Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Drugs ; 84(2): 219-226, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38252335

RESUMEN

Lumasiran (Oxlumo®), a first-in-class synthetic, double-stranded, ribonucleic acid (RNA) interference molecule targeting glycolate oxidase through silencing HAO1 mRNA, is approved in several countries for patients of any age and stage of kidney function with primary hyperoxaluria type 1 (PH1). Approval was based on results from the phase III ILLUMINATE trials. In the double-blind, placebo-controlled, ILLUMINATE-A trial, subcutaneous lumasiran was significantly more effective than placebo in reducing 24-h urinary oxalate excretion in patients aged ≥ 6 years with PH1; this effect was sustained for ≥ 36 months in ongoing longer-term analyses. In the single-arm ILLUMINATE-B trial, lumasiran reduced urinary oxalate:creatinine ratios and plasma oxalate levels in patients aged < 6 years with PH1. In the single-arm ILLUMINATE-C trial, lumasiran reduced plasma oxalate levels in patients with PH1 receiving dialysis as well as those not receiving dialysis. In secondary and exploratory analyses of these trials, nephrocalcinosis grade, kidney stone event rates and estimated glomerular filtration rates were either stable or improved with lumasiran. Lumasiran had an acceptable tolerability profile that remained consistent in longer-term analyses; the most common adverse events were mild and transient injection-site reactions. Thus, lumasiran is an effective treatment option, with an acceptable tolerability profile, in patients with PH1.


Primary hyperoxaluria type 1 (PH1) is a rare genetic disorder that leads to excess oxalate in urine or plasma requiring removal by the kidneys. This overproduction is damaging and can lead to kidney failure. Management of PH1 is typically not curative, eventually ending in kidney and/or liver transplantation. Lumasiran (Oxlumo®) is the first medicine to be approved in several countries for use in patients with PH1, regardless of their age or level of kidney function. It reduces liver oxalate production, lessening damage to the kidneys and potentially reducing the need for organ transplantation. In clinical trials, lumasiran was effective in reducing oxalate levels (in urine and/or plasma) in patients of all ages with PH1, and irrespective of whether they were receiving dialysis or not. Lumasiran either improved or stabilized the severity of calcium deposition in the kidneys, number of kidney stone events and kidney function. Lumasiran had an acceptable tolerability profile; the most common side effects were mild injection-site reactions that resolved quickly. Thus, lumasiran is an effective treatment option, with an acceptable tolerability profile, in patients with PH1.


Asunto(s)
Hiperoxaluria Primaria , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Oxalatos , ARN Interferente Pequeño , Ensayos Clínicos Controlados Aleatorios como Asunto
2.
Urolithiasis ; 51(1): 80, 2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37118061

RESUMEN

Nedosiran is an N-acetyl-D-galactosamine (GalNAc)-conjugated RNA interference agent targeting hepatic lactate dehydrogenase (encoded by the LDHA gene), the putative enzyme mediating the final step of oxalate production in all three genetic subtypes of primary hyperoxaluria (PH). This phase I study assessed the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of subcutaneous nedosiran in patients with PH subtype 3 (PH3) and an estimated glomerular filtration rate  ≥ 30 mL/min/1.73 m2. Single-dose nedosiran 3 mg/kg or placebo was administered in a randomized (2:1), double-blinded manner. Safety/tolerability, 24-h urinary oxalate (Uox) concentrations, and plasma nedosiran concentrations were assessed. The main PD endpoint was the proportion of participants achieving a > 30% decrease from baseline in 24-h Uox at two consecutive visits. Six participants enrolled in and completed the study (nedosiran, n = 4; placebo, n = 2). Nedosiran was well-tolerated and lacked safety concerns. Although the PD response was not met, 24-h Uox excretion declined 24.5% in the nedosiran group and increased 10.5% in the placebo group at Day 85. Three of four nedosiran recipients had a > 30% reduction in 24-h Uox excretion during at least one visit, and one attained near-normal (i.e., ≥ 0.46 to < 0.60 mmol/24 h; ≥ 1.0 to < 1.3 × upper limit of the normal reference range) 24-h Uox excretion from Day 29 to Day 85. Nedosiran displayed predictable plasma PK. The acceptable safety and trend toward Uox-lowering after single-dose nedosiran treatment enables further clinical development of nedosiran in patients with PH3 who currently have no viable therapeutic options. A plain language summary is available in the supplementary information.


Asunto(s)
Hiperoxaluria Primaria , Hiperoxaluria , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/genética , Hiperoxaluria/orina , Oxalatos/orina , Tasa de Filtración Glomerular
4.
Pediatr Nephrol ; 38(4): 1075-1086, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35913563

RESUMEN

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease that causes progressive kidney damage and systemic oxalosis due to hepatic overproduction of oxalate. Lumasiran demonstrated efficacy and safety in the 6-month primary analysis period of the phase 3, multinational, open-label, single-arm ILLUMINATE-B study of infants and children < 6 years old with PH1 (ClinicalTrials.gov: NCT03905694 (4/1/2019); EudraCT: 2018-004,014-17 (10/12/2018)). Outcomes in the ILLUMINATE-B extension period (EP) for patients who completed ≥ 12 months on study are reported here. METHODS: Of the 18 patients enrolled in the 6-month primary analysis period, all entered the EP and completed ≥ 6 additional months of lumasiran treatment (median (range) duration of total exposure, 17.8 (12.7-20.5) months). RESULTS: Lumasiran treatment was previously reported to reduce spot urinary oxalate:creatinine ratio by 72% at month 6, which was maintained at 72% at month 12; mean month 12 reductions in prespecified weight subgroups were 89%, 68%, and 71% for patients weighing < 10 kg, 10 to < 20 kg, and ≥ 20 kg, respectively. The mean reduction from baseline in plasma oxalate level was reported to be 32% at month 6, and this improved to 47% at month 12. Additional improvements were also seen in nephrocalcinosis grade, and kidney stone event rates remained low. The most common lumasiran-related adverse events were mild, transient injection-site reactions (3 patients (17%)). CONCLUSIONS: Lumasiran treatment provided sustained reductions in urinary and plasma oxalate through month 12 across all weight subgroups, with an acceptable safety profile, in infants and young children with PH1. A higher resolution version of the Graphical abstract is available as Supplementary information.


Asunto(s)
Hiperoxaluria Primaria , Cálculos Renales , Niño , Preescolar , Humanos , Lactante , Hiperoxaluria Primaria/complicaciones , Hiperoxaluria Primaria/tratamiento farmacológico , Cálculos Renales/etiología , Oxalatos/efectos adversos
5.
Kidney Int ; 103(1): 207-217, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36007597

RESUMEN

Nedosiran is an investigational RNA interference agent designed to inhibit expression of hepatic lactate dehydrogenase, the enzyme thought responsible for the terminal step of oxalate synthesis. Oxalate overproduction is the hallmark of all genetic subtypes of primary hyperoxaluria (PH). In this double-blind, placebo-controlled study, we randomly assigned (2:1) 35 participants with PH1 (n = 29) or PH2 (n = 6) with eGFR ≥30 mL/min/1.73 m2 to subcutaneous nedosiran or placebo once monthly for 6 months. The area under the curve (AUC) of percent reduction from baseline in 24-hour urinary oxalate (Uox) excretion (primary endpoint), between day 90-180, was significantly greater with nedosiran vs placebo (least squares mean [SE], +3507 [788] vs -1664 [1190], respectively; difference, 5172; 95% CI 2929-7414; P < 0.001). A greater proportion of participants receiving nedosiran vs placebo achieved normal or near-normal (<0.60 mmol/24 hours; <1.3 × ULN) Uox excretion on ≥2 consecutive visits starting at day 90 (50% vs 0; P = 0.002); this effect was mirrored in the nedosiran-treated PH1 subgroup (64.7% vs 0; P < 0.001). The PH1 subgroup maintained a sustained Uox reduction while on nedosiran, whereas no consistent effect was seen in the PH2 subgroup. Nedosiran-treated participants with PH1 also showed a significant reduction in plasma oxalate versus placebo (P = 0.017). Nedosiran was generally safe and well tolerated. In the nedosiran arm, the incidence of injection-site reactions was 9% (all mild and self-limiting). In conclusion, participants with PH1 receiving nedosiran had clinically meaningful reductions in Uox, the mediator of kidney damage in PH.


Asunto(s)
Hiperoxaluria Primaria , Hiperoxaluria , Humanos , Hiperoxaluria/orina , Hiperoxaluria Primaria/diagnóstico , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/genética , Oxalatos/metabolismo , Interferencia de ARN , Método Doble Ciego
6.
J Med Chem ; 65(14): 9718-9734, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35830169

RESUMEN

Primary hyperoxaluria type I (PH1) is a rare kidney disease due to the deficit of alanine:glyoxylate aminotransferase (AGT), a pyridoxal-5'-phosphate-dependent enzyme responsible for liver glyoxylate detoxification, which in turn prevents oxalate formation and precipitation as kidney stones. Many PH1-associated missense mutations cause AGT misfolding. Therefore, the use of pharmacological chaperones (PCs), small molecules that promote correct folding, represents a useful therapeutic option. To identify ligands acting as PCs for AGT, we first performed a small screening of commercially available compounds. We tested each molecule by a dual approach aimed at defining the inhibition potency on purified proteins and the chaperone activity in cells expressing a misfolded variant associated with PH1. We then performed a chemical optimization campaign and tested the resulting synthetic molecules using the same approach. Overall, the results allowed us to identify a promising hit compound for AGT and draw conclusions about the requirements for optimal PC activity.


Asunto(s)
Hiperoxaluria Primaria , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Ligandos , Mutación , Pliegue de Proteína , Transaminasas/metabolismo
7.
Drugs ; 82(10): 1077-1094, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35779234

RESUMEN

The primary hyperoxalurias are three rare inborn errors of the glyoxylate metabolism in the liver, which lead to massively increased endogenous oxalate production, thus elevating urinary oxalate excretion and, based on that, recurrent urolithiasis and/or progressive nephrocalcinosis. Frequently, especially in type 1 primary hyperoxaluria, early end-stage renal failure occurs. Treatment possibilities are scare, namely, hyperhydration and alkaline citrate medication. In type 1 primary hyperoxaluria, vitamin B6, though, is helpful in patients with specific missense or mistargeting mutations. In those vitamin B6 responsive, urinary oxalate excretion and concomitantly urinary glycolate is significantly decreased, or even normalized. In patients non-responsive to vitamin B6, RNA interference medication is now available. Lumasiran® is already available on prescription and targets the messenger RNA of glycolate oxidase, thus blocking the conversion of glycolate into glyoxylate, hence decreasing oxalate, but increasing glycolate production. Nedosiran blocks liver-specific lactate dehydrogenase A and thus the final step of oxalate production. Similar to vitamin B6 treatment, where both RNA interference urinary oxalate excretion can be (near) normalized and plasma oxalate decreases, however, urinary and plasma glycolate increases with lumasiran treatment. Future treatment possibilities are on the horizon, for example, substrate reduction therapy with small molecules or gene editing, induced pluripotent stem cell-derived autologous hepatocyte-like cell transplantation, or gene therapy with newly developed vector technologies. This review provides an overview of current and especially new and future treatment options.


Asunto(s)
Hiperoxaluria Primaria , Glicolatos , Glioxilatos , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Oxalatos/metabolismo , ARN Interferente Pequeño , Vitaminas
8.
s.l; CONETEC; 28 jun. 2022.
No convencional en Español | BRISA/RedTESA | ID: biblio-1381036

RESUMEN

INTRODUCCIÓN: La hiperoxaluria primaria (HOP) es un desorden metabólico hereditario autosómico recesivo del metabolismo del glioxalato, que cursa con una producción excesiva de oxalato. El oxalato es un ácido dicarboxílico que proviene principalmente del metabolismo endógeno y solo una pequeña parte de la dieta. La detoxificación del glioxalato se realiza mayormente por la alaninglioxalato amino transferasa (AGT) en el peroxisoma del hepatocito humano, convirtiendo el glioxalato en glicina. En condiciones normales, sólo parte del glioxalato es transformado en oxalato por la lactato dehidrogenasa (LDH). El oxalato no se une a proteínas plasmáticas y no sufre metabolización, siendo excretado sin cambios por vía renal. Se han descrito tres tipos de trastornos moleculares: el HOP tipo 1 (HOP-1), tipo 2 y tipo 3, donde la HOP-1 es causada por el déficit de la AGT que produce un incremento de glicolato y oxalato. La incidencia de HOP es difícil de estimar dado que en muchos casos son de diagnóstico tardío, generalmente luego de la recidiva de depósitos de oxalatos en el trasplante renal, o no son identificados. Algunos países europeos han estimado una prevalencia de 1 a 3 casos por millón de personas y una tasa de incidencia de 1 cada 100.000 nacidos vivos para esta región. No se han hallado información epidemiológica para Argentina pero está reconocida en el listado de enfermedades poco frecuentes de nuestro país (ORPHACODE 416). TECNOLOGÍA: Lumasiran (Oxlumo™) es un ARN pequeño de interferencia que se dirige al ARNm del gen de la hidroxiácido oxidasa 1 (HAO1; codifica la glicolato oxidasa). Lumasiran reduce los niveles de la enzima glicolato oxidasa en los hepatocitos a través de la interferencia del ARNm. Los niveles reducidos de la enzima reducen la cantidad de glioxilato disponible, un sustrato para la producción de oxalato, que es el metabolito asociado con las manifestaciones clínicas de HOP1. Se administra por vía subcutánea a dosis de carga para adultos de 3 mg/kg una vez al mes durante tres meses, y una dosis de mantenimiento 3 mg/kg una vez cada tres meses comenzando un mes después de la última dosis de carga. Para pediátricos la dosis de ajusta según el peso corporal, según si poseen menos de 10kg, entre 10 y 20kg, o más de 20kg. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de lumasiran para el tratamiento de personas con hiperoxaluria primaria de tipo 1. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. RECOMENDACIONES: No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que mencionen la tecnología en la indicación evaluada. El Instituto Nacional para la Excelencia en Salud y Atención (del inglés, National Institute for Health and Care Excellence) de Reino Unido y la Agencia Canadiense de Medicamentos y Tecnologías en Salud (del inglés, Canadian Agency for Drugs & Technologies in Health) se encuentran evaluando el tratamiento para la indicación mencionada en estos momentos. CONCLUSIONES: La evidencia que sustenta la aprobación de comercialización de lumasiran (Oxlumo™) en pediátricos y adultos con hiperoxaluria primaria tipo 1 se basa en dos estudios de Fase III en centros pertenecientes a países del hemisferio norte. Estos estudios demostrarían que en personas con buen estado salud renal y sin oxalosis sistémica extrarrenal, lumasiran sumado al cuidado estándar reduciría el oxalato urinario y plasmático frente a placebo al corto plazo. Debido al escaso seguimiento, todavía no existen datos maduros sobre desenlaces importantes como la reducción de cálculos renales, de la falla renal y de la oxalosis. No se reportaron eventos adversos serios para lumasiran, y generalmente se informaron reacciones leves y transitorias en el lugar de la inyección. Las agencias regulatorias relevadas han autorizado recientemente la comercialización, junto con la designación de medicamento huérfano, de lumasiran para reducir los niveles de oxalato en orina en pediátricos y adultos con hiperoxaluria primaria tipo 1. No se hallaron guías de práctica clínica actualizadas en Argentina y en el Mundo que mencionen la tecnología en la indicación evaluada. No se hallaron evaluaciones económicas publicadas, aunque el costo del fármaco es muy elevado.


Asunto(s)
Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Argentina , ARN/uso terapéutico , Eficacia , Análisis Costo-Beneficio/economía
9.
Pediatr Nephrol ; 37(4): 907-911, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35015123

RESUMEN

BACKGROUND: Lumasiran, a sub-cutaneous RNA-interference therapy, has been recently approved for primary hyperoxaluria type 1 (PH1), with doses and intervals according to body weight. Little is known as to its use in infants; the aim of this study was to describe treatment outcome in 3 infants who received lumasiran therapy before 2 years of age. CASE-DIAGNOSIS/TREATMENT: Patient 1 was diagnosed antenatally and received lumasiran from day 9. According to the product information template (PIT), he received monthly lumasiran (3 times at 6 mg/kg, then 3 mg/kg), with hyperhydration and potassium citrate. Despite decreased plasma oxalate levels, persistent normal kidney function, and good tolerance, kidney ultrasound performed after 2 months found nephrocalcinosis, without normalization of urinary oxalate (UOx). The dose was increased back to 6 mg/kg, inducing a normalization in UOx. Nephrocalcinosis started to improve at month 10. Patient 2 was diagnosed at 2.5 months (acute kidney failure); nephrocalcinosis was present from diagnosis. She received monthly lumasiran (6 mg/kg), with progressive decrease in UOx and substantial improvement in kidney function but stable nephrocalcinosis after 9 injections. Patient 3 was diagnosed fortuitously (nephrocalcinosis) at 3.5 months and received lumasiran before genetic diagnosis, leading to decreased UOx and maintenance of normal kidney function. Nephrocalcinosis improved after 5 injections. CONCLUSIONS: This report presents the youngest children treated with lumasiran worldwide. Lumasiran seems effective without side effects in infants but does not completely prevent the onset of nephrocalcinosis in the most severe forms. Higher doses than those proposed in the PIT might be required because of hepatic immaturity.


Asunto(s)
Hiperoxaluria Primaria , Nefrocalcinosis , Femenino , Humanos , Hiperoxaluria Primaria/complicaciones , Hiperoxaluria Primaria/diagnóstico , Hiperoxaluria Primaria/tratamiento farmacológico , Lactante , Masculino , Nefrocalcinosis/tratamiento farmacológico , Nefrocalcinosis/etiología , Oxalatos , ARN Interferente Pequeño
10.
Int J Mol Sci ; 23(2)2022 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-35055190

RESUMEN

Hydroxyproline is one of the most prevalent amino acids in animal proteins. It is not a genetically encoded amino acid, but, rather, it is produced by the post-translational modification of proline in collagen, and a few other proteins, by prolyl hydroxylase enzymes. Although this post-translational modification occurs in a limited number of proteins, its biological significance cannot be overestimated. Considering that hydroxyproline cannot be re-incorporated into pro-collagen during translation, it should be catabolized following protein degradation. A cascade of reactions leads to production of two deleterious intermediates: glyoxylate and hydrogen peroxide, which need to be immediately converted. As a result, the enzymes involved in hydroxyproline catabolism are located in specific compartments: mitochondria and peroxisomes. The particular distribution of catabolic enzymes in these compartments, in different species, depends on their dietary habits. Disturbances in hydroxyproline catabolism, due to genetic aberrations, may lead to a severe disease (primary hyperoxaluria), which often impairs kidney function. The basis of this condition is accumulation of glyoxylate and its conversion to oxalate. Since calcium oxalate is insoluble, children with this rare inherited disorder suffer from progressive kidney damage. This condition has been nearly incurable until recently, as significant advances in substrate reduction therapy using small interference RNA led to a breakthrough in primary hyperoxaluria type 1 treatment.


Asunto(s)
Hidroxiprolina/metabolismo , Hiperoxaluria Primaria/genética , ARN Interferente Pequeño/farmacología , Animales , Evolución Molecular , Predisposición Genética a la Enfermedad , Glioxilatos/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/metabolismo , Mitocondrias/metabolismo , Peroxisomas/metabolismo , ARN Interferente Pequeño/uso terapéutico
12.
Kidney Int ; 101(3): 626-634, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34481803

RESUMEN

Primary hyperoxaluria (PH) is a family of ultra-rare autosomal recessive inherited disorders of hepatic glyoxylate metabolism characterized by oxalate overproduction. Nedosiran is an RNA interference agent that inhibits hepatic lactate dehydrogenase, the enzyme responsible for the common, final step of oxalate production in all three genetic subtypes of PH. Here, we assessed in a two-part, randomized, single-ascending-dose, phase 1 study (PHYOX1) the safety, pharmacokinetics, pharmacodynamics, and exposure-response of subcutaneous nedosiran in 25 healthy participants (Group A) and 18 patients with PH1 or PH2 (Group B). Group A received nedosiran (0.3, 1.5, 3.0, 6.0, then 12.0 mg/kg) or placebo, and Group B received open-label nedosiran (1.5, 3.0, or 6.0 mg/kg). No significant safety concerns were identified. Injection site reactions (four or more hours post dose) occurred in 13.3% of participants in Group A and 27.8% of participants in Group B. Mean maximum reduction in 24-hour urinary oxalate excretion from baseline to day 57 (end of study) across Group B dose cohorts was 55% (range: 22%-100%) after single-dose nedosiran, with 33% participants reaching normal 24-hour urinary oxalate excretion. Based on the available modeling and simulation data, a fixed monthly dose of nedosiran 160 mg (free acid; equivalent to 170 mg sodium salt) in adults was associated with the highest proportion of simulated individuals achieving normal or near-normal 24-hour urinary oxalate excretion and fewest fluctuations in urinary oxalate response. Thus, single-dose nedosiran demonstrated acceptable safety and evidence of a pharmacodynamic effect in both PH1 and PH2 subpopulations consistent with its mechanism of action.


Asunto(s)
Hiperoxaluria Primaria , Adulto , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/genética , Oxalatos/orina , Interferencia de ARN
14.
Clin J Am Soc Nephrol ; 16(7): 1025-1036, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33985991

RESUMEN

BACKGROUND AND OBJECTIVES: In the rare disease primary hyperoxaluria type 1, overproduction of oxalate by the liver causes kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an RNA interference therapeutic, suppresses glycolate oxidase, reducing hepatic oxalate production. The objective of this first-in-human, randomized, placebo-controlled trial was to evaluate the safety, pharmacokinetic, and pharmacodynamic profiles of lumasiran in healthy participants and patients with primary hyperoxaluria type 1. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This phase 1/2 study was conducted in two parts. In part A, healthy adults randomized 3:1 received a single subcutaneous dose of lumasiran or placebo in ascending dose groups (0.3-6 mg/kg). In part B, patients with primary hyperoxaluria type 1 randomized 3:1 received up to three doses of lumasiran or placebo in cohorts of 1 or 3 mg/kg monthly or 3 mg/kg quarterly. Patients initially assigned to placebo crossed over to lumasiran on day 85. The primary outcome was incidence of adverse events. Secondary outcomes included pharmacokinetic and pharmacodynamic parameters, including measures of oxalate in patients with primary hyperoxaluria type 1. Data were analyzed using descriptive statistics. RESULTS: Thirty-two healthy participants and 20 adult and pediatric patients with primary hyperoxaluria type 1 were enrolled. Lumasiran had an acceptable safety profile, with no serious adverse events or study discontinuations attributed to treatment. In part A, increases in mean plasma glycolate concentration, a measure of target engagement, were observed in healthy participants. In part B, patients with primary hyperoxaluria type 1 had a mean maximal reduction from baseline of 75% across dosing cohorts in 24-hour urinary oxalate excretion. All patients achieved urinary oxalate levels ≤1.5 times the upper limit of normal. CONCLUSIONS: Lumasiran had an acceptable safety profile and reduced urinary oxalate excretion in all patients with primary hyperoxaluria type 1 to near-normal levels. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER: Study of Lumasiran in Healthy Adults and Patients with Primary Hyperoxaluria Type 1, NCT02706886.


Asunto(s)
Hiperoxaluria Primaria/tratamiento farmacológico , Oxalatos/orina , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/farmacocinética , Fármacos Renales/farmacología , Fármacos Renales/farmacocinética , Adolescente , Adulto , Niño , Femenino , Glicolatos/sangre , Humanos , Hiperoxaluria Primaria/sangre , Hiperoxaluria Primaria/orina , Masculino , ARN Interferente Pequeño/efectos adversos , Fármacos Renales/efectos adversos , Método Simple Ciego , Adulto Joven
15.
J AAPOS ; 25(2): 128-131, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33737053

RESUMEN

Fundus manifestations of primary hyperoxaluria include crystalline deposits, focal or diffuse macular hyperpigmentation, and subretinal fibrosis. Choroidal neovascularization has been hypothesized to underlie the pathogenesis of subretinal fibrosis, yet its manifestations are rarely observed. We report a case of infantile primary hyperoxaluria type 1 in a 17-month-old infant with macular subretinal fluid and subretinal hemorrhage that was associated with leakage on fluorescein angiography and responded to bevacizumab treatment, consistent with choroidal neovascularization. This case suggests that choroidal neovascularization may contribute to subretinal fibrosis and subsequent vision loss in infantile primary hyperoxaluria and may benefit from anti-vascular endothelial growth factor therapy.


Asunto(s)
Neovascularización Coroidal , Hiperoxaluria Primaria , Inhibidores de la Angiogénesis/uso terapéutico , Bevacizumab/uso terapéutico , Neovascularización Coroidal/diagnóstico , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/etiología , Angiografía con Fluoresceína , Humanos , Hiperoxaluria Primaria/tratamiento farmacológico , Lactante , Inyecciones Intravítreas , Tomografía de Coherencia Óptica , Agudeza Visual
16.
N Engl J Med ; 384(13): 1216-1226, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33789010

RESUMEN

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a rare genetic disease caused by hepatic overproduction of oxalate that leads to kidney stones, nephrocalcinosis, kidney failure, and systemic oxalosis. Lumasiran, an investigational RNA interference (RNAi) therapeutic agent, reduces hepatic oxalate production by targeting glycolate oxidase. METHODS: In this double-blind, phase 3 trial, we randomly assigned (in a 2:1 ratio) patients with PH1 who were 6 years of age or older to receive subcutaneous lumasiran or placebo for 6 months (with doses given at baseline and at months 1, 2, 3, and 6). The primary end point was the percent change in 24-hour urinary oxalate excretion from baseline to month 6 (mean percent change across months 3 through 6). Secondary end points included the percent change in the plasma oxalate level from baseline to month 6 (mean percent change across months 3 through 6) and the percentage of patients with 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6. RESULTS: A total of 39 patients underwent randomization; 26 were assigned to the lumasiran group and 13 to the placebo group. The least-squares mean difference in the change in 24-hour urinary oxalate excretion (lumasiran minus placebo) was -53.5 percentage points (P<0.001), with a reduction in the lumasiran group of 65.4% and an effect seen as early as month 1. The between-group differences for all hierarchically tested secondary end points were significant. The difference in the percent change in the plasma oxalate level (lumasiran minus placebo) was -39.5 percentage points (P<0.001). In the lumasiran group, 84% of patients had 24-hour urinary oxalate excretion no higher than 1.5 times the upper limit of the normal range at month 6, as compared with 0% in the placebo group (P<0.001). Mild, transient injection-site reactions were reported in 38% of lumasiran-treated patients. CONCLUSIONS: Lumasiran reduced urinary oxalate excretion, the cause of progressive kidney failure in PH1. The majority of patients who received lumasiran had normal or near-normal levels after 6 months of treatment. (Funded by Alnylam Pharmaceuticals; ILLUMINATE-A ClinicalTrials.gov number, NCT03681184.).


Asunto(s)
Hiperoxaluria Primaria/tratamiento farmacológico , Oxalatos/orina , ARN Interferente Pequeño/uso terapéutico , Tratamiento con ARN de Interferencia , Adolescente , Adulto , Niño , Creatinina/orina , Método Doble Ciego , Femenino , Tasa de Filtración Glomerular , Humanos , Hiperoxaluria Primaria/sangre , Hiperoxaluria Primaria/complicaciones , Hiperoxaluria Primaria/orina , Cálculos Renales/prevención & control , Masculino , Persona de Mediana Edad , Oxalatos/sangre , Oxalatos/metabolismo , ARN Interferente Pequeño/efectos adversos , Adulto Joven
17.
Drugs ; 81(2): 277-282, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33405070

RESUMEN

Lumasiran (Oxlumo™) is a subcutaneously administered small interfering RNA (siRNA) targeting the mRNA for hydroxyacid oxidase 1 gene (HAO1; encodes glycolate oxidase) and was developed by Alnylam Pharmaceuticals for the treatment of primary hyperoxaluria type 1 (PH1). By silencing the gene encoding glycolate oxidase, lumasiran depletes glycolate oxidase and thereby inhibits the synthesis of oxalate, which is the toxic metabolite that is directly associated with the clinical manifestations of PH1. On 19 November 2020, lumasiran received its first global approval in the EU for the treatment of PH1 in all age groups. On 23 November 2020, lumasiran was approved in the USA for the treatment of adult and paediatric patients with PH1. This article summarizes the milestones in the development of lumasiran leading to this first approval.


Asunto(s)
Oxidorreductasas de Alcohol/antagonistas & inhibidores , Hiperoxaluria Primaria/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Adulto , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Niño , Humanos , Hiperoxaluria Primaria/metabolismo , Inyecciones Subcutáneas , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/administración & dosificación
18.
Am J Kidney Dis ; 77(5): 816-819, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32891627

RESUMEN

Primary hyperoxaluria type 1 (PH1) is a genetic disorder characterized by overproduction of oxalate and eventual kidney failure. Kidney failure is usually irreversible in PH1. However, in patients with PH1 homozygous for the G170R mutation (in which the glycine at amino acid 170 is replaced by an arginine), pyridoxine is an enzyme cofactor and decreases urinary oxalate excretion by reducing hepatic oxalate production. We report recovery from dialysis in 3 patients with PH1 homozygous for the G170R mutation in response to pharmacologic-dose pyridoxine treatment. Median age at initiation or resumption of pyridoxine treatment was 37 (range, 20-53) years, and median daily pyridoxine dose was 8.8 (range, 6.8-14.0) mg per kilogram of body weight. Duration of hemodialysis before recovery of kidney function was 10 (range, 5-19) months. Plasma oxalate concentration improved after recovery of kidney function. At a median of 3 (range, 2-46) months following discontinuation of hemodialysis, estimated glomerular filtration rate was 34 (range, 23-52) mL/min/1.73m2, plasma oxalate concentration was 8.8 (range, 4.6-11.3) µmol/L, and urinary oxalate excretion was 0.93 (range, 0.47-1.03) mmol/d. Kidney function was maintained during a median of 3.2 (range, 1.3-3.8) years of follow-up. These observations suggest that kidney failure may be reversible in a subset of patients with PH1 homozygous for the G170R mutation treated with pharmacologic-dose pyridoxine.


Asunto(s)
Hiperoxaluria Primaria/tratamiento farmacológico , Fallo Renal Crónico/terapia , Piridoxina/uso terapéutico , Complejo Vitamínico B/uso terapéutico , Adulto , Femenino , Homocigoto , Humanos , Hiperoxaluria Primaria/sangre , Hiperoxaluria Primaria/complicaciones , Fallo Renal Crónico/etiología , Fallo Renal Crónico/metabolismo , Persona de Mediana Edad , Oxalatos/sangre , Recuperación de la Función , Diálisis Renal , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/terapia , Transaminasas/genética , Transaminasas/metabolismo , Adulto Joven
19.
Mol Genet Metab ; 131(1-2): 171-180, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32792227

RESUMEN

Primary Hyperoxaluria type I (PH1) is a rare disease caused by mutations in the AGXT gene encoding alanine:glyoxylate aminotransferase (AGT), a liver enzyme involved in the detoxification of glyoxylate, the failure of which results in accumulation of oxalate and kidney stones formation. The role of protein misfolding in the AGT deficit caused by most PH1-causing mutations is increasingly being recognized. In addition, the genetic background in which a mutation occurs is emerging as a critical risk factor for disease onset and/or severity. Based on these premises, in this study we have analyzed the clinical, biochemical and cellular effects of the p.Ile56Asn mutation, recently described in a PH1 patient, as a function of the residue at position 11, a hot-spot for both polymorphic (p.Pro11Leu) and pathogenic (p.Pro11Arg) mutations. We have found that the p.Ile56Asn mutation induces a structural defect mostly related to the apo-form of AGT. The effects are more pronounced when the substitution of Ile56 is combined with the p.Pro11Leu and, at higher degree, the p.Pro11Arg mutation. As compared with the non-pathogenic forms, AGT variants display reduced expression and activity in mammalian cells. Vitamin B6, a currently approved treatment for PH1, can overcome the effects of the p.Ile56Asn mutation only when it is associated with Pro at position 11. Our results provide a first proof that the genetic background influences the effects of PH1-causing mutations and the responsiveness to treatment and suggest that molecular and cellular studies can integrate clinical data to identify the best therapeutic strategy for PH1 patients.


Asunto(s)
Hiperoxaluria Primaria/tratamiento farmacológico , Hiperoxaluria Primaria/genética , Hígado/efectos de los fármacos , Transaminasas/genética , Línea Celular , Cristalografía por Rayos X , Glioxilatos/metabolismo , Humanos , Hiperoxaluria Primaria/patología , Hígado/metabolismo , Hígado/patología , Mutación/genética , Conformación Proteica , Pliegue de Proteína/efectos de los fármacos , Relación Estructura-Actividad , Transaminasas/ultraestructura , Vitamina B 6/química , Vitamina B 6/farmacología
20.
Pediatr Nephrol ; 35(9): 1787-1789, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32418144

RESUMEN

BACKGROUND: Primary hyperoxaluria type 1 (PH1) is a multisystemic metabolic disorder caused by an excessive production of oxalate by the liver. The majority of patients presenting in early infancy have end-stage renal disease (ESRD). While awaiting the results of sRNAi trials, the current standard treatment is combined liver-kidney transplantation. Recently, Stiripentol has been reported as a promising drug in the treatment of primary hyperoxaluria by reducing urinary oxalate (UOx). Stiripentol is an anti-convulsive drug used in the treatment of children suffering from Dravet syndrome. It causes blockage of the last step in oxalate production by inhibition of hepatic lactate dehydrogenase 5 (LDH5). CASE: We administered Stiripentol as compassionate use in an anuric infant with dialysis-dependent PH1 over a period of 4 months. Although achieving plasma concentrations of Stiripentol that were recently reported to lower UOx excretion, we did not observe significant reduction to plasma oxalate concentrations (POx). CONCLUSION: We conclude that Stiripentol may not be useful to reduce POx in PH patients with advanced chronic kidney disease (CKD), but larger studies are needed to confirm this finding.


Asunto(s)
Dioxolanos/administración & dosificación , Hiperoxaluria Primaria/tratamiento farmacológico , Humanos , Lactante , Masculino , Oxalatos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...