, it is a suitable target for gene therapy via gene delivery with a recombinant adeno-associated virus (AAV) vector. Here we use the synthetic AAV vector Anc80 via systemic administration to deliver a functional copy of a codon-optimized human PAH gene, with or without an intron spacer, to the Pahenu2 mouse model of PKU. Dose-dependent transduction of the liver and expression of PAH mRNA were present with both vectors, resulting in significant and durable reduction of circulating phenylalanine, reaching near control levels in males. Coat color of treated Pahenu2 mice reflected an increase in pigmentation from brown to the black color of control animals, further indicating functional restoration of phenylalanine metabolism and its byproduct melanin. There were no adverse effects associated with administration of AAV up to 5 × 1012 VG/kg, the highest dose tested. Only minor and/or transient variations in some liver enzymes were observed in some of the AAV-dosed animals which were not associated with pathology findings in the liver. Finally, there was no impact on cell turnover or apoptosis as evaluated by Ki-67 and TUNEL staining, further supporting the safety of this approach. This study demonstrates the therapeutic potential of AAV Anc80 to safely and durably cure PKU in a mouse model, supporting development for clinical consideration.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Phenylalanine Hydroxylase/genetics , Phenylketonurias/therapy , Animals , Cell Line , DNA, Recombinant/administration & dosage , Disease Models, Animal , Female , Genetic Vectors/genetics , Hair Color , Humans , Injections, Intravenous , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Phenylalanine/blood , Phenylalanine Hydroxylase/immunology , Phenylalanine Hydroxylase/metabolism , Transduction, Genetic/methods
6.
PLoS One ; 16(1): e0245831, 2021.
Article in English | MEDLINE | ID: mdl-33493163

ABSTRACT

Phenylketonuria (PKU) is a metabolic disorder whereby phenylalanine metabolism is deficient due to allelic variations in the gene for phenylalanine hydroxylase (PAH). There is no cure for PKU other than orthotopic liver transplantation, and the standard of care for patients is limited to dietary restrictions and key amino acid supplementation. Therefore, Pah was edited in pig fibroblasts for the generation of PKU clone piglets that harbor a common and severe human mutation, R408W. Additionally, the proximal region to the mutation was further humanized by introducing 5 single nucleotide polymorphisms (SNPs) to allow for development of gene editing machinery that could be translated directly from the pig model to human PKU patients that harbor at least one classic R408W allele. Resulting piglets were hypopigmented (a single Ossabaw piglet) and had low birthweight (all piglets). The piglets had similar levels of PAH expression, but no detectable enzymatic activity, consistent with the human phenotype. The piglets were fragile and required extensive neonatal care to prevent failure to thrive and early demise. Phenylalanine levels rose sharply when dietary Phe was unrestricted but could be rapidly reduced with a low Phe diet. Fibroblasts isolated from R408W piglets show susceptibility to correction using CRISPR or TALEN, with subsequent homology-directed recombination to correct Pah. This pig model of PKU provides a powerful new tool for development of all classes of therapeutic candidates to treat or cure PKU, as well as unique value for proof-of-concept studies for in vivo human gene editing platforms in the context of this humanized PKU allele.


Subject(s)
Gene Editing/methods , Mutation , Phenylalanine Hydroxylase/genetics , Phenylketonurias/genetics , Animals , Base Sequence , Disease Models, Animal , Humans , Phenotype , Safety , Swine
8.
Expert Opin Orphan Drugs ; 8(7): 245-256, 2020.
Article in English | MEDLINE | ID: mdl-33224636

ABSTRACT

INTRODUCTION: Inborn errors of metabolism (IEMs) often result from single-gene mutations and collectively cause liver dysfunction in neonates leading to chronic liver and systemic disease. Current treatments for many IEMs are limited to maintenance therapies that may still require orthotropic liver transplantation. Gene therapies offer a potentially superior approach by correcting or replacing defective genes with functional isoforms; however, they face unique challenges from complexities presented by individual diseases and their diverse etiology, presentation, and pathophysiology. Furthermore, immune responses, off-target gene disruption, and tumorigenesis are major concerns that need to be addressed before clinical application of gene therapy. AREAS COVERED: The current treatments for IEMs are reviewed as well as the advances in, and barriers to, gene therapy for IEMs. Attention is then given to ex vivo and in vivo gene therapy approaches for hereditary tyrosinemia type 1 (HT1). Of all IEMs, HT1 is particularly amenable to gene therapy because of a selective growth advantage conferred to corrected cells, thereby lowering the initial transduction threshold for phenotypic relevance. EXPERT OPINION: It is proposed that not only is HT1 a safe indication for gene therapy, its unique characteristics position it to be an ideal IEM to develop for clinical investigation.

9.
Mol Ther Methods Clin Dev ; 18: 738-750, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32913881

ABSTRACT

The effectiveness of cell-based therapies to treat liver failure is often limited by the diseased liver environment. Here, we provide preclinical proof of concept for hepatocyte transplantation into lymph nodes as a cure for liver failure in a large-animal model with hereditary tyrosinemia type 1 (HT1), a metabolic liver disease caused by deficiency of fumarylacetoacetate hydrolase (FAH) enzyme. Autologous porcine hepatocytes were transduced ex vivo with a lentiviral vector carrying the pig Fah gene and transplanted into mesenteric lymph nodes. Hepatocytes showed early (6 h) and durable (8 months) engraftment in lymph nodes, with reproduction of vascular and hepatic microarchitecture. Subsequently, hepatocytes migrated to and repopulated the native diseased liver. The corrected cells generated sufficient liver mass to clinically ameliorate the acute liver failure and HT1 disease as early as 97 days post-transplantation. Integration site analysis defined the corrected hepatocytes in the liver as a subpopulation of hepatocytes from lymph nodes, indicating that the lymph nodes served as a source for healthy hepatocytes to repopulate a diseased liver. Therefore, ectopic transplantation of healthy hepatocytes cures this pig model of liver failure and presents a promising approach for the development of cures for liver disease in patients.

10.
Praxis (Bern 1994) ; 109(9): 736-742, 2020 Jul.
Article in German | MEDLINE | ID: mdl-32635846

ABSTRACT

Rare Cause for Difficulty in Swallowing with Neck Swelling - Paraganglioma Abstract. Pain-free lateral swelling of the neck should be taken seriously and promptly clarified by using sonography. If a cervical paraganglioma is suspected, an interdisciplinary clarification should be initiated. In addition to an MRI of the neck, a vascular-surgical, an ENT specialist and an endocrinological examination should be carried out. If there is a high degree of suspicion of a carotid body tumor, surgical removal or a prospective procedure should be used in accordance with the Shamblin classification, the genetic clarification and the patient's individual life situation. If there is suspicion of vagal paraganglioma due to the location of the tumor and the symptoms, an expectative approach with regular check-ups should be considered in a symptom-poor and elderly patient.


Subject(s)
Carotid Body Tumor , Deglutition , Head and Neck Neoplasms , Paraganglioma , Aged , Carotid Body Tumor/complications , Head and Neck Neoplasms/complications , Humans , Paraganglioma/complications , Prospective Studies
11.
Hepatol Commun ; 3(4): 558-573, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30976745

ABSTRACT

Ex vivo CRISPR/Cas9-mediated gene editing in hepatocytes using homology-directed repair (HDR) is a potential alternative curative therapy to organ transplantation for metabolic liver disease. However, a major limitation of this approach in quiescent adult primary hepatocytes is that nonhomologous end-joining is the predominant DNA repair pathway for double-strand breaks (DSBs). This study explored the hypothesis that ex vivo hepatocyte culture could reprogram hepatocytes to favor HDR after CRISPR/Cas9-mediated DNA DSBs. Quantitative PCR (qPCR), RNA sequencing, and flow cytometry demonstrated that within 24 hours, primary mouse hepatocytes in ex vivo monolayer culture decreased metabolic functions and increased expression of genes related to mitosis progression and HDR. Despite the down-regulation of hepatocyte function genes, hepatocytes cultured for up to 72 hours could robustly engraft in vivo. To assess functionality long-term, primary hepatocytes from a mouse model of hereditary tyrosinemia type 1 bearing a single-point mutation were transduced ex vivo with two adeno-associated viral vectors to deliver the Cas9 nuclease, target guide RNAs, and a 1.2-kb homology template. Adeno-associated viral Cas9 induced robust cutting at the target locus, and, after delivery of the repair template, precise correction of the point mutation occurred by HDR. Edited hepatocytes were transplanted into recipient fumarylacetoacetate hydrolase knockout mice, resulting in engraftment, robust proliferation, and prevention of liver failure. Weight gain and biochemical assessment revealed normalization of metabolic function. Conclusion: The results of this study demonstrate the potential therapeutic effect of ex vivo hepatocyte-directed gene editing after reprogramming to cure metabolic disease in a preclinical model of hereditary tyrosinemia type 1.

12.
Hum Gene Ther Clin Dev ; 30(2): 57-66, 2019 06.
Article in English | MEDLINE | ID: mdl-30860398

ABSTRACT

General safety and toxicology assessments supporting in vivo lentiviral vector-based therapeutic development are sparse. We have previously demonstrated the efficacy of a lentiviral vector expressing fumarylacetoacetate hydrolase (LV-FAH) to cure animal models of hereditary tyrosinemia type 1. Therefore, we performed a complete preclinical toxicological evaluation of LV-FAH, in a large cohort (n = 20/group) of wildtype mice and included matched groups of N-nitrosodiethylamine/carbon tetrachloride (DEN/CCl4)-induced liver injury mice to assess specific toxicity in fibrotic liver tissue. Mice receiving LV-FAH alone (109 TU/mouse) or in combination with DEN/CCl4 presented clinically similar to control animals, with only slight reductions in total body weight gains over the study period (3.2- to 3.7-fold vs. 4.2-fold). There were no indications of toxicity attributed to administration of LV-FAH alone over the duration of this study. The known hepatotoxic combination of DEN/CCl4 induced fibrotic liver injury, and co-administration with LV-FAH was associated with exaggeration of some findings such as an increased liver:body weight ratio and progression to focal hepatocyte necrosis in some animals. Hepatocellular degeneration/regeneration was present in DEN/CCl4-dosed animals regardless of LV-FAH as evaluated by Ki-67 immunohistochemistry and circulating alpha fetoprotein levels, but there were no tumors identified in any tissue in any dose group. These data demonstrate the inherent safety of LV-FAH and support broader clinical development of lentiviral vectors for in vivo administration.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Genetic Therapy , Genetic Vectors , Hydrolases/genetics , Lentivirus/genetics , Animals , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/genetics , Diethylnitrosamine , Disease Models, Animal , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL
13.
Cell Transplant ; 28(1): 79-88, 2019 01.
Article in English | MEDLINE | ID: mdl-30477316

ABSTRACT

Orthotopic liver transplantation remains the only curative therapy for inborn errors of metabolism. Given the tremendous success for primary immunodeficiencies using ex-vivo gene therapy with lentiviral vectors, there is great interest in developing similar curative therapies for metabolic liver diseases. We have previously generated a pig model of hereditary tyrosinemia type 1 (HT1), an autosomal recessive disorder caused by deficiency of fumarylacetoacetate hydrolase (FAH). Using this model, we have demonstrated curative ex-vivo gene and cell therapy using a lentiviral vector to express FAH in autologous hepatocytes. To further evaluate the long-term clinical outcomes of this therapeutic approach, we continued to monitor one of these pigs over the course of three years. The animal continued to thrive off the protective drug NTBC, gaining weight appropriately, and maintaining sexual fecundity for the course of his life. The animal was euthanized 31 months after transplantation to perform a thorough biochemical and histological analysis. Biochemically, liver enzymes and alpha-fetoprotein levels remained normal and abhorrent metabolites specific to HT1 remained corrected. Liver histology showed no evidence of tumorigenicity and Masson's trichrome staining revealed minimal fibrosis and no evidence of cirrhosis. FAH-immunohistochemistry revealed complete repopulation of the liver by transplanted FAH-positive cells. A complete histopathological report on other organs, including kidney, revealed no abnormalities. This study is the first to demonstrate long-term safety and efficacy of hepatocyte-directed gene therapy in a large animal model. We conclude that hepatocyte-directed ex-vivo gene therapy is a rational choice for further exploration as an alternative therapeutic approach to whole organ transplantation for metabolic liver disease, including HT1.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Hydrolases/metabolism , Tyrosinemias/enzymology , Tyrosinemias/therapy , Animals , Computational Biology , Disease Models, Animal , Hydrolases/genetics , Male , Swine , Tyrosinemias/metabolism
14.
J Vis Exp ; (141)2018 11 04.
Article in English | MEDLINE | ID: mdl-30451238

ABSTRACT

Gene therapy is an ideal choice to cure many inborn errors of metabolism of the liver. Ex-vivo, lentiviral vectors have been used successfully in the treatment of many hematopoietic diseases in humans, as their use offers stable transgene expression due to the vector's ability to integrate into the host genome. This method demonstrates the application of ex vivo gene therapy of hepatocytes to a large animal model of hereditary tyrosinemia type I. This process consists of 1) isolation of primary hepatocytes from the autologous donor/recipient animal, 2) ex vivo gene delivery via hepatocyte transduction with a lentiviral vector, and 3) autologous transplant of corrected hepatocytes via portal vein injection. Success of the method generally relies upon efficient and sterile removal of the liver resection, careful handling of the excised specimen for isolation of viable hepatocytes sufficient for re-engrafting, high-percentage transduction of the isolated cells, and aseptic surgical procedures throughout to prevent infection. Technical failure at any of these steps will result in low yield of viable transduced hepatocytes for autologous transplant or infection of the donor/recipient animal. The pig model of human type 1 hereditary tyrosinemia (HT-1) chosen for this approach is uniquely amenable to such a method, as even a small percentage of engraftment of corrected cells will lead to repopulation of the liver with healthy cells based on a powerful selective advantage over native-diseased hepatocytes. Although this growth selection will not be true for all indications, this approach is a foundation for expansion into other indications and allows for manipulation of this environment to address additional diseases, both within the liver and beyond, while controlling for exposure to viral vector and opportunity for off-target toxicity and tumorigenicity.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/genetics , Hepatocytes/transplantation , Transplantation, Autologous/methods , Animals , Disease Models, Animal , Swine
15.
Surgery ; 164(3): 473-481, 2018 09.
Article in English | MEDLINE | ID: mdl-29884476

ABSTRACT

BACKGROUND: Autologous hepatocyte transplantation after ex vivo gene therapy is an alternative to liver transplantation for metabolic liver disease. Here we evaluate ex vivo gene therapy followed by transplantation of single-cell or spheroid hepatocytes. METHODS: Pig and mouse hepatocytes were isolated, labeled with zirconium-89 and returned to the liver as single cells or spheroids. Biodistribution was evaluated through positron emission tomography-computed tomography. Fumarylacetoacetate hydrolase-deficient pig hepatocytes were isolated and transduced with a lentiviral vector containing the Fah gene. Animals received portal vein infusion of single-cell or spheroid autologous hepatocytes after ex vivo gene delivery. Portal pressures were measured and ultrasound was used to evaluate for thrombus. Differences in engraftment and expansion of ex vivo corrected single-cell or spheroid hepatocytes were followed through histologic analysis and animals' ability to thrive off 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione. RESULTS: Positron emission tomography-computed tomography imaging showed spheroid hepatocytes with increased heterogeneity in biodistribution as compared with single cells, which spread more uniformly throughout the liver. Animals receiving spheroids experienced higher mean changes in portal pressure than animals receiving single cells (P < .01). Additionally, two animals from the spheroid group developed portal vein thrombi that required systemic anticoagulation. Immunohistochemical analysis of spheroid- and single-cell-transplanted animals showed similar engraftment and expansion rates of fumarylacetoacetate hydrolase-positive hepatocytes in the liver, correlating with similar weight stabilization curves. CONCLUSION: Ex vivo gene correction of autologous hepatocytes in fumarylacetoacetate hydrolase-deficient pigs can be performed using hepatocyte spheroids or single-cell hepatocytes, with spheroids showing a more heterogeneous distribution within the liver and higher risks for portal vein thrombosis and increased portal pressures.


Subject(s)
Cell Transplantation/methods , Genetic Therapy , Hepatocytes/transplantation , Spheroids, Cellular/transplantation , Tyrosinemias/therapy , Animals , Disease Models, Animal , Female , Male , Mice , Swine , Tyrosinemias/diagnostic imaging , Tyrosinemias/pathology
16.
J Pers Soc Psychol ; 114(1): 110-130, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28557471

ABSTRACT

This study advanced knowledge on charisma by (a) introducing a new personality-based model to conceptualize and assess charisma and by (b) investigating curvilinear relationships between charismatic personality and leader effectiveness. Moreover, we delved deeper into this curvilinear association by (c) examining moderation by the leader's level of adjustment and by (d) testing a process model through which the effects of charismatic personality on effectiveness are explained with a consideration of specific leader behaviors. Study 1 validated HDS charisma (Hogan Development Survey) as a useful trait-based measure of charisma. In Study 2 a sample of leaders (N = 306) were assessed in the context of a 360-degree development center. In line with the too-much-of-a-good-thing effect, an inverted U-shaped relationship between charismatic personality and observer-rated leader effectiveness was found, indicating that moderate levels are better than low or high levels of charisma. Study 3 (N = 287) replicated this curvilinear relationship and further illustrated the moderating role of leader adjustment, in such a way that the inflection point after which the effects of charisma turn negative occurs at higher levels of charisma when adjustment is high. Nonlinear mediation modeling further confirmed that strategic and operational leader behaviors fully mediate the curvilinear relationship. Leaders low on charisma are less effective because they lack strategic behavior; highly charismatic leaders are less effective because they lack operational behavior. In sum, this work provides insight into the dispositional nature of charisma and uncovers the processes through which and conditions under which leader charisma translates into (in)effectiveness. (PsycINFO Database Record


Subject(s)
Employment/psychology , Leadership , Personality , Social Perception , Adult , Female , Humans , Male , Middle Aged , Young Adult
18.
Gerontologist ; 57(1): 46-53, 2017 02.
Article in English | MEDLINE | ID: mdl-27497450

ABSTRACT

As professionals in geriatric medicine and social work, we are caregivers for our widowed mother of 90 years, a woman with neurocognitive disorder and multiple medical conditions. She has had repeated, problematic encounters with the health care system over the past 4 years. Caring successfully for an aging parent requires a comprehensive understanding of her unique medical, psychological, and functional status; need for social support; and overall goals of care. Poor communication between and among clinical teams-and with patients and families-is ubiquitous. The patient and family are not consistently listened to, or integrated, into the clinical team. We recount our experiences of one hospitalization and how we addressed the recurring obstacles we faced. Our training and experience gave us a firm understanding of the hazards of hospitalizing an elderly person and the need to be present, engaged, attentive, active, and vigilant. We caught and corrected major mistakes: failure to follow-up abnormal test results, multiple medication errors, undertreatment of pain, poor fall prevention, and inappropriate assessment and placement for rehabilitation. In a dysfunctional health care system, the family is, and must be, the ultimate fail-safe mechanism. We identify potentially effective solutions for the problems we encountered: adoption of dementia-sensitive and patient- and family-centered care, improved communication, better management of information (including better systems for monitoring lab results and for dispensing and reconciling medications), expediting care, changing reimbursement and regulation, and improving discharge planning and placement.


Subject(s)
Neurocognitive Disorders/nursing , Professional-Family Relations , Aged, 80 and over , Caregivers/psychology , Communication , Female , Hospitalization , Humans , Medical Errors , Medical Records Systems, Computerized , Mother-Child Relations , Neurocognitive Disorders/complications
19.
Article in English | MEDLINE | ID: mdl-27039257

ABSTRACT

INTRODUCTION: The Safety Pharmacology Society (SPS) and National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs) conducted a survey and workshop in 2015 to define current industry practices relating to housing of non-rodents during telemetry recordings in safety pharmacology and toxicology studies. The aim was to share experiences, canvas opinion on the study procedures/designs that could be used and explore the barriers to social housing. METHODS: Thirty-nine sites, either running studies (Sponsors or Contract Research Organisations, CROs) and/or outsourcing work responded to the survey (51% from Europe; 41% from USA). RESULTS: During safety pharmacology studies, 84, 67 and 100% of respondents socially house dogs, minipigs and non-human primates (NHPs) respectively on non-recording days. However, on recording days 20, 20 and 33% of respondents socially house the animals, respectively. The main barriers for social housing were limitations in the recording equipment used, study design and animal temperament/activity. During toxicology studies, 94, 100 and 100% of respondents socially house dogs, minipigs and NHPs respectively on non-recording days. However, on recording days 31, 25 and 50% of respondents socially house the animals, respectively. The main barriers for social housing were risk of damage to and limitations in the recording equipment used, food consumption recording and temperament/activity of the animals. CONCLUSIONS: Although the majority of the industry does not yet socially house animals during telemetry recordings in safety pharmacology and toxicology studies, there is support to implement this refinement. Continued discussions, sharing of best practice and data from companies already socially housing, combined with technology improvements and investments in infrastructure are required to maintain the forward momentum of this refinement across the industry.


Subject(s)
Hemodynamics/drug effects , Housing, Animal , Social Environment , Animals , Dogs , Drug Evaluation, Preclinical , Pharmacology/methods , Primates , Safety , Surveys and Questionnaires , Swine , Swine, Miniature , Telemetry , Temperament , Toxicology/methods
20.
World Hosp Health Serv ; 52(2): 19-22, 2016.
Article in English | MEDLINE | ID: mdl-30716237

ABSTRACT

Service excellence Training is an important continuing priority in Lorma Medical Center. Its design and contents are modified as needed to respond to patients' comments on the quality of service by Lorma staff. Noted inadequacies in customer satisfaction were bared in the Patient Surveys of Lorma in 2013. Floating of Satisfaction Assessments is done monthly by the Patient Relations Officer and summaries of the same are submitted by the Executive Secretary to the Department Heads concerned for immediate action, monitoring and reporting on improvements made. The premise is that process improvements should be based on data (1).


Subject(s)
Hospital Administration , Patient Satisfaction , Quality of Health Care , Health Care Surveys , Hospital Bed Capacity, 100 to 299 , Inservice Training , Job Satisfaction , Organizational Case Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...