Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Br J Dermatol ; 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38366625

ABSTRACT

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable widespread blistering skin disorder caused by mutations in the gene encoding for type VII collagen (C7), the major component of anchoring fibrils. OBJECTIVE: To evaluate the efficacy and safety of intravenous (IV) gentamicin-readthrough therapy in patients with RDEB harboring nonsense mutations. Primary outcomes were increased expression of C7 in patients' skin and assessments for safety (ototoxicity, nephrotoxicity, autoimmune response). Secondary outcomes included measuring wound healing in target wounds and assessment by a validated Epidermolysis Bullosa Disease and Activity Scarring Index (EBDASI) scoring system. METHODS: An open-label pilot trial assessing two different regimens of IV gentamicin between August 2018 and March 2020 with follow-up through 180 days post-treatment. Three RDEB patients with confirmed nonsense mutations in COL7A1 in either one or two alleles and decreased baseline expression of C7 at the dermal-epidermal junction (DEJ) of their skin participated in the study. Three patients received gentamicin at 7.5 mg/kg daily for 14 days and two of three patients further received 7.5 mg/kg IV gentamicin twice weekly for 12 weeks.Patients who had pre-existing auditory or renal impairment, were currently using ototoxic or nephrotoxic medications, or had allergies to aminoglycosides or sulfate compounds were excluded. RESULTS: After gentamicin treatment, skin biopsies from all three patients (ages ranging 18-28 years) exhibited increased C7 in their DEJ. With both regimens, the new C7 persisted at least six months post-treatment. At one and three-months post-treatment, 100% of the monitored wounds exhibited greater than 85% closure. Both IV gentamicin infusion regimens decreased EBDASI total activity scores. Of all patients assessed with the EBDASI, all patients exhibited decreased total activity scores three-month post-treatment. All three patients completed the study, and no adverse effects or anti-C7 antibodies were detected. CONCLUSIONS: IV gentamicin induced readthrough of nonsense mutations in RDEB patients and restored functional C7 in their skin, enhanced wound healing, and improved clinical parameters. IV gentamicin may be a safe, efficacious, low cost, and readily available therapy in this population of RDEB patients. TRIAL REGISTRATION: Clinicaltrials.gov Identifiers: NCT03392909.

2.
JAMA Dermatol ; 158(4): 366-374, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35234826

ABSTRACT

IMPORTANCE: Junctional epidermolysis bullosa (JEB) is an incurable blistering skin disorder with high infant mortality often caused by nonsense variants in the genes that encode laminin 332. OBJECTIVE: To evaluate the safety and outcomes following intravenous gentamicin readthrough therapy and subsequent laminin 332 expression in patients with JEB. DESIGN, SETTING, AND PARTICIPANTS: This open-label, pilot nonrandomized clinical trial assessed 1 course of low- or high-dose intravenous gentamicin, including follow-up at 30 and 90 days after treatment. Five pediatric patients with JEB (2 with intermediate JEB and 3 with severe JEB) and confirmed nonsense variants in LAMA3 or LAMB3 in 1 or 2 alleles and decreased expression of laminin 332 at the dermal-epidermal junction of their skin participated in the study, which was performed at a single institution in collaboration with physicians and home infusion services near the patients from April 1, 2019, to February 28, 2021, with follow-up until May 31, 2021. INTERVENTIONS: Three patients received gentamicin at 7.5 mg/kg daily for 14 days, and 2 patients received gentamicin at 10 mg/kg daily for 24 days. MAIN OUTCOMES AND MEASURES: Primary outcomes were change in expression of laminin 332 in patients' skin and assessments for safety (ototoxic effects, nephrotoxic effects, and autoimmune response). Secondary outcomes included wound healing in monitored wounds and Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI) score. RESULTS: After gentamicin treatment, all 5 patients (age range, 3 months to 10 years, 4 [80%] female) exhibited increased laminin 332 in the dermal-epidermal junction. By 1 month, 7 of 9 wounds in patients receiving low-dose intravenous gentamicin and all wounds in patients receiving high-dose intravenous gentamicin exhibited at least 50% wound closure. By 3 months, 8 of 9 wounds in patients receiving low-dose gentamicin and all wounds in patients receiving high-dose intravenous gentamicin exhibited greater than 85% closure. All 3 patients who were evaluated with EBDASI showed a decrease in total activity scores that met minimal clinically important differences 1 month after treatment. All 5 patients completed the study, and no ototoxic effects, nephrotoxic effects, or anti-laminin 332 antibodies were detected. CONCLUSIONS AND RELEVANCE: In this nonrandomized clinical trial, intravenous gentamicin therapy was associated with induced readthrough of nonsense variants in patients with JEB, restored functional laminin 332 in their skin, and wound closure during the 3-month study period. Although long-term safety and efficacy requires further evaluation, a single cycle of intravenous gentamicin may be a safe and readily available therapy in the short term for this population of patients with JEB. TRIAL REGISTRATION: ClinicalTrials.gov Identifiers: NCT03526159 and NCT04140786.


Subject(s)
Epidermolysis Bullosa, Junctional , Alleles , Child , Epidermolysis Bullosa, Junctional/drug therapy , Epidermolysis Bullosa, Junctional/genetics , Female , Gentamicins/metabolism , Gentamicins/therapeutic use , Humans , Infant , Laminin , Male , Skin/metabolism , Wound Healing
5.
Cancer Gene Ther ; 28(9): 1058-1070, 2021 09.
Article in English | MEDLINE | ID: mdl-33664459

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1), a master transcriptional factor for protecting cells from hypoxia, plays a critical role in spermatogenesis and tumorigenesis. For the past two decades, numerous small molecule inhibitors that block mRNA synthesis, protein translation, or DNA binding of HIF-1α have entered clinical trials. To date, few have advanced to FDA approval for clinical applications due to limited efficacy at their toxicity-tolerable dosages. New windows for developing effective and safe therapeutics require better understanding of the specific mechanism of action. The finding that a chaperone-defective mutant heat shock protein-90-alpha (Hsp90α) blocks spermatogenesis, a known hypoxia-driven process in mouse testis prompted us to focus on the role of Hsp90α in HIF-1α. Here we demonstrate that Hsp90α gene knockout causes a dramatic reduction of the high steady-state level of HIF-1α in the testis, blocking sperm production and causing infertility of the mice. In HIF-1α-dependent tumor cells, we found that Hsp90α forms protein complexes with hypoxia-elevated HIF-1α and Hsp90α knockout prevents hypoxia-induced HIF-1α accumulation. In contrast, downregulation of Hsp90ß had little effect on hypoxia-induced accumulation of HIF-1α. Instead, Hsp90ß protects signaling molecules responsible for cellular homeostasis from assault by 17-AAG (17-N-allylamino-17-demethoxygeldanamycin), a general ATPase inhibitor of both Hsp90α and Hsp90ß. Since targeting Hsp90ß gene is lethal in both cultured cells and in mice, our new finding explains the toxicity of the previous inhibitor trials and identifies the specific binding of Hsp90α to HIF-1α as a new therapeutic window for developing safer and more effective treatment of male infertility and cancer.


Subject(s)
Carcinogenesis/genetics , HSP90 Heat-Shock Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Spermatogenesis/genetics , Animals , Cell Transformation, Neoplastic , Humans , Male , Mice , Mice, Knockout
6.
Adv Wound Care (New Rochelle) ; 10(8): 415-435, 2021 08.
Article in English | MEDLINE | ID: mdl-32966158

ABSTRACT

Significance: Since the last Food and Drug Administration (FDA) approval of a wound healing therapeutic in 1997, no new therapeutic candidate (excluding physical therapies, devices, dressings, and antimicrobial agents) has advanced to clinical applications. During this period, the FDA drug approvals for tumors, which have been referred to as "wounds that do not heal," have reached a total of 284 (by end of 2018). Both political and scientific factors may explain this large discrepancy in drug approvals for the two seemingly related and equally complex pathophysiological conditions. Recent Advances: Using the current research funding ratio of 1:150 for wound healing to cancer and the 5% FDA drug approval rate for oncology, we reach a crude estimate of a 0.03% success rate for wound healing therapeutics. Unless a drastic improvement of the current situation, we express a pessimistic outlook toward new and effective wound healing drugs. Critical Issues: We argue that successful development of wound healing therapeutics will rely on identification of wound healing driver genes (WDGs), and the focus should be on WDGs for the wound closure phase of wound healing. Therefore, WDGs must be both necessary and sufficient for wound closure; the absence of a WDG disrupts wound closure, while its supplementation alone is sufficient to restore full wound closure. Successful translation of a WDG into therapeutics requires availability of well-defined animal models with a high degree of relevance to humans. This review discusses the main hurdles faced by the wound healing research community behind the development of so-called "rescuing drugs" for wound healing. Future Directions: Given the lack of new wound healing drugs for the past 23 years, there is a need for a wide range of fresh, innovative, and thorough debates on wound healing drug development, including an organized movement to raise public support for wound healing research.


Subject(s)
HSP90 Heat-Shock Proteins/therapeutic use , Wound Closure Techniques , Wound Healing/drug effects , Wound Healing/genetics , Wounds and Injuries/drug therapy , HSP90 Heat-Shock Proteins/pharmacology , Humans , United States
7.
Int J Surg Case Rep ; 74: 218-221, 2020.
Article in English | MEDLINE | ID: mdl-32892123

ABSTRACT

INTRODUCTION: Our case report describes a patient where multiple laparoscopies five years from initial presentation of symptoms were performed prior to laparotomy for benign multi-cystic peritoneal mesothelioma (BMPM), which has not been documented. PRESENTATION: A 61-year-old woman presented with years of chronic abdominal pain. Computerized tomography (CT) demonstrated a multi-cystic mass near the porta hepatis, and ultrasound was concerning for contained gallbladder perforation. Fine needle aspiration (FNA) demonstrated benign ductal epithelial cells in a background of mucin and bile without the presence of malignant cells. During laparotomy, a cystic mass attached to the porta hepatis seen emanating from the small bowel mesentery, and additional small cystic lesions through the abdomen were removed. The specimen, measuring 26 × 18 × 8 cm, showed multi-loculated cysts filled with serous fluid. DISCUSSION: BMPM is a rare neoplasm of mesothelioma cells originating from serosa of viscous organs. BMPMs appear as cystic structures with thin walls containing mucinous/gelatinous fluid. Microscopic features include a lack of invasion and no increased cellularity in the stroma, with or without inflammation (Myers & Babiker, 2018). It is postulated to be either a reactive or neoplastic process. There is no gold-standard treatment for BMPM. Our case is unique in the sense that our patient required several surgical biopsies before final diagnosis could be made. CONCLUSION: This case highlights the difficulty of diagnosing BMPM and differentiating it from malignant diseases that can present similarly and can be associated with significantly worse prognosis. Defined management strategies have yet to be demonstrated.

8.
Curr Opin Pediatr ; 32(4): 539-546, 2020 08.
Article in English | MEDLINE | ID: mdl-32692051

ABSTRACT

PURPOSE OF REVIEW: Over the past decade many previously poorly understood vascular malformation disorders have been linked to somatic activating mutations in PIK3CA, which regulates cell survival and growth via activation of the mTOR1-AKT pathway. The goal of this article is to describe and provide an update on the clinical features, complications, and management strategies for the PIK3CA-related overgrowth spectrum (PROS). RECENT FINDINGS: PROS encompasses a heterogenous group of disorders with complications related to the tissues harboring the mutation. Vascular malformation syndromes, such as Klippel-Trenaunay syndrome and Congenital Lipomatous Overgrowth Vascular malformations Epidermal nevi and Skeletal abnormalities, have an increased risk of thromboembolic complications, which is accentuated postprocedurally. Asymmetric overgrowth, particularly of limbs, results in a high rate of orthopedic complications. Hypoglycemia screening in the neonatal period and ongoing monitoring for growth failure is recommended in megalencephaly capillary malformation due to its association with multiple endocrinopathies. Recently, sirolimus, an mTOR1 inhibitor, has shown promise in vascular anomalies and now PROS. PIK3CA direct inhibitor, Alpelisib (BYL719), was recently trialed with significant clinical benefit. SUMMARY: As the pathogenesis of these conditions is better elucidated and targeted treatments are developed, recognizing the clinical features, comorbidities, and evolving therapeutic landscape across the PROS spectrum becomes more crucial for optimization of care.


Subject(s)
Abnormalities, Multiple/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Growth Disorders/diagnosis , Growth Disorders/genetics , Musculoskeletal Abnormalities , Vascular Malformations/genetics , Abnormalities, Multiple/diagnosis , Biomarkers/blood , Class I Phosphatidylinositol 3-Kinases/metabolism , Genetic Testing , Humans , Mosaicism , Mutation , Syndrome
9.
Am J Emerg Med ; 38(12): 2761.e1-2761.e3, 2020 12.
Article in English | MEDLINE | ID: mdl-32518024

ABSTRACT

BACKGROUND: Erythema multiforme (EM), an acute dermatologic condition frequently encountered in the Emergency Department, classically presents with a targetoid rash. We reviewed all recent EM cases seen at the LAC-USC County Hospital in order to ascertain the proportion of Herpes associated EM (HAEM) cases and to inform the diagnostic workup of these patients. METHODS: ICD-9 and ICD-10 codes were used to extract a list of EM cases at our institution from 2013 to 2019. Two non-blinded abstractors screened records to confirm an EM diagnosis and entered patient data utilizing a standardized data abstraction form. Cohen's kappa statistic was used to measure inter-rater reliability on various variables. Kappa (κ) values ranged from 0.803 to 1.0. RESULTS: 70 pediatric and 56 adult EM patients were included in the study. A likely etiology was ascribed to 63% of pediatric and adult EM cases. Pediatric EM was most commonly attributed to upper respiratory infection (URI) (n = 23; 33%), Mycoplasma pneumoniae infection (n = 5; 7%), and medications (n = 4; 6%). Adult EM was most commonly attributed to HSV infection (n = 11; 20%), medications (n = 5; 9%), URIs (n = 4; 7%), and other infections (n = 4; 7%). CONCLUSION: HSV-1/2 serologic testing should be considered in most EM patients to potentially prevent repeated ED visits. In EM cases not clearly attributable to herpes or drug exposure, physicians can consider further workup: Mycoplasma serology, nasal PCR, and a respiratory viral panel in pediatric patients. Identification of an etiologic cause may suggest a different treatment approach and prevent mislabeling of medication allergies in patient charts.


Subject(s)
Erythema Multiforme/etiology , Herpes Simplex/complications , Adult , Antibodies, Viral , Child , Child, Preschool , Drug Eruptions/complications , Erythema Multiforme/physiopathology , Female , Herpes Simplex/diagnosis , Humans , Infant , Male , Middle Aged , Pneumonia, Mycoplasma/complications , Respiratory Tract Infections/complications , Retrospective Studies , Serologic Tests , Young Adult
10.
Mol Ther ; 28(5): 1327-1338, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32222156

ABSTRACT

Generalized severe junctional epidermolysis bullosa (GS-JEB) is an incurable and fatal autosomal recessively inherited blistering skin disease caused by mutations in the LAMA3, LAMB3, or LAMC2 genes. Most of these mutations are nonsense mutations that create premature termination codons that lead to impaired production of functional laminin 332, a protein needed for epidermal-dermal adherence. Gentamicin induces readthrough of nonsense mutations and restores the full-length protein in various genetic diseases. Using primary keratinocytes from three GS-JEB patients, we showed that gentamicin induced functional laminin 332 that reversed a JEB-associated, abnormal cell phenotype. In a subsequent open-label trial involving the same patients, we examined whether 0.5% gentamicin ointment applied topically to open skin wounds could promote nonsense mutation readthrough and create new laminin 332 in the patients' skin. Gentamicin-treated wounds exhibited increased expression of laminin 332 at the dermal-epidermal junction for at least 3 months and were associated with improved wound closure. There were no untoward side effects from topical gentamicin. The newly induced laminin 332 did not generate anti-laminin 332 autoantibodies in either the patients' blood or skin. Gentamicin readthrough therapy may be a treatment for GS-JEB patients with nonsense mutations.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Cell Adhesion Molecules/metabolism , Codon, Nonsense/genetics , Epidermolysis Bullosa, Junctional/drug therapy , Epidermolysis Bullosa, Junctional/genetics , Gentamicins/administration & dosage , Signal Transduction/drug effects , Wound Healing/drug effects , Administration, Cutaneous , Anti-Bacterial Agents/adverse effects , Cell Survival/drug effects , Cells, Cultured , Child , Child, Preschool , Epidermolysis Bullosa, Junctional/pathology , Female , Follow-Up Studies , Gentamicins/adverse effects , Humans , Infant , Keratinocytes/metabolism , Male , Skin/metabolism , Treatment Outcome , Kalinin
11.
Proc Natl Acad Sci U S A ; 115(28): E6536-E6545, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29946029

ABSTRACT

Herlitz junctional epidermolysis bullosa (H-JEB) is an incurable, devastating, and mostly fatal inherited skin disease for which there is only supportive care. H-JEB is caused by loss-of-function mutations in LAMA3, LAMB3, or LAMC2, leading to complete loss of laminin 332, the major component of anchoring filaments, which mediate epidermal-dermal adherence. LAMB3 (laminin ß3) mutations account for 80% of patients with H-JEB, and ∼95% of H-JEB-associated LAMB3 mutations are nonsense mutations leading to premature termination codons (PTCs). In this study, we evaluated the ability of gentamicin to induce PTC readthrough in H-JEB laminin ß3-null keratinocytes transfected with expression vectors encoding eight different LAMB3 nonsense mutations. We found that gentamicin induced PTC readthrough in all eight nonsense mutations tested. We next used lentiviral vectors to generate stably transduced H-JEB cells with the R635X and C290X nonsense mutations. Incubation of these cell lines with various concentrations of gentamicin resulted in the synthesis and secretion of full-length laminin ß3 in a dose-dependent and sustained manner. Importantly, the gentamicin-induced laminin ß3 led to the restoration of laminin 332 assembly, secretion, and deposition within the dermal/epidermal junction, as well as proper polarization of α6ß4 integrin in basal keratinocytes, as assessed by immunoblot analysis, immunofluorescent microscopy, and an in vitro 3D skin equivalent model. Finally, newly restored laminin 332 corrected the abnormal cellular phenotype of H-JEB cells by reversing abnormal cell morphology, poor growth potential, poor cell-substratum adhesion, and hypermotility. Therefore, gentamicin may offer a therapy for H-JEB and other inherited skin diseases caused by PTC mutations.


Subject(s)
Cell Adhesion Molecules , Codon, Nonsense , Epidermolysis Bullosa, Junctional , Gentamicins/pharmacology , Keratinocytes/metabolism , Mutagenesis/drug effects , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/metabolism , Epidermolysis Bullosa, Junctional/pathology , HEK293 Cells , Humans , Integrin alpha6beta4/genetics , Integrin alpha6beta4/metabolism , Keratinocytes/pathology , Kalinin
SELECTION OF CITATIONS
SEARCH DETAIL
...