Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Astrid Pañeda is active.

Publication


Featured researches published by Astrid Pañeda.


Hepatology | 2011

Molecular therapy for obesity and diabetes based on a long-term increase in hepatic fatty-acid oxidation †‡

Josep M. Orellana-Gavaldà; Laura Herrero; Maria Ida Malandrino; Astrid Pañeda; Maria Sol Rodríguez-Peña; Harald Petry; Guillermina Asins; Sander J. H. van Deventer; Fausto G. Hegardt; Dolors Serra

Obesity‐induced insulin resistance is associated with both ectopic lipid deposition and chronic, low‐grade adipose tissue inflammation. Despite their excess fat, obese individuals show lower fatty‐acid oxidation (FAO) rates. This has raised the question of whether burning off the excess fat could improve the obese metabolic phenotype. Here we used human‐safe nonimmunoreactive adeno‐associated viruses (AAV) to mediate long‐term hepatic gene transfer of carnitine palmitoyltransferase 1A (CPT1A), the key enzyme in fatty‐acid β‐oxidation, or its permanently active mutant form CPT1AM, to high‐fat diet‐treated and genetically obese mice. High‐fat diet CPT1A‐ and, to a greater extent, CPT1AM‐expressing mice showed an enhanced hepatic FAO which resulted in increased production of CO2, adenosine triphosphate, and ketone bodies. Notably, the increase in hepatic FAO not only reduced liver triacylglyceride content, inflammation, and reactive oxygen species levels but also systemically affected a decrease in epididymal adipose tissue weight and inflammation and improved insulin signaling in liver, adipose tissue, and muscle. Obesity‐induced weight gain, increase in fasting blood glucose and insulin levels, and augmented expression of gluconeogenic genes were restored to normal only 3 months after AAV treatment. Thus, CPT1A‐ and, to a greater extent, CPT1AM‐expressing mice were protected against obesity‐induced weight gain, hepatic steatosis, diabetes, and obesity‐induced insulin resistance. In addition, genetically obese db/db mice that expressed CPT1AM showed reduced glucose and insulin levels and liver steatosis. Conclusion: A chronic increase in liver FAO improves the obese metabolic phenotype, which indicates that AAV‐mediated CPT1A expression could be a potential molecular therapy for obesity and diabetes. (HEPATOLOGY 2011)


Human Gene Therapy | 2009

Effect of Adeno-Associated Virus Serotype and Genomic Structure on Liver Transduction and Biodistribution in Mice of Both Genders

Astrid Pañeda; Lucia Vanrell; Itsaso Mauleón; Julien Crettaz; Pedro Berraondo; Eric J. Timmermans; Stuart G. Beattie; Jaap Twisk; Sander J. H. van Deventer; Jesús Prieto; Antonio Fontanellas; Maria Sol Rodríguez-Peña; Gloria González-Aseguinolaza

Recombinant adeno-associated viral (AAV) vectors have unique properties, which make them suitable vectors for gene transfer. Here we assess the liver transduction efficiency and biodistribution of AAV-pseudotyped capsids (serotypes) 1, 5, 6, and 8, combined with single-stranded and double-stranded genomic AAV2 structures carrying the luciferase reporter gene after systemic administration. The analysis was performed in vivo and ex vivo, in male and female mice. Gender-related differences in AAV-mediated transduction and biodistribution were shown for the four serotypes. Our data confirm the superiority of AAV8 over the rest of the serotypes, as well as a significant advantage of double-stranded genomes in terms of liver transduction efficiency, particularly in females. Regarding biodistribution, AAV5 displayed a narrower tropism than the other serotypes tested, transducing, almost exclusively, the liver. Interestingly, AAV1 and AAV8, in particular those having single-stranded genomes, showed high transduction efficiency of female gonads. However, no inadvertent germ line transmission of AAV genomes was observed after breeding single-stranded AAV8-injected female mice with untreated males. In conclusion, double-stranded AAV8 vectors led to the highest levels of liver transduction in mice, as demonstrated by luciferase expression. Nevertheless, the transduction of other organs with AAV8 vectors could favor the use of less efficient serotypes, such as AAV5, which display a narrow tropism.


Human Gene Therapy | 2013

Safety and Liver Transduction Efficacy of rAAV5-cohPBGD in Nonhuman Primates: A Potential Therapy for Acute Intermittent Porphyria

Astrid Pañeda; Esperanza López-Franco; Christine Kaeppel; Carmen Unzu; Ana Gloria Gil-Royo; Delia D'Avola; Stuart G. Beattie; Cristina Olagüe; Roberto Ferrero; Ana Sampedro; Itsaso Mauleón; Stephan Hermening; Florence Salmon; Alberto Benito; Juan J. Gavira; María Eugenia Cornet; María del Mar Municio; Christof von Kalle; Harald Petry; Jesús Prieto; Manfred Schmidt; Antonio Fontanellas; Gloria González-Aseguinolaza

Acute intermittent porphyria (AIP) results from haplo-insufficient activity of porphobilinogen deaminase (PBGD) and is characterized clinically by life-threatening, acute neurovisceral attacks. To date, liver transplantation is the only curative option for AIP. The aim of the present preclinical nonhuman primate study was to determine the safety and transduction efficacy of an adeno-associated viral vector encoding PBGD (recombinant AAV serotype 5-codon-optimized human porphobilinogen deaminase, rAAV5-cohPBGD) administered intravenously as part of a safety program to start a clinical study in patients with AIP. Macaques injected with either 1 × 10(13) or 5 × 10(13) vector genomes/kg of clinical-grade rAAV5-cohPBGD were monitored by standardized clinical parameters, and vector shedding was analyzed. Liver transduction efficacy, biodistribution, vector integration, and histopathology at day 30 postvector administration were determined. There was no evidence of acute toxicity, and no adverse effects were observed. The vector achieved efficient and homogenous hepatocellular transduction, reaching transgenic PBGD expression levels equivalent to 50% of the naturally expressed PBGD mRNA. No cellular immune response was detected against the human PBGD or AAV capsid proteins. Integration site analysis in transduced liver cells revealed an almost random integration pattern supporting the good safety profile of rAAV5-cohPBGD. Together, data obtained in nonhuman primates indicate that rAAV5-cohPBGD represents a safe therapy to correct the metabolic defect present in AIP patients.


Journal of Virology | 2009

Treatment of Chronic Viral Hepatitis in Woodchucks by Prolonged Intrahepatic Expression of Interleukin-12

Julien Crettaz; Itziar Otano; Laura Ochoa; Alberto Benito; Astrid Pañeda; Igor Aurrekoetxea; Pedro Berraondo; Juan R. Rodriguez-Madoz; Aurora Astudillo; Florian Kreppel; Stefan Kochanek; J.J. Ruiz; Stephan Menne; Jesús Prieto; Gloria González-Aseguinolaza

ABSTRACT Chronic hepatitis B is a major cause of liver-related death worldwide. Interleukin-12 (IL-12) induction accompanies viral clearance in chronic hepatitis B virus infection. Here, we tested the therapeutic potential of IL-12 gene therapy in woodchucks chronically infected with woodchuck hepatitis virus (WHV), an infection that closely resembles chronic hepatitis B. The woodchucks were treated by intrahepatic injection of a helper-dependent adenoviral vector encoding IL-12 under the control of a liver-specific RU486-responsive promoter. All woodchucks with viral loads below 1010 viral genomes (vg)/ml showed a marked and sustained reduction of viremia that was accompanied by a reduction in hepatic WHV DNA, a loss of e antigen and surface antigen, and improved liver histology. In contrast, none of the woodchucks with higher viremia levels responded to therapy. The antiviral effect was associated with the induction of T-cell immunity against viral antigens and a reduction of hepatic expression of Foxp3 in the responsive animals. Studies were performed in vitro to elucidate the resistance to therapy in highly viremic woodchucks. These studies showed that lymphocytes from healthy woodchucks or from animals with low viremia levels produced gamma interferon (IFN-γ) upon IL-12 stimulation, while lymphocytes from woodchucks with high viremia failed to upregulate IFN-γ in response to IL-12. In conclusion, IL-12-based gene therapy is an efficient approach to treat chronic hepadnavirus infection in woodchucks with viral loads below 1010 vg/ml. Interestingly, this therapy is able to break immunological tolerance to viral antigens in chronic WHV carriers.


Journal of Hepatology | 2016

Phase I open label liver-directed gene therapy clinical trial for acute intermittent porphyria

D. D’Avola; Esperanza López-Franco; Bruno Sangro; Astrid Pañeda; Nadina Grossios; Irene Gil-Farina; Alberto Benito; Jaap Twisk; María Paz; J.J. Ruiz; Manfred Schmidt; Harald Petry; Pauline Harper; Rafael Enríquez de Salamanca; Antonio Fontanellas; Jesús Prieto; Gloria González-Aseguinolaza

BACKGROUND & AIMS Acute intermittent porphyria (AIP) results from porphobilinogen deaminase (PBGD) haploinsufficiency, which leads to hepatic over-production of the neurotoxic heme precursors porphobilinogen (PBG) and delta-aminolevulinic acid (ALA) and the occurrence of neurovisceral attacks. Severe AIP is a devastating disease that can only be corrected by liver transplantation. Gene therapy represents a promising curative option. The objective of this study was to investigate the safety of a recombinant adeno-associated vector expressing PBGD (rAAV2/5-PBGD) administered for the first time in humans for the treatment of AIP. METHODS In this phase I, open label, dose-escalation, multicenter clinical trial, four cohorts of 2 patients each received a single intravenous injection of the vector ranging from 5×10(11) to 1.8×10(13) genome copies/kg. Adverse events and changes in urinary PBG and ALA and in the clinical course of the disease were periodically evaluated prior and after treatment. Viral shedding, immune response against the vector and vector persistence in the liver were investigated. RESULTS Treatment was safe in all cases. All patients developed anti-AAV5 neutralizing antibodies but no cellular responses against AAV5 or PBGD were observed. There was a trend towards a reduction of hospitalizations and heme treatments, although ALA and PBG levels remained unchanged. Vector genomes and transgene expression could be detected in the liver one year after therapy. CONCLUSIONS rAAV2/5-PBGD administration is safe but AIP metabolic correction was not achieved at the doses tested in this trial. Notwithstanding, the treatment had a positive impact in clinical outcomes in most patients. LAY SUMMARY Studies in an acute intermittent porphyria (AIP) animal model have shown that gene delivery of PBGD to hepatocytes using an adeno-associated virus vector (rAAV2/5-PBG) prevent mice from suffering porphyria acute attacks. In this phase I, open label, dose-escalation, multicenter clinical trial we show that the administration of rAAV2/5-PBGD to patients with severe AIP is safe but metabolic correction was not achieved at the doses tested; the treatment, however, had a positive but heterogeneous impact on clinical outcomes among treated patients and 2 out of 8 patients have stopped hematin treatment. CLINICAL TRIAL NUMBER The observational phase was registered at Clinicaltrial.gov as NCT 02076763. The interventional phase study was registered at EudraCT as n° 2011-005590-23 and at Clinicaltrial.gov as NCT02082860.


Human Gene Therapy | 2011

Adeno-Associated Virus Liver Transduction Efficiency Measured by in Vivo [18F]FHBG Positron Emission Tomography Imaging in Rodents and Nonhuman Primates

Astrid Pañeda; María Collantes; Stuart G. Beattie; Itzia Otano; Jolanda Snapper; Eric Jacobus Hubertus Timmermans; Laura Guembe; Harald Petry; José L. Lanciego; Alberto Benito; Jesús Prieto; Maria Sol Rodríguez-Peña; Iván Peñuelas; Gloria González-Aseguinolaza

Recombinant adeno-associated virus 5 (rAAV5) represents a candidate vector with unique advantages for the treatment of hepatic disorders because of its narrow hepatic tropism. Noninvasive in vivo imaging of transgene expression provides an important tool with which to quantify the transduction efficiency, and duration and location, of transgene expression. In this study, we used positron emission tomography (PET) and positron emission tomography-computed tomography (PET-CT) imaging to monitor liver transduction efficacy in rodents and nonhuman primates that received rAAV5 vector encoding herpes simplex virus thymidine kinase (HSV-TK). HSV-TK expression in liver was also measured by immunohistochemistry. Notable differences in liver transduction efficiency were found, dependent on the animal species and sex. Male rodents were better transduced than females, as previously described. Moreover, male nonhuman primates also displayed increased hepatic expression of the rAAV5-delivered transgene, indicating that differences in rAAV-mediated liver transduction can be anticipated in humans. Our results demonstrate the high sensitivity and reproducibility of PET, using HSV-TK and [(18)F]FHBG, to detect gene expression after rAAV vector administration into living animals, confirming the utility of this technology in the quantification of transgene expression, even at low expression levels. However, we also describe how an immune response against HSV-TK hampered analysis of long-term expression in nonhuman primates.


Expert Opinion on Biological Therapy | 2006

Gene therapy for viral hepatitis

Gloria González-Aseguinolaza; Julien Crettaz; Laura Ochoa; Itziar Otano; Rafael Aldabe; Astrid Pañeda

Hepatitis B and C infections are two of the most prevalent viral diseases in the world. Existing therapies against chronic viral hepatitis are far from satisfactory due to low response rates, undesirable side effects and selection of resistant viral strains. Therefore, new therapeutic approaches are urgently needed. This review, after briefly summarising the in vitro and in vivo systems for the study of both diseases and the genetic vehicles commonly used for liver gene transfer, examines the existing status of gene therapy-based antiviral strategies that have been employed to prevent, eliminate or reduce viral infection. In particular, the authors focus on the results obtained in clinical trials and experimental clinically relevant animal models.


Journal of Hepatology | 2006

37 Gene therapy of chronic hepatitis B infection using a high-capacity adenovirus expressing interleukin 12 under the control of a liver specific mifepristone-inducible promoter

Julien Crettaz; Laura Ochoa; Astrid Pañeda; C. Olagüe; A. Vales; Stefan Kochanek; Jesús Prieto; Gloria González-Aseguinolaza

Three hundred fifty millions people are estimated to be infected by the hepatitis B virus (HBV). Eighty-seven millions of them would develop HBV-related liver cancer or cirrhosis. In this study we developed a high-capacity adenovirus (HC-Ad), characterized by its low toxicity and long-term expression, encoding for the murine interleukin 12 (mIL12) directed by a liver-specific promoter and inducible by the administration of mifepristone (HC-RU-mIL12). We assayed its antiviral effect using two animal models of chronic HBV infection: transgenic mice for the HBV genome and woodchucks chronically infected by the woodchuck hepatitis virus (WHV).


Human Gene Therapy | 2006

Intrahepatic Injection of Recombinant Adeno-Associated Virus Serotype 2 Overcomes Gender-Related Differences in Liver Transduction

Pedro Berraondo; Julien Crettaz; Laura Ochoa; Astrid Pañeda; Jesús Prieto; Iñaki F. Trocóniz; Gloria González-Aseguinolaza


Toxicology Letters | 2013

Non-clinical safety evaluation of AAV5-PBGD in mice and cynomolgus macaques

Gloria González-Aseguinolaza; Astrid Pañeda; Esperanza López-Franco; Cristina Olagüe; Antonio Fontanellas; Carmen Unzu; Ana Sampedro; María Eugenia Cornet; María del Mar Municio; Paul Heal; Harald Petry; Marc Sonnemans; Lisa Spronck; Florence Salmon

Collaboration


Dive into the Astrid Pañeda's collaboration.

Top Co-Authors

Avatar

Gloria González-Aseguinolaza

Chartered Institute of Management Accountants

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stuart G. Beattie

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge