Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Genet Metab ; 139(4): 107631, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37453187

RESUMO

Acid sphingomyelinase deficiency (ASMD) is a rare LSD characterized by lysosomal accumulation of sphingomyelin, primarily in macrophages. With the recent availability of enzyme replacement therapy, the need for biomarkers to assess severity of disease has increased. Glycoprotein non-metastatic protein B (GPNMB) plasma levels were demonstrated to be elevated in Gaucher disease. Given the similarities between Gaucher disease and ASMD, the hypothesis was that GPNMB might be a potential biochemical marker for ASMD as well. Plasma samples of ASMD patients were analyzed and GPNMB plasma levels were compared to those of healthy volunteers. Visceral disease severity was classified as severe when splenic, hepatic and pulmonary manifestations were all present and as mild to moderate if this was not the case. Median GPNMB levels in 67 samples of 19 ASMD patients were 185 ng/ml (range 70-811 ng/ml) and were increased compared to 10 healthy controls (median 36 ng/ml, range 9-175 ng/ml, p < 0.001). Median plasma GPNMB levels of ASMD patients with mild to moderate visceral disease compared to patients with severe visceral disease differed significantly and did not overlap (respectively 109 ng/ml, range 70-304 ng/ml and 325 ng/ml, range 165-811 ng/ml, p < 0.001). Correlations with other biochemical markers of ASMD (i.e. chitotriosidase activity, CCL18 and lysosphingomyelin, respectively R = 0.28, p = 0.270; R = 0.34, p = 0.180; R = 0.39, p = 0.100) and clinical parameters (i.e. spleen volume, liver volume, diffusion capacity and forced vital capacity, respectively R = 0.59, p = 0.061, R = 0.5, p = 0.100, R = 0.065, p = 0.810, R = -0.38, p = 0.160) could not be established within this study. The results of this study suggest that GPNMB might be suitable as a biomarker of visceral disease severity in ASMD. Correlations between GPNMB and biochemical or clinical markers of ASMD and response to therapy have to be studied in a larger cohort.


Assuntos
Glicoproteínas de Membrana , Doença de Niemann-Pick Tipo B , Humanos , Masculino , Feminino , Criança , Adolescente , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Glicoproteínas de Membrana/sangue , Doença de Niemann-Pick Tipo B/sangue , Doença de Niemann-Pick Tipo B/diagnóstico , Biomarcadores/sangue , Doença de Niemann-Pick Tipo A/sangue , Doença de Niemann-Pick Tipo A/diagnóstico , Gravidade do Paciente , Doença de Gaucher/sangue , Doença de Gaucher/diagnóstico , Estudos de Casos e Controles
2.
Orphanet J Rare Dis ; 17(1): 383, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36271424

RESUMO

INTRODUCTION: Several new treatment modalities are being developed for lysosomal storage disorders (LSDs), including gene therapy. As the currently available treatment options and their influence on disease progression differ greatly within the spectrum of LSDs, willingness to undergo gene therapy might vary among patients with LSDs and/or their representatives. The width of the LSD spectrum is illustrated by the differences between type 1 Gaucher disease, Fabry disease and Mucopolysaccharidosis type III (MPS III). For type 1 Gaucher and Fabry disease several therapies are available, resulting in a near normal or improved, but individually varying, prognosis. No treatment options are available for MPS III. AIM: To identify factors influencing patients' and/or their representatives' decisions regarding undergoing gene therapy. METHODS: Focus group discussions and semi-structured interviews were conducted with patients with type 1 Gaucher disease, Fabry disease and MPS III. Parents of MPS III patients were included as patients' representatives. RESULTS: Nine Gaucher patients, 23 Fabry patients, two adult MPS III patients and five parents of MPS III patients participated in the study. The five main themes that arose were: outcome of gene therapy, risks and side effects, burden of gene therapy treatment, current situation and ethical aspects. Participants' views ranged from hesitance to eagerness to undergo gene therapy, which seemed to be mostly related to disease severity and currently available treatment options. Severe disease, limited treatment options and limited effectiveness of current treatment augmented the willingness to choose gene therapy. Gaucher and Fabry patients deemed the burden of treatment important. Fabry and MPS III patients and parents considered outcome important, suggesting hope for improvement. When asked to rank the factors discussed in the focus group discussions, Gaucher patients ranked outcome low, which could indicate a more cautious attitude towards gene therapy. CONCLUSION: This study underlines the importance of exploring patients' needs and expectations before using limited resources in the development of therapies for patient groups of which a significant subset may not be willing to undergo that specific therapy.


Assuntos
Doença de Fabry , Doença de Gaucher , Doenças por Armazenamento dos Lisossomos , Mucopolissacaridose III , Adulto , Humanos , Doença de Fabry/genética , Doença de Fabry/terapia , Doença de Gaucher/genética , Doença de Gaucher/terapia , Terapia Genética , Doenças por Armazenamento dos Lisossomos/terapia , Doenças por Armazenamento dos Lisossomos/tratamento farmacológico , Lisossomos
3.
Mol Genet Metab ; 130(1): 16-26, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32088119

RESUMO

Acid Sphingomyelinase Deficiency (ASMD), or Niemann-Pick type A/B disease, is a rare lipid storage disorder leading to accumulation of sphingomyelin and its precursors primarily in macrophages. The disease has a broad phenotypic spectrum ranging from a fatal infantile form with severe neurological involvement (the infantile neurovisceral type) to a primarily visceral form with different degrees of pulmonary, liver, spleen and skeletal involvement (the chronic visceral type). With the upcoming possibility of treatment with enzyme replacement therapy, the need for biomarkers that predict or reflect disease progression has increased. Biomarkers should be validated for their use as surrogate markers of clinically relevant endpoints. In this review, clinically important endpoints as well as biochemical and imaging markers of ASMD are discussed and potential new biomarkers are identified. We suggest as the most promising biomarkers that may function as surrogate endpoints in the future: diffusion capacity measured by spirometry, spleen volume, platelet count, low-density lipoprotein cholesterol, liver fibrosis measured with a fibroscan, lysosphingomyelin and walked distance in six minutes. Currently, no biomarkers have been validated. Several plasma markers of lipid-laden cells, fibrosis or inflammation are of high potential as biomarkers and deserve further study. Based upon current guidelines for biomarkers, recommendations for the validation process are provided.


Assuntos
Doença de Niemann-Pick Tipo A/sangue , Doença de Niemann-Pick Tipo A/diagnóstico por imagem , Doença de Niemann-Pick Tipo B/sangue , Doença de Niemann-Pick Tipo B/diagnóstico por imagem , Esfingolipídeos/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , Doenças Ósseas/imunologia , Doenças Ósseas/metabolismo , Doenças Cardiovasculares/sangue , LDL-Colesterol/sangue , Humanos , Hepatopatias/sangue , Hepatopatias/diagnóstico por imagem , Hepatopatias/enzimologia , Pneumopatias/diagnóstico por imagem , Pneumopatias/enzimologia , Pneumopatias/metabolismo , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/metabolismo , Doença de Niemann-Pick Tipo A/fisiopatologia , Doença de Niemann-Pick Tipo B/fisiopatologia , Baço/diagnóstico por imagem , Baço/crescimento & desenvolvimento , Baço/patologia
4.
J Clin Lipidol ; 13(2): 272-278, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30795984

RESUMO

BACKGROUND: Homozygous familial hypercholesterolemia (hoFH) is either diagnosed on the identification of pathogenic genetic variants in LDLR, APOB, or PCSK9 or by phenotypic parameters of which an extremely elevated LDL-C level >13 mmol/L (>500 mg/dL) is the most prominent hallmark. Little is known about the clinical spectrum in children with hoFH. OBJECTIVE: We set out to investigate the phenotypical spectrum of genetically defined hoFH in our pediatric cohort and evaluated how many pediatric patients, now classified as heterozygous, carry a second mutation, which would reclassify these patients as hoFH. METHODS: We analyzed the data of a total of 1903 children with molecularly proven FH. Subsequently we performed candidate gene sequencing in the cohort of heterozygous familial hypercholesterolemia children in whom the LDL-C level was above the lowest level measured in the pediatric patients with hoFH. RESULTS: Of our 13 hoFH children, 8 (62%) had LDL-C levels below the clinical hoFH criteria of 13 mmol/L (500 mg/dL). In the remaining 1890 patients with heterozygous familial hypercholesterolemia, 64 (3.4%) had LDL-C levels equal to or above the lowest LDL-C level in a patient with hoFH carrying 2 deleterious variants (8.36 mmol/L or 323.3 mg/dL). No additional pathogenic variants in LDLR and APOB were identified. In 2 related patients, a PCSK9 gain of function mutation was found. CONCLUSION: We show that LDL-C levels vary among pediatric patients with molecularly proven hoFH, and that most of these patients do not meet the clinical LDL-C criteria for hoFH. The levels overlap with LDL-C levels in true heterozygous patients. This warrants a critical reappraisal of the current LDL-C cutoffs for the phenotypic diagnosis of hoFH in children.


Assuntos
Variação Genética , Homozigoto , Hiperlipoproteinemia Tipo II/genética , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Fenótipo , Adulto Jovem
5.
Atherosclerosis ; 278: 174-179, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30286343

RESUMO

BACKGROUND AND AIMS: Lysosomal acid lipase deficiency (LAL-D) is a lysosomal storage disorder. In severe cases, it can cause life-threatening organ failure due to lipid substrates accumulation. However, mild phenotypes of this disorder are increasingly recognized. The aim of this study is to determine the number of missed LAL-D patients in a large pediatric hospital population. METHODS: In a retrospective data mining study, the medical files of children, who visited the outpatient clinic at a university hospital between 2000 and 2016, with high plasma low density lipoprotein cholesterol (LDL-C) levels, were evaluated. Previously developed LAL-D screening criteria, with lipid and alanine aminotransferase (ALT) values adjusted for children, were used to analyze which children are suspect for LAL-D. For suspicion of LAL-D, at least 3 out of 5 screening criteria had to be met. Subsequently data on presentation and follow-up were collected to determine if the clinical picture was compatible with LAL-D. RESULTS: We identified 2037 children with high LDL-C levels. Of those, 36 children complied with ≥3 screening criteria. Thirty-one of those had an underlying disorder other than LAL-D that explained the abnormalities and, in the 5 remaining children, ALT and lipid levels normalized spontaneously, thus excluding LAL-D. CONCLUSIONS: This study shows that retrospective data mining is unlikely to yield a significant number of LAL-D cases in children. The screening algorithm adjusted for children seems useful and accurate in the selection of children for further testing, suggesting it can be applied prospectively, although further validation is warranted.


Assuntos
Doença do Armazenamento de Colesterol Éster/diagnóstico , LDL-Colesterol/sangue , Mineração de Dados , Doença de Wolman/diagnóstico , Adolescente , Alanina Transaminase/metabolismo , Algoritmos , Cardiologia/normas , Criança , Pré-Escolar , Doença do Armazenamento de Colesterol Éster/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Fígado/metabolismo , Masculino , Programas de Rastreamento/métodos , Pacientes Ambulatoriais , Fenótipo , Estudos Retrospectivos , Esterol Esterase/genética , Doença de Wolman/metabolismo , Doença de Wolman
6.
J Clin Lipidol ; 11(2): 507-514, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28502508

RESUMO

BACKGROUND: Patients with autosomal dominant hypercholesterolemia (ADH), caused by mutations in either low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB), or proprotein convertase subtilisin-kexin type 9 (PCSK9) are characterized by high low-density lipoprotein cholesterol levels and in some studies also high lipoprotein(a) (Lp(a)) levels were observed. The question remains whether this effect on Lp(a) levels is gene-dose-dependent in individuals with either 0, 1, or 2 LDLR or APOB mutations. OBJECTIVE: We set out to study whether Lp(a) levels differ among bi-allelic ADH mutation carriers, and their relatives, in the Netherlands. METHODS: Bi-allelic ADH mutation carriers were identified in the database of the national referral laboratory for DNA diagnostics of inherited dyslipidemias. Family members were invited by the index cases to participate. Clinical parameters and Lp(a) levels were measured in bi-allelic ADH mutation carriers and their heterozygous and unaffected relatives. RESULTS: We included a total of 119 individuals; 34 bi-allelic ADH mutation carriers (20 homozygous/compound heterozygous LDLR mutation carriers (HoFH), 2 homozygous APOB mutation carriers (HoFDB), and 12 double heterozygotes for an LDLR and APOB mutation), 63 mono-allelic ADH mutation carriers (50 heterozygous LDLR [HeFH], 13 heterozygous APOB [HeFDB] mutation carriers), and 22 unaffected family members. Median Lp(a) levels in unaffected relatives, HeFH, and HoFH patients were 19.9 (11.1-41.5), 24.4 (5.9-70.6), and 47.3 (14.9-111.7) mg/dL, respectively (P = .150 for gene-dose dependency). Median Lp(a) levels in HeFDB and HoFDB patients were 50.3 (18.7-120.9) and 205.5 (no interquartile range calculated), respectively (P = .012 for gene-dose-dependency). Double heterozygous carriers of LDLR and APOB mutations had median Lp(a) levels of 27.0 (23.5-45.0), which did not significantly differ from HoFH and HoFDB patients (P = .730 and .340, respectively). CONCLUSION: A (trend toward) increased plasma Lp(a) levels in homozygous ADH patients compared with both heterozygous ADH and unaffected relatives was observed. Whether increased Lp(a) levels in homozygous ADH patients add to the increased cardiovascular disease risk and whether this risk can be reduced by therapies that lower both low-density lipoprotein cholesterol and Lp(a) levels remains to be elucidated.


Assuntos
Homozigoto , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/genética , Lipoproteínas/sangue , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Adulto Jovem
7.
J Clin Lipidol ; 10(6): 1462-1469, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27919364

RESUMO

INTRODUCTION: Autosomal dominant hypercholesterolemia (ADH), characterized by high-plasma low-density lipoprotein cholesterol (LDL-C) levels and premature cardiovascular disease (CVD) risk, is caused by mutations in LDLR, APOB, and/or PCSK9. OBJECTIVE: To describe the clinical characteristics of "double-heterozygous carriers," with 2 mutations in 2 different ADH causing genes, that is, LDLR and APOB or LDLR and PCSK9. METHODS: Double heterozygotes were identified in the database of the national referral laboratory for DNA diagnostics of inherited dyslipidemias. We collected the medical data (comprising lipids and CVD events) from double heterozygotes and compared these with data from their heterozygous and unaffected relatives and homozygote/compound heterozygous LDLR mutation carriers, identified in a previously described cohort (n = 45). RESULTS: A total of 28 double heterozygotes (23 LDLR/APOB and 5 LDLR/PCSK9 mutation carriers) were identified. Off treatment, LDL-C levels were significantly higher in double heterozygotes (mean ± SD, 8.4 ± 2.8 mmol/L) compared with 28 heterozygous (5.6 ± 2.2) and 18 unaffected relatives (2.5 ± 1.1; P ≤ .01 for all comparisons) and significantly lower compared with homozygous/compound heterozygous LDLR mutation carriers (13.0 ± 5.1; P < .001). CONCLUSIONS: Double-heterozygous carriers of mutations in ADH genes express an intermediate phenotype compared with heterozygous and homozygous/compound heterozygous carriers and might well be misconceived to suffer from a severe form of heterozygous ADH. The molecular identification of double heterozygosity is of relevance from both a screening and an educational perspective.


Assuntos
Hiperlipoproteinemia Tipo II/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Apolipoproteínas B/genética , Doenças Cardiovasculares/etiologia , Criança , LDL-Colesterol/sangue , Feminino , Heterozigoto , Humanos , Hiperlipoproteinemia Tipo II/complicações , Hiperlipoproteinemia Tipo II/genética , Masculino , Pessoa de Meia-Idade , Fenótipo , Polimorfismo de Nucleotídeo Único , Pró-Proteína Convertase 9/genética , Receptores de LDL/genética , Estudos Retrospectivos , Fatores de Risco , Adulto Jovem
8.
Atherosclerosis ; 251: 263-265, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27423329

RESUMO

BACKGROUND AND AIMS: We recently identified lysosomal acid lipase (LAL) deficiency, a recessive disease caused by mutations in LIPA, in 3 patients with a clinical diagnosis of familial hypercholesterolemia (FH). We aimed to determine the prevalence of LIPA mutations among individuals with a clinical FH diagnosis. METHODS: In 276 patients with phenotypic FH, in whom no genetic basis for their phenotype was found, LIPA was sequenced. All variants were assessed for pathogenicity using a literature search and in silico prediction models. RESULTS: We included 213 adults and 63 children with mean (±SD) LDL-C levels of 7.8 ± 1.3 and 4.4 ± 1.5 mmol/L, respectively. Twenty-one variants were identified. Six patients were heterozygous carrier of a (potentially) pathogenic mutation. No homozygous LIPA mutation carriers were identified. CONCLUSIONS: Our data show that LAL deficiency was not missed as diagnosis in our study population but the frequency of heterozygous LIPA mutations implies that the FH population might be relatively enriched with LIPA mutation carriers.


Assuntos
Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Mutação , Esterol Esterase/genética , Doença de Wolman/genética , Adolescente , Adulto , Idoso , Criança , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Prevalência , Triglicerídeos/sangue , Doença de Wolman
9.
Atherosclerosis ; 248: 238-44, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27017151

RESUMO

AIMS: Homozygous familial hypercholesterolaemia (HoFH) is a rare disorder usually caused by mutations in both alleles of the low-density lipoprotein receptor gene (LDLR). Premature death, often before the age of 20 years, was a common fate for patients with HoFH prior to the introduction of statins in 1990 and the use of lipoprotein apheresis. Consequently, HoFH has been widely considered a condition exclusive to a population comprising very young patients with extremely high LDL cholesterol (LDL-C) levels. However, recent epidemiologic and genetic studies have shown that the HoFH patient population is far more diverse in terms of age, LDL-C levels, and genetic aetiology than previously realised. We set out to investigate the clinical characteristics regarding age and LDL-C ranges of patients with HoFH. METHODS AND RESULTS: We analysed the data from 3 recent international studies comprising a total of 167 HoFH patients. The age of the patients ranged from 1 to 75 years, and a large proportion of the patients, both treated and untreated, exhibited LDL-C levels well below the recommended clinical diagnostic threshold for HoFH. LDL-C levels ranged from 4.4 mmol/L to 27.2 mmol/L (170-1052 mg/dL) for untreated patients, and from 2.6 mmol/L to 20.3 mmol/L (101-785 mg/dL) for treated patients. When patients were stratified according to LDLR functionality, a similarly wide range of age and LDL-C values was observed regardless of LDLR mutation status. CONCLUSION: These results demonstrate that HoFH is not restricted to very young patients or those with extremely high LDL-C levels.


Assuntos
Homozigoto , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Adolescente , Adulto , Idoso , Alelos , Anticolesterolemiantes/uso terapêutico , Criança , Pré-Escolar , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Estudos de Coortes , Análise Mutacional de DNA , Interpretação Estatística de Dados , Método Duplo-Cego , Humanos , Lactente , Cooperação Internacional , Pessoa de Meia-Idade , Mutação , Oligonucleotídeos/uso terapêutico , Fenótipo , Receptores de LDL/genética , Sistema de Registros , Estudos Retrospectivos , Adulto Jovem
10.
Circ Cardiovasc Genet ; 8(6): 823-31, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26374825

RESUMO

BACKGROUND: Patients with PCSK9 gene gain of function (GOF) mutations have a rare form of autosomal dominant hypercholesterolemia. However, data examining their clinical characteristics and geographic distribution are lacking. Furthermore, no randomized treatment study in this population has been reported. METHODS AND RESULTS: We compiled clinical characteristics of PCSK9 GOF mutation carriers in a multinational retrospective, cross-sectional, observational study. We then performed a randomized placebo-phase, double-blind study of alirocumab 150 mg administered subcutaneously every 2 weeks to 13 patients representing 4 different PCSK9 GOF mutations with low-density lipoprotein cholesterol (LDL-C) ≥70 mg/dL on their current lipid-lowering therapies at baseline. Observational study: among 164 patients, 16 different PCSK9 GOF mutations distributed throughout the gene were associated with varying severity of untreated LDL-C levels. Coronary artery disease was common (33%; average age of onset, 49.4 years), and untreated LDL-C concentrations were higher compared with matched carriers of mutations in the LDLR (n=2126) or apolipoprotein B (n=470) genes. Intervention study: in PCSK9 GOF mutation patients randomly assigned to receive alirocumab, mean percent reduction in LDL-C at 2 weeks was 62.5% (P<0.0001) from baseline, 53.7% compared with placebo-treated PCSK9 GOF mutation patients (P=0.0009; primary end point). After all subjects received 8 weeks of alirocumab treatment, LDL-C was reduced by 73% from baseline (P<0.0001). CONCLUSIONS: PCSK9 GOF mutation carriers have elevated LDL-C levels and are at high risk of premature cardiovascular disease. Alirocumab, a PCSK9 antibody, markedly lowers LDL-C levels and seems to be well tolerated in these patients. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique Identifier: NCT01604824.


Assuntos
Anticorpos Monoclonais/administração & dosagem , LDL-Colesterol/sangue , Doença da Artéria Coronariana , Hiperlipoproteinemia Tipo II , Mutação , Pró-Proteína Convertases , Serina Endopeptidases , Adolescente , Adulto , Idoso , Anticorpos Monoclonais Humanizados , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/genética , Método Duplo-Cego , Feminino , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/genética , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/antagonistas & inibidores , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
11.
Atherosclerosis ; 241(2): 597-606, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26115072

RESUMO

In this review, we discuss the screening and treatment of familial hypercholesterolemia (FH), an autosomal dominant inherited disease, characterized by severely increased levels of low-density lipoprotein cholesterol (LDL-C) and increased risk for premature coronary heart disease (CHD). Genetic family based cascade screening for FH was shown to be cost-effective and a screening program with such an approach was carried out in the Netherlands from 1994 to 2014. Over 64,000 persons have participated in this program of whom 40.3% were found to carry an FH causing mutation. We will discuss the results of this screening program, as well as the scientific opportunities it has provided. Currently, statins and ezetimibe are the only registered LDL-C lowering treatment options for FH patients. Many of them do not attain the treatment goals that are recommended by treatment guidelines. In this review, we will also provide a comprehensive overview of promising new modalities that could lower LDL-C in FH patients.


Assuntos
Doença das Coronárias/diagnóstico , Doença das Coronárias/terapia , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/terapia , Anticolesterolemiantes/uso terapêutico , Apolipoproteínas B/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Doença das Coronárias/genética , Análise Custo-Benefício , Ezetimiba/uso terapêutico , Predisposição Genética para Doença , Humanos , Hiperlipoproteinemia Tipo II/genética , Programas de Rastreamento , Mutação , Países Baixos , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/metabolismo , Receptores de LDL/genética , Risco , Serina Endopeptidases/metabolismo
12.
Curr Opin Lipidol ; 26(3): 200-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25950706

RESUMO

PURPOSE OF REVIEW: Homozygous autosomal dominant hypercholesterolemia (hoADH) is a rare genetic disorder caused by mutations in LDL receptor, apolipoprotein B, and/or proprotein convertase subtilisin-kexin type 9. Both the genetic mutations and the clinical phenotype vary largely among individual patients, but patients with hoADH are typically characterized by extremely elevated LDL-cholesterol (LDL-C) levels, and a very high-risk for premature cardiovascular disease. Current lipid-lowering therapies include bile acid sequestrants, statins, and ezetimibe. To further decrease LDL-C levels in hoADH, lipoprotein apheresis is recommended, but this therapy is not available in all countries. RECENT FINDINGS: Recently, the microsomal triglyceride transfer protein inhibitor lomitapide and the RNA antisense inhibitor of apolipoprotein B mipomersen were approved by the Food and Drug Administration/European Medicine Agency and the Food and Drug Administration, respectively. Several other LDL-C-lowering strategies and therapeutics targeting the HDL-C pathway are currently in the clinical stage of development. SUMMARY: Novel therapies have been introduced for LDL-C-lowering and innovative drug candidates for HDL-C modulation for the treatment of hoADH. Here, we review the current available literature on the prevalence, diagnosis, and therapeutic strategies for hoADH.


Assuntos
Hiperlipoproteinemia Tipo II/terapia , Animais , Anticolesterolemiantes/farmacologia , Anticolesterolemiantes/uso terapêutico , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , HDL-Colesterol/administração & dosagem , Terapia Genética , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/epidemiologia , Prevalência , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/antagonistas & inibidores , Serina Endopeptidases
13.
Eur Heart J ; 36(9): 560-5, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24585268

RESUMO

AIMS: Homozygous autosomal dominant hypercholesterolaemia (hoADH), an orphan disease caused by mutations in low-density lipoprotein receptor (LDLR), apolipoprotein B (APOB), or proprotein convertase subtilisin-kexin type 9 (PCSK9), is characterized by elevated plasma low-density lipoprotein-cholesterol (LDL-C) levels and high risk for premature cardiovascular disease (CVD). The exact prevalence of molecularly defined hoADH is unknown. Therefore, we investigated the prevalence and phenotypical characteristics of this disease in an open society, i.e. the Netherlands. METHODS AND RESULTS: The database of the nationwide ADH molecular diagnostic center was queried to identify all molecularly defined hoADH patients. Carriers of non-pathogenic mutations were excluded. Medical records were analysed for data regarding lipid levels and CVD events. Of 104,682 individuals screened for molecular defects, 49 were classified as hoADH (0.05%); 20 were true homozygotes, 25 were compound heterozygotes for LDLR mutations, and 4 were homozygous for APOB mutations. No bi-allelic PCSK9 mutation carriers were identified. Consequently, the prevalence of hoADH was estimated to be ∼1 : 300,000. Mean LDL-C levels prior to lipid-lowering treatment were 12.9 ± 5.1 mmol/L (range 4.4-21.5 mmol/L). Surprisingly, only 50% of the patients met the clinical criteria for hoADH (LDL-C >13.0 mmol/L); 29% of patients suffered from a CVD event. CONCLUSION: The prevalence of molecularly defined hoADH is much higher and the clinical phenotype is more variable than previously assumed. In light of the fact that novel therapies are, or will be registered for the treatment of hoADH patients, an uniform definition of hoADH either as a phenotypic or molecular entity is warranted in order to identify patients who are considered to be eligible for these novel agents.


Assuntos
Hiperlipoproteinemia Tipo II/epidemiologia , Adolescente , Adulto , Idoso , Apolipoproteína B-100/genética , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Heterozigoto , Homozigoto , Humanos , Hiperlipoproteinemia Tipo II/genética , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mutação/genética , Países Baixos/epidemiologia , Fenótipo , Prevalência , Prognóstico , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/genética , Receptores de LDL/genética , Serina Endopeptidases/genética , Adulto Jovem
14.
J Am Coll Cardiol ; 64(14): 1418-26, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25277610

RESUMO

BACKGROUND: Patients with familial hypercholesterolemia (FH) are characterized by elevated atherogenic lipoprotein particles, predominantly low-density lipoprotein cholesterol (LDL-C), which is associated with accelerated atherogenesis and increased cardiovascular risk. OBJECTIVES: This study used (18)F-fluorodeoxyglucose positron emission tomography ((18)FDG-PET) to investigate whether arterial inflammation is higher in patients with FH and, moreover, whether lipoprotein apheresis attenuates arterial wall inflammation in FH patients. METHODS: In total, 38 subjects were recruited: 24 FH patients and 14 normolipidemic controls. All subjects underwent FDG-PET imaging at baseline. Twelve FH patients who met the criteria for lipoprotein apheresis underwent apheresis procedures followed by a second FDG-PET imaging 3 days (range 1 to 4 days) after apheresis. Subsequently, the target-to-background ratio (TBR) of FDG uptake within the arterial wall was assessed. RESULTS: In FH patients, the mean arterial TBR was higher compared with healthy controls (2.12 ± 0.27 vs. 1.92 ± 0.19; p = 0.03). A significant correlation was observed between baseline arterial TBR and LDL-C (R = 0.37; p = 0.03) that remained significant after adjusting for statin use (ß = 0.001; p = 0.02) and atherosclerosis risk factors (ß = 0.001; p = 0.03). LDL-C levels were significantly reduced after lipoprotein apheresis (284 ± 118 mg/dl vs. 127 ± 50 mg/dl; p < 0.001). There was a significant reduction of arterial inflammation after lipoprotein apheresis (TBR: 2.05 ± 0.31 vs. 1.91 ± 0.33; p < 0.02). CONCLUSIONS: The arterial wall of FH patients is characterized by increased inflammation, which is markedly reduced after lipoprotein apheresis. This lends support to a causal role of apoprotein B-containing lipoproteins in arterial wall inflammation and supports the concept that lipoprotein-lowering therapies may impart anti-inflammatory effects by reducing atherogenic lipoproteins.


Assuntos
Artérias/patologia , Remoção de Componentes Sanguíneos/métodos , Hiperlipoproteinemia Tipo II/terapia , Inflamação/terapia , Idoso , Aterosclerose/sangue , Estudos de Casos e Controles , LDL-Colesterol/sangue , Estudos Transversais , Feminino , Fluordesoxiglucose F18/química , Humanos , Hiperlipoproteinemia Tipo II/complicações , Inflamação/complicações , Lipoproteínas/química , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Tomografia por Emissão de Pósitrons/métodos , Estudos Prospectivos , Fatores de Risco , Tomografia Computadorizada por Raios X
15.
Lancet Diabetes Endocrinol ; 2(6): 455-63, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24731671

RESUMO

BACKGROUND: Eprotirome is a liver-selective thyroid hormone receptor agonist that has been shown to lower plasma LDL cholesterol concentrations in previous phase 1 and 2 studies of patients with dyslipidaemia. We aimed to assess the long-term safety and efficacy of 50 µg and 100 µg eprotirome in patients with familial hypercholesterolaemia. METHODS: For this randomised, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial, we enrolled patients between Oct 3, 2011, and Feb 14, 2012, at 53 sites in 11 countries in Europe, Africa, and south Asia. Patients were eligible for enrolment if they were aged 18 years or older, diagnosed with heterozygous familial hypercholesterolaemia, and had not reached target LDL cholesterol concentrations after at least 8 weeks of statin therapy with or without ezetimibe. We used a computer-generated randomisation sequence to allocate patients to one of three groups: 50 µg eprotirome, 100 µg eprotirome, or placebo. This trial was planned for 52-76 weeks, with primary efficacy analysis at 12 weeks, but it was prematurely terminated when another study found that eprotirome causes cartilage damage in dogs. Although it was impossible to meet the predefined study outcomes, we analysed changes in the concentrations of LDL cholesterol and other lipids, liver parameters, thyroid hormone concentrations, and adverse effects of treatment with eprotirome versus placebo at 6 weeks of treatment. Analysis was done in all patients who received 6 weeks of treatment. This study is registered with ClinicalTrials.gov, number NCT01410383. FINDINGS: We enrolled 236 patients, randomly allocating 80 to receive placebo, 79 to receive 50 µg eprotirome, and 77 to receive 100 µg eprotirome. 69 patients reached the 6 week timepoint (23 given placebo, 24 given 50 µg eprotirome, and 22 given 100 µg eprotirome). Mean LDL cholesterol concentrations increased by 9% (95% CI -2 to 20) in the placebo group, decreased by 12% (-28 to 4%; p=0.0677 vs placebo) in the 50 µg eprotirome group, and decreased by 22% (-32 to -13%; p=0.0045 vs placebo) in the 100 µg eprotirome group. We noted statistically significant increases between both eprotirome groups and placebo in aspartate aminotransferase (AST; p<0.0001), alanine aminotransferase (ALT; p<0.0001), conjugated bilirubin (p=0.0006), and gamma-glutamyltranspeptidase (p<0.0001). Four patients had to discontinue or interrupt study treatment before trial termination due to AST increases between the upper limit of normal (ULN) and six times ULN, and ALT concentrations between three and seven times ULN. Although we detected no changes in serum concentrations of thyroid-stimulating hormone or free tri-iodothyronine, free tetra-iodothyronine decreased by 19% (23 to 16) in the 50 µg eprotirome group and 27% (30 to 23) in the 100 µg eprotirome group (p<0.0001 vs placebo for both groups). INTERPRETATION: Our findings show that eprotirome can lower LDL cholesterol concentrations in patients with familial hypercholesterolaemia when added to conventional statin treatment with or without ezetimibe, but that it has the potential to induce liver injury. These findings, along with findings of cartilage damage in dogs, raise serious doubts about selective thyroid hormone mimetics as a therapeutic approach to lower LDL cholesterol concentrations. FUNDING: Karo Bio AB.


Assuntos
Anilidas/uso terapêutico , Anticolesterolemiantes/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/epidemiologia , LDL-Colesterol/efeitos dos fármacos , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Fígado/efeitos dos fármacos , África , Alanina Transaminase/efeitos dos fármacos , Análise de Variância , Anilidas/efeitos adversos , Anticolesterolemiantes/efeitos adversos , Apolipoproteína B-100/efeitos dos fármacos , Ásia , LDL-Colesterol/sangue , Método Duplo-Cego , Europa (Continente) , Feminino , Humanos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/epidemiologia , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento
16.
Curr Opin Lipidol ; 24(4): 301-6, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23759796

RESUMO

PURPOSE OF REVIEW: Mipomersen has been approved by the US Food and Drug Administration as an orphan drug for patients with homozygous familial hypercholesterolemia (HoFH). In contrast, the European Medicines Agency advised negatively on the use of mipomersen. In this review, we discuss the efficacy and safety considerations for this discrepancy. RECENT FINDINGS: On the basis of the results of clinical trials with mipomersen, safety concerns have been raised regarding cardiovascular risk reduction and development of hepatic steatosis. In addition, (long-term) tolerability concerns have been raised predominantly regarding injection site reactions. A pooled analysis of cardiovascular events in phase III trials with mipomersen did not provide evidence for either a positive or negative effect on cardiovascular disease. Although long-term studies with mipomersen are eagerly awaited, hepatic fat content appears to stabilize after 6-12 months notwithstanding continued mipomersen administration. SUMMARY: HoFH is a disease with an unmet medical need for new lipid-lowering therapies. On the basis of a mean 2.9 mmol/l LDL-cholesterol reduction, mipomersen is expected to reduce cardiovascular risk in HoFH. Available evidence suggests that the fat accumulation associated with this treatment differs from steatohepatitis, which is a progressive and damaging liver disease. No evidence is available suggesting that injection site reactions because of mipomersen treatment will result in safety issues.


Assuntos
Anticolesterolemiantes/uso terapêutico , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Animais , Anticolesterolemiantes/efeitos adversos , Anticolesterolemiantes/farmacologia , LDL-Colesterol/sangue , Ensaios Clínicos Fase III como Assunto , Aprovação de Drogas , Europa (Continente) , Fígado Gorduroso/induzido quimicamente , Humanos , Hiperlipoproteinemia Tipo II/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Metanálise como Assunto , Oligonucleotídeos/efeitos adversos , Oligonucleotídeos/farmacologia , Estados Unidos
17.
Arterioscler Thromb Vasc Biol ; 32(12): 3058-65, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23065826

RESUMO

OBJECTIVE: Although the association between circulating levels of lipoprotein(a) [Lp(a)] and risk of coronary artery disease (CAD) and stroke is well established, its role in risk of peripheral arterial disease (PAD) remains unclear. Here, we examine the association between Lp(a) levels and PAD in a large prospective cohort. To contextualize these findings, we also examined the association between Lp(a) levels and risk of stroke and CAD and studied the role of low-density lipoprotein as an effect modifier of Lp(a)-associated cardiovascular risk. METHODS AND RESULTS: Lp(a) levels were measured in apparently healthy participants in the European Prospective Investigation of Cancer (EPIC)-Norfolk cohort. Cox regression was used to quantify the association between Lp(a) levels and risk of PAD, stroke, and CAD outcomes. During 212 981 person-years at risk, a total of 2365 CAD, 284 ischemic stroke, and 596 PAD events occurred in 18 720 participants. Lp(a) was associated with PAD and CAD outcomes but not with ischemic stroke (hazard ratio per 2.7-fold increase in Lp(a) of 1.37, 95% CI 1.25-1.50, 1.13, 95% CI 1.04-1.22 and 0.91, 95% CI 0.79-1.03, respectively). Low-density lipoprotein cholesterol levels did not modify these associations. CONCLUSIONS: Lp(a) levels were associated with future PAD and CAD events. The association between Lp(a) and cardiovascular disease was not modified by low-density lipoprotein cholesterol levels.


Assuntos
Transtornos Cerebrovasculares/epidemiologia , Doença da Artéria Coronariana/epidemiologia , Lipoproteína(a)/sangue , Doença Arterial Periférica/epidemiologia , Acidente Vascular Cerebral/epidemiologia , Adulto , Idoso , Biomarcadores/sangue , Transtornos Cerebrovasculares/sangue , Estudos de Coortes , Doença da Artéria Coronariana/sangue , Europa (Continente) , Feminino , Humanos , Lipoproteínas LDL/sangue , Masculino , Pessoa de Meia-Idade , Doença Arterial Periférica/sangue , Estudos Prospectivos , Análise de Regressão , Fatores de Risco , Acidente Vascular Cerebral/sangue , Reino Unido
18.
Hum Mutat ; 33(2): 448-55, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22095935

RESUMO

Autosomal Dominant Hypercholesterolemia (ADH) is caused by LDLR and APOB mutations. However, genetically diagnosed ADH patients do not always exhibit the expected hypercholesterolemic phenotype. Of 4,669 genetically diagnosed ADH patients, identified through the national identification screening program for ADH, 75 patients (1.6%) had LDL-cholesterol (LDL-C) levels below the 50th percentile for age and gender prior to lipid-lowering therapy. The genes encoding APOB, PCSK9, and ANGPTL3 were sequenced in these subjects to address whether monogenic dominant loss-of-function mutations underlie this paradoxical phenotype. APOB mutations, resulting in truncated APOB, were found in five (6.7%) probands, reducing LDL-C by 56%. Rare variants in PCSK9, and ANGPTL3 completely correcting the hypercholesterolemic phenotype were not found. The common variants p.N902N, c.3842+82T>A, p.D2312D, and p.E4181K in APOB, and c.1863+94A>G in PCSK9 were significantly more prevalent in our cohort compared to the general European population. Interestingly, 40% of our probands carried at least one minor allele for all four common APOB variants compared to 1.5% in the general European population. While we found a low prevalence of rare variants in our cohort, our data suggest that regions in proximity of the analyzed loci, and linked to specific common haplotypes, might harbor additional variants that correct an ADH phenotype.


Assuntos
Angiopoietinas/genética , Apolipoproteínas B/genética , LDL-Colesterol/sangue , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/genética , Mutação , Pró-Proteína Convertases/genética , Serina Endopeptidases/genética , Adulto , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Feminino , Heterozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Pró-Proteína Convertase 9 , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA