Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
EMBO Mol Med ; 16(4): 870-884, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38462666

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is a rare inherited skin disease characterized by defects in type VII collagen leading to a range of fibrotic pathologies resulting from skin fragility, aberrant wound healing, and altered dermal fibroblast physiology. Using a novel in vitro model of fibrosis based on endogenously produced extracellular matrix, we screened an FDA-approved compound library and identified antivirals as a class of drug not previously associated with anti-fibrotic action. Preclinical validation of our lead hit, daclatasvir, in a mouse model of RDEB demonstrated significant improvement in fibrosis as well as overall quality of life with increased survival, weight gain and activity, and a decrease in pruritus-induced hair loss. Immunohistochemical assessment of daclatasvir-treated RDEB mouse skin showed a reduction in fibrotic markers, which was supported by in vitro data demonstrating TGFß pathway targeting and a reduction of total collagen retained in the extracellular matrix. Our data support the clinical development of antivirals for the treatment of patients with RDEB and potentially other fibrotic diseases.


Subject(s)
Carbamates , Epidermolysis Bullosa Dystrophica , Imidazoles , Pyrrolidines , Valine/analogs & derivatives , Humans , Animals , Mice , Epidermolysis Bullosa Dystrophica/drug therapy , Epidermolysis Bullosa Dystrophica/pathology , Quality of Life , Collagen Type VII/metabolism , Collagen Type VII/therapeutic use , Fibrosis , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Skin/metabolism , Skin/pathology
2.
Brasília; CONITEC; nov. 2022.
Non-conventional in Portuguese | BRISA/RedTESA | ID: biblio-1443191

ABSTRACT

CONDIÇÃO CLÍNICA: A Epidermólise Bolhosa (EB) é uma doença congênita, não contagiosa pertencente a um grupo de doenças cutâneas geneticamente transmitidas, cuja principal característica é a formação de bolhas após trauma mínimo espontâneo ou mecânico. Alguns indivíduos podem apresentar deformidades das mãos e nos pés (pseudosindactilia), anemia ferropriva, perdas de unhas e dentes, escaras na córnea, atrasos de desenvolvimento devido à desnutrição e risco de desenvolvimento de câncer nas lesões crônicas. A EB é causada por mutações em pelo menos 20 genes diferentes, sendo os KRT5, KRT14, PLEC e COL17A1 os principais genes citados na literatura. Ademais, sua classificação é complexa, porque mutações nesses mesmos genes podem resultar em fenótipos clínicos distintos. As mutações causam a ausência ou a diminuição da codificação de proteínas estruturais podendo levar a redução da resistência da pele à tração da ferida. TRATAMENTO: O tratamento atual da EB é principalmente preventivo e de suporte, incluindo proteção contra forças mecânicas evitando fricção, tratamento precoce de feridas para prevenir infecções e proteção da ferida com curativos não adesivos adequados para permitir a cicatrização. O Protocolo Clínico e Diretrizes Terapêuticas (PCDT) da EB, publicado em 2021, descreve os principais tratamentos para os pacientes com essa condição. As medidas terapêuticas da EB inclui terapia medicamentosa e não medicam


Subject(s)
Humans , Triterpenes/therapeutic use , Cannabinol/therapeutic use , Keratinocytes , Anthraquinones/therapeutic use , Epidermolysis Bullosa/drug therapy , Collagen Type VII/therapeutic use , Technological Development and Innovation Projects , Mesenchymal Stem Cells , Fibroblasts , Brazil , Efficacy , Cost-Benefit Analysis/economics
3.
Exp Dermatol ; 29(11): 1112-1122, 2020 11.
Article in English | MEDLINE | ID: mdl-33043517

ABSTRACT

Epidermolysis bullosa (EB) is the prototypical example of genetic skin fragility disorders. Genotypic heterogeneity, modifier genes, epigenetic, biochemical and environmental factors alter and determine pathogenic traits and, ultimately, the wide and striking phenotypic variability in EB. Besides the primary structural-functional defect, chronic tissue damage with induction and dysregulation of inflammatory pathways is a common pathogenic mechanism in EB. In localized variants, the inflammatory aberrations may mainly affect the micromilieu of lesional skin, while a systemic inflammatory response was shown to contribute to the systemic morbidity in severe EB subtypes with extensive cutaneous involvement. Our continued understanding of the pathophysiology of EB, as well as advances in molecular technologies, has paved the way for translational therapeutic approaches. The spectrum comprises of corrective and symptom-relieving therapies that include innovative therapeutic options garnered from the bench, repurposed drugs approved for other diseases, as well as strategies for gene-, protein- and cell-based therapies. Immunological traits further define new targets of therapy, aimed at improving skin barrier restoration, microbial surveillance and infection control, wound healing and anti-neoplastic effects. Clinical availability and feasibility of these approaches for all EB patients and subtypes are currently limited, reflecting issues of efficacy, specificity, tolerability and safety. A multistep targeting approach and highly individualized, risk-stratified combinatory treatment plans will thus be essential for sustained efficacy and improved overall quality of life in EB.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Epidermolysis Bullosa/therapy , Genetic Therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/etiology , Cell- and Tissue-Based Therapy , Clinical Trials as Topic , Codon, Nonsense , Collagen Type VII/therapeutic use , Epidermolysis Bullosa/complications , Epidermolysis Bullosa/genetics , Humans , Immunotherapy , Molecular Targeted Therapy , Pain/drug therapy , Pain/etiology , Pruritus/drug therapy , Pruritus/etiology , Recombinant Proteins/therapeutic use , Skin Neoplasms/etiology , Translational Research, Biomedical
4.
JAMA ; 316(17): 1808-1817, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27802546

ABSTRACT

Importance: Recessive dystrophic epidermolysis bullosa (RDEB) is a devastating, often fatal, inherited blistering disorder caused by mutations in the COL7A1 gene encoding type VII collagen. Support and palliation are the only current therapies. Objective: To evaluate the safety and wound outcomes following genetically corrected autologous epidermal grafts in patients with RDEB. Design, Setting, and Participants: Single-center phase 1 clinical trial conducted in the United States of 4 patients with severe RDEB with a measured area of wounds suitable for grafting of at least 100 cm2. Patients with undetectable type VII collagen keratinocyte expression were excluded. Interventions: Autologous keratinocytes isolated from biopsy samples collected from 4 patients with RDEB were transduced with good manufacturing practice-grade retrovirus carrying full-length human COL7A1 and assembled into epidermal sheet grafts. Type VII collagen gene-corrected grafts (approximately 35 cm2) were transplanted onto 6 wounds in each of the patients (n = 24 grafts). Main Outcomes and Measures: The primary safety outcomes were recombination competent retrovirus, cancer, and autoimmune reaction. Molecular correction was assessed as type VII collagen expression measured by immunofluorescence and immunoelectron microscopy. Wound healing was assessed using serial photographs taken at 3, 6, and 12 months after grafting. Results: The 4 patients (mean age, 23 years [range, 18-32 years]) were all male with an estimated body surface area affected with RDEB of 4% to 30%. All 24 grafts were well tolerated without serious adverse events. Type VII collagen expression at the dermal-epidermal junction was demonstrated on the graft sites by immunofluorescence microscopy in 9 of 10 biopsy samples (90%) at 3 months, in 8 of 12 samples (66%) at 6 months, and in 5 of 12 samples (42%) at 12 months, including correct type VII collagen localization to anchoring fibrils. Wounds with recombinant type VII collagen graft sites displayed 75% or greater healing at 3 months (21 intact graft sites of 24 wound sites; 87%), 6 months (16/24; 67%), and 12 months (12/24; 50%) compared with baseline wound sites. Conclusions and Relevance: In this preliminary study of 4 patients with RDEB, there was wound healing in some type VII collagen gene-corrected grafts, but the response was variable among patients and among grafted sites and generally declined over 1 year. Long-term follow-up is necessary for these patients, and controlled trials are needed with a broader range of patients to better understand the potential long-term efficacy of genetically corrected autologous epidermal grafts. Trial Registration: clinicaltrials.gov Identifier: NCT01263379.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/therapy , Gene Transfer Techniques , Keratinocytes/transplantation , Wound Healing , Adolescent , Adult , Collagen Type VII/metabolism , Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Humans , Male , Moloney murine leukemia virus/genetics , Pyrimidines , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Surgical Flaps , Time Factors , Wounds and Injuries/metabolism , Wounds and Injuries/therapy , Young Adult
5.
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
6.
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
7.
Actas Dermosifiliogr ; 106(5): 376-82, 2015 Jun.
Article in English, Spanish | MEDLINE | ID: mdl-25796272

ABSTRACT

Recessive dystrophic epidermolysis bullosa (RDEB) is among the most serious rare skin diseases. It is also the rare skin disease for which most effort has been expended in developing advanced therapeutic interventions. RDEB is caused by collagen VII deficiency resulting from COL7A1 mutations. Therapeutic approaches seek to replenish collagen VII and thus restore dermal-epidermal adhesion. Therapeutic options under development include protein therapy and different cell-based and gene-based therapies. In addition to treating skin defects, some of these therapies may also target internal mucosa. In the coming years, these novel therapeutic approaches should substantially improve the quality of life of patients with RDEB.


Subject(s)
Epidermolysis Bullosa Dystrophica/therapy , Therapies, Investigational , Animals , Cell- and Tissue-Based Therapy , Collagen Type VII/administration & dosage , Collagen Type VII/deficiency , Collagen Type VII/genetics , Collagen Type VII/therapeutic use , Disease Models, Animal , Forecasting , Genes, Recessive , Genetic Therapy , Humans , Mice , Mosaicism , Mutation , Tissue Engineering
9.
Sci Transl Med ; 6(264): 264ra163, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429056

ABSTRACT

Patients with recessive dystrophic epidermolysis bullosa (RDEB) lack functional type VII collagen owing to mutations in the gene COL7A1 and suffer severe blistering and chronic wounds that ultimately lead to infection and development of lethal squamous cell carcinoma. The discovery of induced pluripotent stem cells (iPSCs) and the ability to edit the genome bring the possibility to provide definitive genetic therapy through corrected autologous tissues. We generated patient-derived COL7A1-corrected epithelial keratinocyte sheets for autologous grafting. We demonstrate the utility of sequential reprogramming and adenovirus-associated viral genome editing to generate corrected iPSC banks. iPSC-derived keratinocytes were produced with minimal heterogeneity, and these cells secreted wild-type type VII collagen, resulting in stratified epidermis in vitro in organotypic cultures and in vivo in mice. Sequencing of corrected cell lines before tissue formation revealed heterogeneity of cancer-predisposing mutations, allowing us to select COL7A1-corrected banks with minimal mutational burden for downstream epidermis production. Our results provide a clinical platform to use iPSCs in the treatment of debilitating genodermatoses, such as RDEB.


Subject(s)
Collagen Type VII/genetics , Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/therapy , Genes, Recessive , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Animals , Base Sequence , Epidermolysis Bullosa Dystrophica/genetics , Genetic Predisposition to Disease , Genetic Therapy , Genome, Human , Homologous Recombination/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Keratinocytes/pathology , Mice , Molecular Sequence Data , Mutation/genetics , Sequence Analysis, DNA
10.
J Invest Dermatol ; 133(7): 1719-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23760051

ABSTRACT

In this issue, Woodley et al. report restoration of anchoring fibril formation and dermal-epidermal adherence in a murine model of recessive dystrophic epidermolysis bullosa (RDEB) by intravenous injection of recombinant human type VII collagen. This work follows a previous report by the same group of the surprising capability of intradermally injected type VII collagen protein to reverse RDEB, and it opens new therapeutic avenues.


Subject(s)
Collagen Type VII/pharmacology , Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/drug therapy , Epidermolysis Bullosa Dystrophica/pathology , Skin/drug effects , Wounds and Injuries/drug therapy , Animals , Humans
11.
Mol Ther ; 21(7): 1335-44, 2013 07.
Article in English | MEDLINE | ID: mdl-23670575

ABSTRACT

Patients with recessive dystrophic epidermolysis bullosa (RDEB) have incurable skin fragility, blistering, and skin wounds due to mutations in the gene that codes for type VII collagen (C7) that mediates dermal-epidermal adherence in human skin. In this study, we evaluated if topically applied human recombinant C7 (rC7) could restore C7 at the dermal-epidermal junction (DEJ) and enhance wound healing. We found that rC7 applied topically onto murine skin wounds stably incorporated into the newly formed DEJ of healed wounds and accelerated wound closure by increasing re-epithelialization. Topical rC7 decreased the expression of fibrogenic transforming growth factor-ß2 (TGF-ß2) and increased the expression of anti-fibrogenic TGF-ß3. These were accompanied by the reduced expression of connective tissue growth factor, fewer α smooth muscle actin (α-SMA)-positive myofibroblasts, and less deposition of collagen in the healed neodermis, consistent with less scar formation. In addition, using a mouse model in which skin from C7 knock out mice was grafted onto immunodeficient mice, we showed that applying rC7 onto RDEB grafts with wounds restored C7 and anchoring fibrils (AFs) at the DEJ of the grafts and corrected the dermal-epidermal separation. The topical application of rC7 may be useful for treating patients with RDEB and patients who have chronic skin wounds.


Subject(s)
Collagen Type VII/therapeutic use , Dermis/metabolism , Epidermis/metabolism , Recombinant Proteins/therapeutic use , Administration, Topical , Animals , Collagen Type VII/administration & dosage , Epidermolysis Bullosa Dystrophica/drug therapy , Fluorescent Antibody Technique , Humans , Mice , Mice, Nude , Recombinant Proteins/administration & dosage , Wound Healing/drug effects
13.
Methods Mol Biol ; 961: 425-40, 2013.
Article in English | MEDLINE | ID: mdl-23325662

ABSTRACT

Dystrophic epidermolysis bullosa (DEB) is a clinically heterogeneous heritable skin fragility disorder characterized by mechanically induced mucocutaneous blistering. On the molecular level DEB is caused by mutations leading to deficiency in collagen VII (CVII), a large extracellular protein building anchoring fibrils that attach the epidermis to the dermis. Severely affected patients suffer from wounds, which heal with excessive scarring causing mutilating deformities of hands and feet. The patients are also predisposed to development of aggressive squamous cell carcinomas at sites of chronic wounds. Currently no available therapies exist for this extremely disabling and stigmatizing disorder. We are developing and evaluating cell- and protein-based therapies for the management of DEB. Dermal fibroblasts are easy to propagate in vitro, they produce CVII, and they have immunomodulating capacities, which makes it possible to use allogeneic fibroblasts for therapy without risking major adverse effects from the host's immune system. Hence, fibroblasts, and fibroblast-like cells such as mesenchymal stromal cells, are prime candidates for cell-based DEB therapies. An alternative for management of disorders caused by defects in proteins with relatively low turnover rate is to introduce the protein de novo to the tissue by direct application of the protein. CVII is long-lived and expressed in moderate amounts in the skin; this makes injection of collagen VII protein a realistic approach for the treatment of DEB. Here we present methods and protocols that we are using for fibroblast- and recombinant CVII-based therapies of DEB in our model of this disease, the CVII hypomorphic mouse. These protocols are directed towards management of DEB but they can be easily adapted for the treatment of other skin fragility disorders.


Subject(s)
Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts/transplantation , Skin/pathology , Animals , Cell Culture Techniques/methods , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Collagen Type VII/isolation & purification , Disease Models, Animal , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Fibroblasts/metabolism , Fluorescent Antibody Technique/methods , Humans , Injections, Intradermal , Mice , Recombinant Proteins/isolation & purification , Recombinant Proteins/therapeutic use , Skin/metabolism
15.
Mol Ther ; 17(1): 26-33, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19018253

ABSTRACT

Patients with recessive dystrophic epidermolysis bullosa (RDEB) have incurable skin fragility, blistering, and scarring due to mutations in the gene that encodes for type VII collagen (C7) that mediates dermal-epidermal adherence in human skin. We showed previously that intradermal injection of recombinant C7 into transplanted human DEB skin equivalents stably restored C7 expression at the basement membrane zone (BMZ) and reversed the RDEB disease features. In this study, we evaluated the feasibility of protein therapy in a C7 null mouse (Col7a1(-/-)) which recapitulates the features of human RDEB. We intradermally injected purified human C7 into DEB mice and found that the injected human C7 stably incorporated into the mouse BMZ, formed anchoring fibrils, and corrected the DEB murine phenotype, as demonstrated by decreased skin fragility, reduced new blister formation, and markedly prolonged survival. After 4 weeks, treated DEB mice developed circulating anti-human C7 antibodies. Most surprisingly, these anti-C7 antibodies neither bound directly to the mouse's BMZ nor prevented the incorporation of newly injected human C7 into the BMZ. Anti-C7 antibody production was prevented by treating the mice with an anti-CD40L monoclonal antibody, MR1. We conclude that protein therapy may be feasible for the treatment of human patients with RDEB.


Subject(s)
Collagen Type VII/administration & dosage , Collagen Type VII/therapeutic use , Epidermolysis Bullosa Dystrophica/therapy , Animals , Antibodies/immunology , Basement Membrane/metabolism , Collagen Type VII/immunology , Collagen Type VII/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epidermolysis Bullosa Dystrophica/pathology , Humans , Injections, Intradermal , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Skin/metabolism , Skin/pathology , Skin/ultrastructure
16.
Anesth Analg ; 99(2): 595-7, table of contents, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15271747

ABSTRACT

We report a case of a chronically anticoagulated 59-yr-old woman who underwent an L4 to L5 epidural block to relieve her low back pain and subsequently developed a T7 to L5 epidural hematoma with cauda equina and conus compression. Fresh frozen plasma and vitamin K were given before surgery, whereas recombinant activated factor VII was administered during surgery to reverse the coagulopathy and to enable the emergent laminectomy and hematoma evacuation. Recombinant activated factor VII administration proved to be a useful adjunct in the emergent surgical management of a thoracolumbar epidural hematoma.


Subject(s)
Analgesia, Epidural/adverse effects , Collagen Type VII/therapeutic use , Hematoma, Epidural, Cranial/drug therapy , Anticoagulants/adverse effects , Female , Hematoma, Epidural, Cranial/etiology , Hemostatics/therapeutic use , Humans , Intraoperative Period , Low Back Pain/therapy , Magnetic Resonance Imaging , Middle Aged , Plasma , Recombinant Proteins/therapeutic use , Vitamin K/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...