Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
1.
Adv Biol Regul ; 87: 100920, 2023 01.
Article in English | MEDLINE | ID: mdl-36216757

ABSTRACT

Mutations of splicing factor genes (including SF3B1, SRSF2, U2AF1 and ZRSR2) occur in more than half of all patients with myelodysplastic syndromes (MDS), a heterogeneous group of myeloid neoplasms. Splicing factor mutations lead to aberrant pre-mRNA splicing of many genes, some of which have been shown in functional studies to impact on hematopoiesis and to contribute to the MDS phenotype. This clearly demonstrates that impaired spliceosome function plays an important role in MDS pathophysiology. Recent studies that harnessed the power of induced pluripotent stem cell (iPSC) and CRISPR/Cas9 gene editing technologies to generate new iPSC-based models of splicing factor mutant MDS, have further illuminated the role of key downstream target genes. The aberrantly spliced genes and the dysregulated pathways associated with splicing factor mutations in MDS represent potential new therapeutic targets. Emerging data has shown that IRAK4 is aberrantly spliced in SF3B1 and U2AF1 mutant MDS, leading to hyperactivation of NF-κB signaling. Pharmacological inhibition of IRAK4 has shown efficacy in pre-clinical studies and in MDS clinical trials, with higher response rates in patients with splicing factor mutations. Our increasing knowledge of the effects of splicing factor mutations in MDS is leading to the development of new treatments that may benefit patients harboring these mutations.


Subject(s)
DNA, Recombinant , Myelodysplastic Syndromes , Humans , RNA Splicing Factors/genetics , DNA, Recombinant/metabolism , DNA, Recombinant/pharmacology , DNA, Recombinant/therapeutic use , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/pharmacology , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism , Myelodysplastic Syndromes/genetics , Spliceosomes/genetics , RNA Splicing , Mutation
2.
Buenos Aires; CONETEC; nov. 2019.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1048252

ABSTRACT

INTRODUCCIÓN: La hipofosfatasia (HPP) es una enfermedad hereditaria "ultra rara" sumamente heterogénea, del metabolismo fosfo-cálcico generada por un déficit enzimático. Se presenta con múltiples formas clínicas, que van desde alteraciones dentales hasta falta de mineralización ósea generalizada, que lleva a una alta tasa de mortalidad. El pronóstico empeora cuanto más precoz es el inicio de los síntomas (formas perinatales e infantil). No se encuentran disponibles datos de prevalencia locales; se estima una incidencia de aproximadamente 2 casos nuevos de las formas severas cada año en Argentina. DESCRIPCIÓN DE LA TECNOLOGÍA: Asfotase alfa es una enzima de reemplazo diseñada para suplementar la actividad de la TN-SALP, la misma es obtenida por técnicas de ADN recombinante utilizando células de ovario de hámster chino. Se administra de forma subcutánea, la dosis recomendada por el fabricante es de 2 mg/kg tres veces por semana o 1 mg/kg seis veces por semana, aunque se ha utilizado en dosis superiores en los estudios encontrados. METODOLOGÍA: Se realizó una búsqueda en Medline, Lilacs, Cochrane, Tripdatabase, ClínicalTrials.gov, Orphanet y buscadores genéricos de internet. Se utilizaron las palabras clave "asfotase", "hypophosphatasia AND enzyme replacement", "asfotase AND hypophosphatasia", "strensiq". Se consideraron criterios de inclusión a ensayos clínicos controlados aleatorizados (ECCAs), ensayos clínicos no controlados, cohortes prospectivas, casos y controles y estudios de fase 2 o más avanzado, realizados en humanos; en idioma inglés, español, francés o portugués. RESULTADOS: Para la forma clínica perinatal severa e infantil, evidencia de muy baja calidad basada en estudios de fase 2 mostró un beneficio considerable para los desenlaces sobrevida, soporte ventilatorio y cambios radiológicos. Los costos directos anuales de la tecnología para el tratamiento de esta forma clínica se estimaron entre US$ 178.308 y US$ 217.620. En el caso de la forma clínica infanto-juvenil se incluyó un estudio de fase 2 donde se valoró un beneficio considerable para el desenlace cambios radiológicos. Sin embargo, el efecto del tratamiento fue incierto para los desenlaces sobrevida y calidad de vida. La calidad de la evidencia hallada en este caso también fue muy baja. Finalmente, para la forma clínica adulta, en el estudio de fase 2 incluido, no observó beneficio para los desenlaces mineralización ósea y dolor; y un efecto incierto del tratamiento para los desenlaces sobrevida y calidad de vida. La calidad de la evidencia hallada fue baja. CONCLUSIÓN: La información de este reporte contiene las opiniones y perspectivas de una cuidadora de un paciente pediátrico sobre la evolución de la enfermedad hasta llegar al tratamiento, y sobre la vida cotidiana. En el caso de este paciente de 6 años, con diagnostico a los 8 meses de vida, que recibe asfotase alfa hace 3 años, el tratamiento presentó buenos resultados en cuanto al dolor y al desarrollo de la marcha.


Subject(s)
Humans , DNA, Recombinant/therapeutic use , Enzyme Replacement Therapy , Hypophosphatasia/drug therapy , Technology Assessment, Biomedical , Cost-Benefit Analysis/economics
4.
J Biol Chem ; 292(44): 18062-18074, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28928221

ABSTRACT

Parkinson's disease (PD) is one of the most epidemic neurodegenerative diseases and is characterized by movement disorders arising from loss of midbrain dopaminergic (DA) neurons. Recently, the relationship between PD and autophagy has received considerable attention, but information about the mechanisms involved is lacking. Here, we report that autophagy-related gene 5 (ATG5) is potentially important in protecting dopaminergic neurons in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model in zebrafish. Using analyses of zebrafish swimming behavior, in situ hybridization, immunofluorescence, and expressions of genes and proteins related to PD and autophagy, we found that the ATG5 expression level was decreased and autophagy flux was blocked in this model. The ATG5 down-regulation led to the upgrade of PD-associated proteins, such as ß-synuclein, Parkin, and PINK1, aggravation of MPTP-induced PD-mimicking pathological locomotor behavior, DA neuron loss labeled by tyrosine hydroxylase (TH) or dopamine transporter (DAT), and blocked autophagy flux in the zebrafish model. ATG5 overexpression alleviated or reversed these PD pathological features, rescued DA neuron cells as indicated by elevated TH/DAT levels, and restored autophagy flux. The role of ATG5 in protecting DA neurons was confirmed by expression of the human atg5 gene in the zebrafish model. Our findings reveal that ATG5 has a role in neuroprotection, and up-regulation of ATG5 may serve as a goal in the development of drugs for PD prevention and management.


Subject(s)
Autophagy-Related Protein 5/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Genetic Therapy , Parkinsonian Disorders/prevention & control , Zebrafish Proteins/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/therapeutic use , Behavior, Animal/drug effects , Biomarkers/metabolism , Brain/cytology , Brain/metabolism , Brain/pathology , Cell Line, Tumor , DNA, Recombinant/therapeutic use , Dopaminergic Neurons/cytology , Dopaminergic Neurons/pathology , Embryo, Nonmammalian , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Larva , Microinjections , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/therapeutic use , Neuroprotection/drug effects , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
5.
Ann N Y Acad Sci ; 1407(1): 5-16, 2017 11.
Article in English | MEDLINE | ID: mdl-28905423

ABSTRACT

The most advanced regulatory processes for complex biological products have been put in place in many countries to provide appropriate regulatory oversight of biotherapeutic products in general, and similar biotherapeutics in particular. This process is still ongoing and requires regular updates to national regulatory requirements in line with scientific developments and up-to-date standards. For this purpose, strong knowledge of and expertise in evaluating biotherapeutics in general and similar biotherapeutic products, also called biosimilars, in particular is essential. Here, we discuss the World Health Organization's international standard-setting role in the regulatory evaluation of recombinant DNA-derived biotherapeutic products, including biosimilars, and provide examples that may serve as models for moving forward with nonbiological complex medicinal products. A number of scientific challenges and regulatory considerations imposed by the advent of biosimilars are described, together with the lessons learned, to stimulate future discussions on this topic. In addition, the experiences of facilitating the implementation of guiding principles for evaluation of similar biotherapeutic products into regulatory and manufacturers' practices in various countries over the past 10 years are briefly explained, with the aim of promoting further developments and regulatory convergence of complex biological and nonbiological products.


Subject(s)
Biological Products/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , DNA, Recombinant/therapeutic use , Drug Industry/standards , Drug Evaluation/standards , Drug Industry/trends , Drug Therapy/methods , Drug Therapy/standards , Drug Therapy/trends , Guidelines as Topic/standards , Humans , World Health Organization
6.
Curr Top Med Chem ; 17(13): 1529-1541, 2017.
Article in English | MEDLINE | ID: mdl-28017152

ABSTRACT

Protein bioconjugates can be synthesized by using chemical reactions, enzymatic reactions or genetic engineering technologies. Naturally occurred protein fusion event is used on purpose in the development of better biopharmaceuticals by applying genetic engineering methodologies. This review will mainly focus on the types of fusion proteins produced with the use of recombinant DNA technology, by combining genes or parts of genes from the same or different organisms, in order to be used in pharmaceutical applications for several purposes. Main concerns for the development of better biopharmaceuticals include quality, efficacy, safety, immunogenicity and toxicity issues. Extending half-life of the drug to increase patient compliance, targeting the drugs to reduce toxicity, improving the manufacturing environment to reduce the costs and revealing protein interaction technologies to find novel and superior drugs are the main aims of fusion protein production. Here, related tags and examples of fusion methods for different purposes will be explained precisely.


Subject(s)
DNA, Recombinant/metabolism , Pharmaceutical Preparations/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Animals , DNA, Recombinant/chemistry , DNA, Recombinant/therapeutic use , Humans , Pharmaceutical Preparations/chemistry , Recombinant Fusion Proteins/chemistry
7.
Cancer Control ; 23(1): 12-20, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27009452

ABSTRACT

BACKGROUND: Locoregional advanced melanoma poses a complex clinical challenge that requires a multidisciplinary, patient-centered approach. Numerous agents have been studied for their suitability as intralesional therapy in the past decades, but few have successfully completed phase 3 clinical trial testing. METHODS: The relevant medical literature was searched for articles regarding use of intralesional therapies in metastatic melanoma. Therapies with data from phase 2 or higher studies were selected for review. This review also summarizes the mechanisms of action, adverse-event profiles, and clinical data for these agents. RESULTS: Intralesional therapies demonstrate promising effects in select patients with advanced melanoma. The optimal approach should be individually tailored and consist of a combination of intralesional therapies, regional perfusions, systemic immunotherapies, targeted therapies, and surgery, if necessary. CONCLUSIONS: Due to its relatively good local response rates and tolerable adverse-event profile, intralesional therapy may be a treatment option for select patients with unresectable, locally advanced or metastatic melanoma.


Subject(s)
Genetic Therapy , Immunotherapy , Injections, Intralesional/methods , Melanoma/therapy , Oncolytic Viruses , Skin Neoplasms/therapy , Administration, Cutaneous , BCG Vaccine/administration & dosage , BCG Vaccine/adverse effects , BCG Vaccine/therapeutic use , DNA, Recombinant/administration & dosage , DNA, Recombinant/therapeutic use , Electrochemotherapy/methods , Fluorescent Dyes/administration & dosage , Fluorescent Dyes/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , HLA-B7 Antigen/genetics , Herpesvirus 1, Human , Humans , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Lipids/administration & dosage , Lipids/therapeutic use , Melanoma/genetics , Rose Bengal/administration & dosage , Rose Bengal/therapeutic use , Skin Neoplasms/genetics
8.
Expert Opin Biol Ther ; 16(5): 619-26, 2016.
Article in English | MEDLINE | ID: mdl-26898656

ABSTRACT

INTRODUCTION: Intralesional immunotherapy supplements systemic treatments and often achieves higher remission rates as compared to systemic therapy. Its indication is metastatic melanoma with limited tumor burden, particularly in loco-regional metastasis and distant soft tissue metastasis. AREAS COVERED: This review describes intralesional immunotherapy with talimogene laherparepvec (T-VEC), with velimogene aliplasmid (Allovectin-7) and with intralesional interleukin-2. These therapies function exclusively by activating the immune system. Furthermore, Rose Bengal and electrochemotherapy have been included, as bystander effects have been observed with these treatments. EXPERT OPINION: Objective remissions are achieved in a higher percentage with intralesional immunotherapies, such as intralesional interleukin-2 with up to 69% of complete remissions, as compared to systemic treatment. Therefore, intralesional immunotherapy may act as supplement in the armament against metastatic melanoma. In particular, for patients with multiple cutaneous and subcutaneous metastases (20­≥ 100) and in patients with subcutaneous bulky disease intralesional immunotherapy can improve the disease outcome. The exact role of intralesional immunotherapy in the age of immune checkpoint blockade has still to be determined. A number of clinical trials are on the way in order to better understand synergistic actions of intralesional and systemic immunotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Immunotherapy , Melanoma/drug therapy , Melanoma/pathology , Skin Neoplasms/drug therapy , DNA, Recombinant/therapeutic use , Humans , Injections, Intralesional , Lipids/therapeutic use , Oncolytic Virotherapy , Remission Induction , Skin Neoplasms/pathology
9.
Methods Mol Biol ; 1349: 151-61, 2016.
Article in English | MEDLINE | ID: mdl-26458835

ABSTRACT

The ability to manipulate capripoxvirus through gene knockouts and gene insertions has become an increasingly valuable research tool in elucidating the function of individual genes of capripoxvirus, as well as in the development of capripoxvirus-based recombinant vaccines. The homologous recombination technique is used to generate capripoxvirus knockout viruses (KO), and is based on the targeting a particular viral gene of interest. This technique can also be used to insert a gene of interest. A protocol for the generation of a viral gene knockout is described. This technique involves the use of a plasmid which encodes the flanking sequences of the regions where the homologous recombination will occur, and will result in the insertion of an EGFP reporter gene for visualization of recombinant virus, as well as the E. coli gpt gene as a positive selection marker. If an additional gene is to be incorporated, this can be achieved by inserting a gene of interest for expression under a poxvirus promoter into the plasmid between the flanking regions for insertion. This chapter describes a protocol for generating such recombinant capripoxviruses.


Subject(s)
Capripoxvirus/genetics , DNA, Recombinant/genetics , Gene Knockout Techniques/methods , Vaccines, Synthetic/genetics , Capripoxvirus/immunology , Cloning, Molecular , DNA, Recombinant/immunology , DNA, Recombinant/therapeutic use , Escherichia coli/genetics , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Promoter Regions, Genetic , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
10.
Adv Exp Med Biol ; 871: 73-85, 2015.
Article in English | MEDLINE | ID: mdl-26374213

ABSTRACT

Advanced therapy medicinal products, a new class of products with promising therapeutic effects, have been classified as medicinal products and as such should be developed according to a well-structured development plan, to establish their quality, safety and efficacy profile and conclude, at the time of the marketing authorisation evaluation, on a positive risk/benefit balance for patients. An important part of this development plan is achieved through clinical trials, which have also to be approved according to a well-established regulatory process, prior any initiation. This chapter is dedicated to describe the regulatory pathway to be followed in France, before initiating any clinical trial with those investigational advanced therapy medicinal products. In France, to get the final authorisation to initiate a clinical trial, the legislation imposes to run in parallel two independent but complementary authorisation procedures. The first procedure is aimed at assessing the ethical aspect of the biomedical research, while the second has to review the safety and regulatory aspects. A third procedure has to be envisaged where in case the investigational product consists or contains a genetically modified organism. The French system herein described is in line with the EU regulation on clinical trial and follows the respective deadlines for granting the final approval. The complexity of the procedure is in fact more due to the complexity of the products and protocols to be assessed than to the procedure itself which is now very close to the well-known procedure applied routinely for more conventional chemical or biological candidate medicinal products.


Subject(s)
Cell- and Tissue-Based Therapy/ethics , Drug Approval/legislation & jurisprudence , Drug and Narcotic Control/legislation & jurisprudence , Genetic Therapy/legislation & jurisprudence , Translational Research, Biomedical/legislation & jurisprudence , Animals , Biological Products/therapeutic use , Cell- and Tissue-Based Therapy/methods , Clinical Trials as Topic , DNA, Recombinant/therapeutic use , France , Genetic Therapy/ethics , Genetic Vectors/therapeutic use , Humans , Investigational New Drug Application/legislation & jurisprudence , Patient Safety/legislation & jurisprudence , Practice Guidelines as Topic , Research Design , Translational Research, Biomedical/ethics
11.
Adv Exp Med Biol ; 871: 31-47, 2015.
Article in English | MEDLINE | ID: mdl-26374211

ABSTRACT

The National Institutes of Health (NIH) oversight of human gene transfer research, which is defined as the deliberate transfer of recombinant and/or synthetic nucleic acid molecules to humans, originates with the NIH Guidelines for Research Involving Recombinant or Synthetic Nucleic Acid Molecules (NIH Guidelines). The NIH Guidelines, which were first published in the Federal Register almost 40 years ago, have been amended numerous times to remain responsive to scientific progress and to clearly define the responsibilities of NIH, the Recombinant DNA Advisory Committee (RAC), investigators, and institutions. Human gene transfer trials conducted at clinical sites in the United States (USA) are subject to the NIH Guidelines if they are conducted at, or sponsored by, an institution that receives any support for recombinant or synthetic nucleic acid research from the NIH. Human gene transfer trials conducted either in the USA or abroad are also subject to the NIH Guidelines if the investigational agent was developed with NIH funds and the institution that developed the investigational materials sponsors or participates in these projects. Trials are registered with the NIH Office Biotechnology Activities (OBA) and there are ongoing reporting requirements. Each new trial is reviewed by the RAC, and those that are novel or raise unique ethical or social issues are selected for review at quarterly public RAC meetings. The RAC also advises the NIH on policy and other matters relating to clinical gene transfer research and biosafety.


Subject(s)
DNA, Recombinant/therapeutic use , Drug and Narcotic Control/legislation & jurisprudence , Genetic Therapy/legislation & jurisprudence , Genetic Vectors/therapeutic use , Translational Research, Biomedical/legislation & jurisprudence , Animals , Clinical Trials as Topic , Genetic Therapy/ethics , Humans , Informed Consent/ethics , Informed Consent/legislation & jurisprudence , National Institutes of Health (U.S.) , Patient Safety/legislation & jurisprudence , Practice Guidelines as Topic , Research Design , Translational Research, Biomedical/ethics , United States
12.
Adv Exp Med Biol ; 871: 49-71, 2015.
Article in English | MEDLINE | ID: mdl-26374212

ABSTRACT

Health Canada regulates gene therapy products and many cell therapy products as biological drugs under the Canadian Food and Drugs Act and its attendant regulations. Cellular products that meet certain criteria, including minimal manipulation and homologous use, may be subjected to a standards-based approach under the Safety of Human Cells, Tissues and Organs for Transplantation Regulations. The manufacture and clinical testing of cell and gene therapy products (CGTPs) presents many challenges beyond those for protein biologics. Cells cannot be subjected to pathogen removal or inactivation procedures and must frequently be administered shortly after final formulation. Viral vector design and manufacturing control are critically important to overall product quality and linked to safety and efficacy in patients through concerns such as replication competence, vector integration, and vector shedding. In addition, for many CGTPs, the value of nonclinical studies is largely limited to providing proof of concept, and the first meaningful data relating to appropriate dosing, safety parameters, and validity of surrogate or true determinants of efficacy must come from carefully designed clinical trials in patients. Addressing these numerous challenges requires application of various risk mitigation strategies and meeting regulatory expectations specifically adapted to the product types. Regulatory cooperation and harmonisation at an international level are essential for progress in the development and commercialisation of these products. However, particularly in the area of cell therapy, new regulatory paradigms may be needed to harness the benefits of clinical progress in situations where the resources and motivation to pursue a typical drug product approval pathway may be lacking.


Subject(s)
Cell- and Tissue-Based Therapy/ethics , Drug and Narcotic Control/legislation & jurisprudence , Genetic Therapy/legislation & jurisprudence , Translational Research, Biomedical/legislation & jurisprudence , Animals , Biological Products/therapeutic use , Canada , Cell- and Tissue-Based Therapy/methods , DNA, Recombinant/therapeutic use , Drug Approval , Genetic Therapy/ethics , Genetic Vectors/therapeutic use , Humans , Patient Safety/legislation & jurisprudence , Practice Guidelines as Topic , Research Design , Translational Research, Biomedical/ethics , Validation Studies as Topic
13.
Ann N Y Acad Sci ; 1350: 17-28, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26273800

ABSTRACT

Among the five enzyme complexes in the oxidative phosphorylation system, NADH-coenzyme Q oxidoreductase (also called complex I) is the largest, most intricate, and least understood. This enzyme complex spans the inner mitochondrial membrane and catalyzes the first step of electron transfer by the oxidation of NADH, and thereby provides two electrons for the reduction of quinone to quinol. Complex I deficiency is associated with many severe mitochondrial diseases, including Leber hereditary optic neuropathy and Leigh syndrome. However, to date, conventional treatments for the majority of genetic mitochondrial diseases are only palliative. Developing a reliable and convenient therapeutic approach is therefore considered to be an urgent need. Targeted proteins fused with the protein transduction domain of human immunodeficiency virus 1 transactivator of transcription (TAT) have been shown to enter cells by crossing plasma membranes while retaining their biological activities. Recent developments show that, in fusion with mitochondrial targeting sequences (MTSs), TAT-MTS-bound cargo can be correctly transported into mitochondria and restore the missing function of the cargo protein in patients' cells. The available evidence suggests that the TAT-mediated protein transduction system holds great promise as a potential therapeutic approach to treat complex I deficiency, as well as other mitochondrial diseases.


Subject(s)
DNA, Mitochondrial/metabolism , Electron Transport Complex I/deficiency , Electron Transport Complex I/metabolism , Mitochondrial Diseases/metabolism , Models, Biological , Protein Sorting Signals , Signal Transduction , tat Gene Products, Human Immunodeficiency Virus/metabolism , Animals , DNA, Mitochondrial/therapeutic use , DNA, Recombinant/metabolism , DNA, Recombinant/therapeutic use , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Electron Transport Complex I/therapeutic use , Enzyme Replacement Therapy/methods , HIV-1/metabolism , Humans , Mitochondrial Diseases/genetics , Oxidative Phosphorylation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Targeted Gene Repair/methods , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/genetics
14.
PLoS Pathog ; 11(5): e1004907, 2015 May.
Article in English | MEDLINE | ID: mdl-25996983

ABSTRACT

Herpes simplex virus (HSV) enters cells by means of four essential glycoproteins - gD, gH/gL, gB, activated in a cascade fashion by gD binding to one of its receptors, nectin1 and HVEM. We report that the engineering in gH of a heterologous ligand - a single-chain antibody (scFv) to the cancer-specific HER2 receptor - expands the HSV tropism to cells which express HER2 as the sole receptor. The significance of this finding is twofold. It impacts on our understanding of HSV entry mechanism and the design of retargeted oncolytic-HSVs. Specifically, entry of the recombinant viruses carrying the scFv-HER2-gH chimera into HER2+ cells occurred in the absence of gD receptors, or upon deletion of key residues in gD that constitute the nectin1/HVEM binding sites. In essence, the scFv in gH substituted for gD-mediated activation and rendered a functional gD non-essential for entry via HER2. The activation of the gH moiety in the chimera was carried out by the scFv in cis, not in trans as it occurs with wt-gD. With respect to the design of oncolytic-HSVs, previous retargeting strategies were based exclusively on insertion in gD of ligands to cancer-specific receptors. The current findings show that (i) gH accepts a heterologous ligand. The viruses retargeted via gH (ii) do not require the gD-dependent activation, and (iii) replicate and kill cells at high efficiency. Thus, gH represents an additional tool for the design of fully-virulent oncolytic-HSVs retargeted to cancer receptors and detargeted from gD receptors.


Subject(s)
DNA, Recombinant/metabolism , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/metabolism , Simplexvirus/physiology , Single-Chain Antibodies/metabolism , Viral Envelope Proteins/metabolism , Viral Tropism , Virus Internalization , Animals , Binding Sites , Cell Line , Cell Survival , DNA, Recombinant/chemistry , DNA, Recombinant/therapeutic use , Genetic Therapy , Humans , Ligands , Mutation , Neoplasms/metabolism , Neoplasms/therapy , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Protein Engineering , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Receptor, ErbB-2/therapeutic use , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Signal Transduction , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/therapeutic use
15.
Adv Exp Med Biol ; 848: 131-48, 2015.
Article in English | MEDLINE | ID: mdl-25757619

ABSTRACT

There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous ß-herpes virus that infects 60-95 % of adults worldwide. Infection is a major cause of congenital abnormalities in newborns, contributes to development of childhood cerebral palsy and medulloblastoma, can result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. While CMV has been increasingly associated with numerous inflammatory diseases and cancers, only recently has it been correlated with increased risk of heart disease in adults, the number-one killer in the USA. These data, among others, suggest that subclinical CMV infection, or microinfection, in healthy individuals may play more of a causative role than an epiphenomenon in development of CMV-associated pathologies. Due to the myriad of diseases and complications associated with CMV, an efficacious vaccine would be highly valuable in reducing human morbidity and mortality as well as saving billions of dollars in annual health-care costs and disability adjusted life years (DALY) in the developing world. Therefore, the development of a safe efficacious CMV vaccine or immune therapy is paramount to the public health. This review aims to provide a brief overview on aspects of CMV infection and disease and focuses on current vaccine strategies. The use of new synthetic DNA vaccines might offer one such approach to this difficult problem.


Subject(s)
Cloning, Molecular/methods , Cytomegalovirus Infections/therapy , Cytomegalovirus Vaccines/therapeutic use , Immunotherapy, Active/methods , Vaccines, DNA/therapeutic use , Adult , Animals , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/genetics , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Humans , Vaccines, DNA/genetics
16.
Immunol Cell Biol ; 91(10): 642-51, 2013.
Article in English | MEDLINE | ID: mdl-24145856

ABSTRACT

Recombinant DNA vaccines represent a novel method for generating in situ expression of vaccine antigens. Intramuscular injections of naked DNA are able to elicit potent humoral and cellular immune responses but still numerous factors limit the immunogenicity of DNA vaccines. Co-expression of cytokines with antigen encoding genes in DNA vectors can improve the immune responses and modify Th1/Th2 balance. In this study, the immunomodulatory effect of Interleukin 22 (IL-22) as an adjuvant was studied by DNA vaccination with S. Typhi Heat shock protein 60 (HSP60/GroEL) in mice. Further, DNA construct of IL-22 gene fused with GroEL was developed and immunization studies were carried out in mice. DNA vaccination with GroEL alone stimulated humoral and cell-mediated immune responses. Co-immunization (IL-22+GroEL) further resulted in increase in T-cell proliferative responses, antibody titres (IgG, IgG1, IgG2a) and secretion of IFNγ (Th1), IL-1ß and Th2 (IL-4, IL-6) cytokines. Co-expression (IL-22-GroEL DNA) also promoted antibody titres and cytokine levels were significantly higher as compared to co-immunized group. A reduction in bacterial load in spleen, liver and intestine was seen in all the immunized groups as compared to control, with least organ burden in fusion DNA construct group (co-expression). Improved protective efficacy (90%) against lethal challenge by Salmonella was observed with IL-22-GroEL co-expressing DNA vector as compared with plasmid encoding GroEL only (50-60%) or co-immunization group (75-80%). This study thus shows that co-expression of IL-22 and GroEL genes enhances the immune responses and protective efficacy, circumventing the need of any adjuvant.


Subject(s)
Chaperonin 60/genetics , Immunity/genetics , Interleukins/genetics , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Animals , Antibody Formation/immunology , Bacterial Load/immunology , Cell Proliferation , Cytokines/metabolism , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Female , Gene Expression , Genetic Vectors/metabolism , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Protein Biosynthesis , Salmonella Infections, Animal/drug therapy , Salmonella Infections, Animal/prevention & control , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transcription, Genetic , Treatment Outcome , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Interleukin-22
18.
Hum Reprod ; 28(9): 2398-406, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820419

ABSTRACT

STUDY QUESTION: Is targeted adenovirus vector, Ad-SSTR-RGD-TK (Adenovirus -human somatostatin receptor subtype 2- arginine, glycine and aspartate-thymidine kinase), given in combination with ganciclovir (GCV) against immortalized human leiomyoma cells (HuLM) a potential therapy for uterine fibroids? SUMMARY ANSWER: Ad-SSTR-RGD-TK/GCV, a targeted adenovirus, effectively reduces cell growth in HuLM cells and to a significantly greater extent than in human uterine smooth muscle cells (UtSM). WHAT IS KNOWN ALREADY: Uterine fibroids (leiomyomas), a major cause of morbidity and the most common indication for hysterectomy in premenopausal women, are well-defined tumors, making gene therapy a suitable and potentially effective non-surgical approach for treatment. Transduction of uterine fibroid cells with adenoviral vectors such as Ad-TK/GCV (herpes simplex virus thymidine kinase gene) decreases cell proliferation. STUDY DESIGN, SIZE, DURATION: An in vitro cell culture method was set up to compare and test the efficacy of a modified adenovirus vector with different multiplicities of infection in two human immortalized cell lines for 5 days. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immortalized human leiomyoma cells and human uterine smooth muscle cells were infected with different multiplicities of infection (MOI) (5-100 plaque-forming units (pfu)/cell) of a modified Ad-SSTR-RGD-TK vector and subsequently treated with GCV. For comparison, HuLM and UtSM cells were transfected with Ad-TK/GCV and Ad-LacZ/GCV. Cell proliferation was measured using the CyQuant assay in both cell types. Additionally, western blotting was used to assess the expression of proteins responsible for regulating proliferation and apoptosis in the cells. MAIN RESULTS AND THE ROLE OF CHANCE: Transduction of HuLM cells with Ad-SSTR-RGD-TK/GCV at 5, 10, 50 and 100 pfu/cell decreased cell proliferation by 28, 33, 45, and 84%, respectively (P < 0.05) compared with untransfected cells, whereas cell proliferation in UtSM cells transfected with the same four MOIs of Ad-SSTR-RGD-TK/GCV compared with that of untransfected cells was decreased only by 8, 23, 25, and 28%, respectively (P < 0.01). Western blot analysis showed that, in comparison with the untargeted vector Ad-TK, Ad-SSTR-RGD-TK/GCV more effectively reduced expression of proteins that regulate the cell cycle (Cyclin D1) and proliferation (PCNA, Proliferating Cell Nuclear Antigen), and it induced expression of the apoptotic protein BAX, in HuLM cells. LIMITATIONS, REASONS FOR CAUTION: Results from this study need to be replicated in an appropriate animal model before testing this adenoviral vector in a human trial. WIDER IMPLICATIONS OF THE FINDINGS: Effective targeting of gene therapy to leiomyoma cells enhances its potential as a non-invasive treatment of uterine fibroids.


Subject(s)
DNA, Recombinant/metabolism , Gene Expression Regulation, Neoplastic , Genetic Vectors/metabolism , Leiomyoma/metabolism , Myometrium/metabolism , Transduction, Genetic , Uterine Neoplasms/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , DNA, Recombinant/adverse effects , DNA, Recombinant/therapeutic use , Female , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/adverse effects , Genetic Vectors/therapeutic use , Humans , Leiomyoma/therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Uterine Neoplasms/therapy
19.
Pharmacol Rep ; 65(5): 1075-85, 2013.
Article in English | MEDLINE | ID: mdl-24399704

ABSTRACT

Pharmaceutical biotechnology has a long tradition and is rooted in the last century, first exemplified by penicillin and streptomycin as low molecular weight biosynthetic compounds. Today, pharmaceutical biotechnology still has its fundamentals in fermentation and bioprocessing, but the paradigmatic change affected by biotechnology and pharmaceutical sciences has led to an updated definition. The biotechnology revolution redrew the research, development, production and even marketing processes of drugs. Powerful new instruments and biotechnology related scientific disciplines (genomics, proteomics) make it possible to examine and exploit the behavior of proteins and molecules. Recombinant DNA (rDNA) technologies (genetic, protein, and metabolic engineering) allow the production of a wide range of peptides, proteins, and biochemicals from naturally nonproducing cells. This technology, now approximately 25 years old, is becoming one of the most important technologies developed in the 20(th) century. Pharmaceutical products and industrial enzymes were the first biotech products on the world market made by means of rDNA. Despite important advances regarding rDNA applications in mammalian cells, yeasts still represent attractive hosts for the production of heterologous proteins. In this review we describe these processes.


Subject(s)
Biotechnology/methods , DNA, Recombinant/biosynthesis , Genetic Engineering , Recombinant Proteins/biosynthesis , Animals , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Darbepoetin alfa , Erythropoietin/analogs & derivatives , Erythropoietin/biosynthesis , Erythropoietin/therapeutic use , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/therapeutic use , Genomics , Hematinics/metabolism , Hematinics/therapeutic use , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Insulin, Regular, Human/biosynthesis , Insulin, Regular, Human/therapeutic use , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Proteomics , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Somatostatin/biosynthesis , Somatostatin/therapeutic use , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/therapeutic use
20.
Cancer Gene Ther ; 19(12): 811-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23037806

ABSTRACT

Allovectin (velimogene aliplasmid) is a cancer immunotherapeutic currently completing a pivotal phase 3 study for metastatic melanoma. Consisting of a bicistronic plasmid encoding both major histocompatibility complex (MHC) class I heavy and light chains (HLA-B7 and ß2-microglobulin, respectively) formulated with a cationic lipid-based system, it is designed for direct intratumoral administration. Following injection into a single lesion, the product is intended to induce anti-tumor immune responses against both treated and distal lesions. Both the plasmid and lipid components of Allovectin contribute to the biological activity of the drug product, and its therapeutic activity is hypothesized to derive from multiple mechanisms of actions (MOAs). These include the induction of both cytotoxic T-cell and innate immune responses directed against allogeneic as well as tumor-derived targets, consequences of both an increased MHC class I expression on tumor cells and the induction of a localized immune/inflammatory response. In this paper, we review Allovectin's proposed MOAs, placing their contributions in the context of anti-tumor immunity and highlighting both preclinical and clinical supporting data.


Subject(s)
DNA, Recombinant/therapeutic use , Lipids/therapeutic use , Melanoma/drug therapy , Melanoma/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Animals , DNA, Recombinant/pharmacology , HLA-B7 Antigen/immunology , Humans , Immunotherapy/methods , Lipids/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL