110 results on '"Sondhi, D."'
Search Results
2. AAV2-mediated CLN2 gene transfer to rodent and non-human primate brain results in long-term TPP-I expression compatible with therapy for LINCL
- Author
-
Sondhi, D, Peterson, D A, Giannaris, E L, Sanders, C T, Mendez, B S, De, B, Rostkowski, A B, Blanchard, B, Bjugstad, K, Sladek, Jr, J R, Redmond, Jr, D E, Leopold, P L, Kaminsky, S M, Hackett, N R, and Crystal, R G
- Published
- 2005
- Full Text
- View/download PDF
3. The Natural History of Late Infantile CLN2 Disease: Striking Homogeneity of Clinical Progression in Two Independently Obtained Large Clinical Cohorts: PS2 - 4.
- Author
-
Schulz, A, Nickel, M, Downs, M, Mezey, J, Landy, H, Sondhi, D, Jacoby, D, Wittes, J, Crystal, R G, and Kohlschuetter, A
- Published
- 2013
4. DOES CATHETER SIZE MATTER? INTRAPLEURAL FIBRINOLYTIC THERAPY FOR MANAGEMENT OF EMPYEMA AND COMPLICATED PLEURAL EFFUSIONS
- Author
-
Chhabria, M., primary and Sondhi, D., additional
- Published
- 2020
- Full Text
- View/download PDF
5. Radioiodinated Capsids Facilitate In Vivo Non-Invasive Tracking of Adeno-Associated Gene Transfer Vectors
- Author
-
Kothari, P., primary, De, B. P., additional, He, B., additional, Chen, A., additional, Chiuchiolo, M. J., additional, Kim, D., additional, Nikolopoulou, A., additional, Amor-Coarasa, A., additional, Dyke, J. P., additional, Voss, H. U., additional, Kaminsky, S. M., additional, Foley, C. P., additional, Vallabhajosula, S., additional, Hu, B., additional, DiMagno, S. G., additional, Sondhi, D., additional, Crystal, R. G., additional, Babich, J. W., additional, and Ballon, D., additional
- Published
- 2017
- Full Text
- View/download PDF
6. Brain Region–Specific Degeneration with Disease Progression in Late Infantile Neuronal Ceroid Lipofuscinosis (CLN2 Disease)
- Author
-
Dyke, J.P., primary, Sondhi, D., additional, Voss, H.U., additional, Yohay, K., additional, Hollmann, C., additional, Mancenido, D., additional, Kaminsky, S.M., additional, Heier, L.A., additional, Rudser, K.D., additional, Kosofsky, B., additional, Casey, B.J., additional, Crystal, R.G., additional, and Ballon, D., additional
- Published
- 2016
- Full Text
- View/download PDF
7. Assessment of Disease Severity in Late Infantile Neuronal Ceroid Lipofuscinosis Using Multiparametric Magnetic Resonance Imaging
- Author
-
Dyke, J. P., Sondhi, D., Voss, H. U., Shungu, D. C., Mao, X., Yohay, K., Worgall, S., Hackett, N. R., Hollmann, C., Yeotsas, M. E., Jeong, A.L., Van de Graaf, B., Cao, I., Kaminsky, S. M., Heier, L. A., Rudser, K.D., Souweidane, M. M., Kaplitt, M.G., Kosofsky, B., Crystal, R. G., and Ballon, D.
- Subjects
Male ,Databases, Factual ,Tripeptidyl-Peptidase 1 ,Age Factors ,Brain ,Aminopeptidases ,Magnetic Resonance Imaging ,Severity of Illness Index ,Article ,Neuronal Ceroid-Lipofuscinoses ,Child, Preschool ,Disease Progression ,Humans ,Female ,Serine Proteases ,Artifacts ,Child ,Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,Biomarkers - Abstract
LINCL is a uniformly fatal lysosomal storage disease resulting from mutations in the CLN2 gene that encodes for tripeptidyl peptidase 1, a lysosomal enzyme necessary for the degradation of products of cellular metabolism. With the goal of developing quantitative noninvasive imaging biomarkers sensitive to disease progression, we evaluated a 5-component MR imaging metric and tested its correlation with a clinically derived disease-severity score.MR imaging parameters were measured across the brain, including quantitative measures of the ADC, FA, nuclear spin-spin relaxation times (T2), volume percentage of CSF (%CSF), and NAA/Cr ratios. Thirty MR imaging datasets were prospectively acquired from 23 subjects with LINCL (2.5-8.4 years of age; 8 male/15 female). Whole-brain histograms were created, and the mode and mean values of the histograms were used to characterize disease severity.Correlation of single MR imaging parameters against the clinical disease-severity scale yielded linear regressions with R2 ranging from 0.25 to 0.70. Combinations of the 5 biomarkers were evaluated by using PCA. The best combination included ADC, %CSF, and NAA/Cr (R2=0.76, P.001).The multiparametric disease-severity score obtained from the combination of ADC, %CSF, and NAA/Cr whole-brain MR imaging techniques provided a robust measure of disease severity, which may be useful in clinical therapeutic trials of LINCL in which an objective assessment of therapeutic response is desired.
- Published
- 2012
8. Brain gene therapy for metachromatic leukodystrophy: towards clinical trial
- Author
-
Piguet, F., Sondhi, D., Colle, Marie-Anne, Raoul, Solène, Roujeau, T., Piraud, M., Ahouansou, O., Bouquet, C., Vanier, M.T., Moullier, P., Cherel, Yan, Hackett, N.R, Zerah, M., Aubourg, P., Crystal, R.G., Cartier, N., Sevin, C., Genetique et Biotherapies des Maladies Degeneratives et Proliferatives du Systeme Nerveux (Inserm U745), Institut des sciences du Médicament -Toxicologie - Chimie - Environnement (IFR71), Institut National de la Santé et de la Recherche Médicale (INSERM)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Ecole Nationale Supérieure de Chimie de Paris - Chimie ParisTech-PSL (ENSCP), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche pour le Développement (IRD)-Université Paris Descartes - Paris 5 (UPD5)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Cornell University [New York], Développement et Pathologie du Tissu Musculaire (DPTM), Institut National de la Recherche Agronomique (INRA)-Ecole Nationale Vétérinaire de Nantes, Centre hospitalier universitaire de Nantes (CHU Nantes), CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Centre Hospitalier Universitaire de Lyon, Métabolomique et maladies métaboliques, Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM), Vecteurs viraux et transfert de gènes in vivo, Université de Nantes (UN)-IFR26-Institut National de la Santé et de la Recherche Médicale (INSERM), and ProdInra, Migration
- Subjects
[SDV] Life Sciences [q-bio] ,brain gene therapy ,[SDV]Life Sciences [q-bio] ,ComputingMilieux_MISCELLANEOUS ,metachromatic leukodystrophy - Abstract
International audience
- Published
- 2010
9. Genetic modification of neurons to express bevacizumab for local anti-angiogenesis treatment of glioblastoma
- Author
-
Hicks, M J, primary, Funato, K, additional, Wang, L, additional, Aronowitz, E, additional, Dyke, J P, additional, Ballon, D J, additional, Havlicek, D F, additional, Frenk, E Z, additional, De, B P, additional, Chiuchiolo, M J, additional, Sondhi, D, additional, Hackett, N R, additional, Kaminsky, S M, additional, Tabar, V, additional, and Crystal, R G, additional
- Published
- 2014
- Full Text
- View/download PDF
10. O62 – 1673 The natural history of late infantile CLN2 disease: striking homogeneity of clinical progression in two independently obtained large clinical cohorts
- Author
-
Schulz, A, primary, Nickel, M, additional, Downs, M, additional, Mezey, J, additional, Landy, H, additional, Sondhi, D, additional, Jacoby, D, additional, Wittes, J, additional, Crystal, R, additional, and Kohlschütter, A, additional
- Published
- 2013
- Full Text
- View/download PDF
11. O71 – 2078 Brain gene therapy for metachromatic leukodystrophy
- Author
-
Sevin, C, primary, Roujeau, T, additional, Piguet, F, additional, Sondhi, D, additional, Colle, MA, additional, Raoul, S, additional, Deschamps, JY, additional, and Bouquet, C, additional
- Published
- 2013
- Full Text
- View/download PDF
12. Assessment of Disease Severity in Late Infantile Neuronal Ceroid Lipofuscinosis Using Multiparametric MR Imaging
- Author
-
Dyke, J.P., primary, Sondhi, D., additional, Voss, H.U., additional, Shungu, D.C., additional, Mao, X., additional, Yohay, K., additional, Worgall, S., additional, Hackett, N.R., additional, Hollmann, C., additional, Yeotsas, M.E., additional, Jeong, A.L., additional, Van de Graaf, B., additional, Cao, I., additional, Kaminsky, S.M., additional, Heier, L.A., additional, Rudser, K.D., additional, Souweidane, M.M., additional, Kaplitt, M.G., additional, Kosofsky, B., additional, Crystal, R.G., additional, and Ballon, D., additional
- Published
- 2012
- Full Text
- View/download PDF
13. P05.4 Brain gene therapy for Metachromatic Leukodystrophy: from bench to bedside
- Author
-
Sevin, C., primary, Roujeau, T., additional, Piguet, F., additional, Sondhi, D., additional, Colle, M.-A., additional, Raoul, S., additional, Deschamps, J.-Y., additional, Bouquet, C., additional, Vanier, M., additional, Moullier, P., additional, Cherel, Y., additional, Hackett, N.R., additional, Zerah, M., additional, Crystal, R.G., additional, Cartier, N., additional, and Aubourg, P., additional
- Published
- 2011
- Full Text
- View/download PDF
14. Neurological deterioration in late infantile neuronal ceroid lipofuscinosis
- Author
-
Worgall, S., primary, Kekatpure, M. V., additional, Heier, L., additional, Ballon, D., additional, Dyke, J. P., additional, Shungu, D., additional, Mao, X., additional, Kosofsky, B., additional, Kaplitt, M. G., additional, Souweidane, M. M., additional, Sondhi, D., additional, Hackett, N. R., additional, Hollmann, C., additional, and Crystal, R. G., additional
- Published
- 2007
- Full Text
- View/download PDF
15. Assessing Disease Severity in Late Infantile Neuronal Ceroid Lipofuscinosis Using Quantitative MR Diffusion-Weighted Imaging
- Author
-
Dyke, J.P., primary, Voss, H.U., additional, Sondhi, D., additional, Hackett, N.R., additional, Worgall, S., additional, Heier, L.A., additional, Kosofsky, B.E., additional, Ulug, A.M., additional, Shungu, D.C., additional, Mao, X., additional, Crystal, R.G., additional, and Ballon, D., additional
- Published
- 2007
- Full Text
- View/download PDF
16. RETROPERITONEAL HEMORRHAGE(RPH)- REVIEW OF NATURAL HISTORY, MORBIDITY AND OUTCOME
- Author
-
MURALI, GANESAN, primary, Devine, S, additional, Walkenstein, M, additional, Goldberg, S, additional, Lippmann, M, additional, Sondhi, D, additional, Supaongprapa, W, additional, and Hamed, R, additional
- Published
- 2002
- Full Text
- View/download PDF
17. Design, fabrication and operation of a biomass fermentation facility. First quarterly report, October 1-December 31, 1978
- Author
-
O'Neil, D.J., primary, Colcord, A.R., additional, Bery, M.K., additional, Day, S.W., additional, Roberts, R.S., additional, El-Barbary, I.A., additional, Havlicek, S.C., additional, Anders, M.E., additional, and Sondhi, D., additional
- Published
- 1978
- Full Text
- View/download PDF
18. Airway compromise due to angiotensin-converting enzyme inhibitor-induced angioedema: clinical experience at a large community teaching hospital.
- Author
-
Sondhi D, Lippmann M, and Murali G
- Abstract
STUDY OBJECTIVE: To evaluate the incidence of airway compromise, clinical presentation and morbidity of angiotensin-converting enzyme inhibitor (ACEI)-related angioedema (AE). METHOD: A retrospective chart review was conducted of all patients admitted to our hospital between 1996 and 2001 with the diagnosis of AE. RESULTS: A total of 70 charts on which a diagnosis of AE had been entered were reviewed. Of those, 45 patients (64%) had AE that was thought to be related to ACEI therapy. Of those 45 patients, 29 were women. The mean age was 62 years, and 41 (91%) were African-American. The duration of ACEI therapy before presentation varied from 1 day to 5 years postingestion. Twenty-one of 32 patients presented within 2 months of the initiation of therapy. The mean duration between the onset of symptoms and presentation to the hospital was 9 h. Lip and tongue swelling was seen in all patients. Pulmonary manifestations were noted in 17 of 45 patients (38%) [dyspnea 17 of 17 patients; stridor/respiratory failure, 5 of 17 patients; cough, 2 of 17 patients]. Dysphagia was noted in 9 of 45 patients, drooling of saliva in 8 of 45 patients, and pruritus in 6 of 45 patients. Ten of 45 patients had a history of AE. In five of those patients (50%), ACEI use was a presumed cause of the AE for the current hospital admission. Eighteen of 45 (40%) patients required ICU admission. The mean ICU length of stay was 2.2 days. Five of 45 patients required endotracheal intubation. The mean time spent receiving ventilation was 2.2 days. Discontinuation of the initiating agent and supportive care were the keys to therapy. All of our patients responded to supportive management, and there was no mortality. CONCLUSION: Sixty-four percent of patients in this series had AE due to receiving an ACEI. The majority of the patients were African-American women. Most patients presented within 2 months of starting to receive the drug, although longer durations of therapy were not uncommon. Lip and tongue swelling was the most common airway manifestation. Based on our observations, the discontinuation of ACEI therapy and supportive management are the recommended approaches to therapy to prevent an untoward outcome. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
19. Chemical Approaches to the Study of Protein Tyrosine Kinases and Their Implications for Mechanism and Inhibitor Design - routes of metabolism and selective toxicity
- Author
-
Cole, P.A., Sondhi, D., and Kim, K.
- Published
- 1999
- Full Text
- View/download PDF
20. Expression and processing of mature human frataxin after gene therapy in mice.
- Author
-
Rojsajjakul T, Selvan N, De B, Rosenberg JB, Kaminsky SM, Sondhi D, Janki P, Crystal RG, Mesaros C, Khanna R, and Blair IA
- Subjects
- Humans, Animals, Mice, Heart, Protein Processing, Post-Translational, Liver metabolism, Genetic Therapy, Iron-Binding Proteins metabolism, Frataxin, Friedreich Ataxia therapy, Friedreich Ataxia drug therapy
- Abstract
Friedreich's ataxia is a degenerative and progressive multisystem disorder caused by mutations in the highly conserved frataxin (FXN) gene that results in FXN protein deficiency and mitochondrial dysfunction. While gene therapy approaches are promising, consistent induction of therapeutic FXN protein expression that is sub-toxic has proven challenging, and numerous therapeutic approaches are being tested in animal models. FXN (hFXN in humans, mFXN in mice) is proteolytically modified in mitochondria to produce mature FXN. However, unlike endogenous hFXN, endogenous mFXN is further processed into N-terminally truncated, extra-mitochondrial mFXN forms of unknown function. This study assessed mature exogenous hFXN expression levels in the heart and liver of C57Bl/6 mice 7-10 months after intravenous administration of a recombinant adeno-associated virus encoding hFXN (AAVrh.10hFXN) and examined the potential for hFXN truncation in mice. AAVrh.10hFXN induced dose-dependent expression of hFXN in the heart and liver. Interestingly, hFXN was processed into truncated forms, but found at lower levels than mature hFXN. However, the truncations were at different positions than mFXN. AAVrh.10hFXN induced mature hFXN expression in mouse heart and liver at levels that approximated endogenous mFXN levels. These results suggest that AAVrh.10hFXN can likely induce expression of therapeutic levels of mature hFXN in mice., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
21. Expression and processing of mature human frataxin after gene therapy in mice.
- Author
-
Rojsajjakul T, Selvan N, De B, Rosenberg JB, Kaminsky SM, Sondhi D, Janki P, Crystal RG, Mesaros C, Khanna R, and Blair IA
- Abstract
Friedreich's ataxia is a degenerative and progressive multisystem disorder caused by mutations in the highly conserved frataxin (FXN) gene that results in FXN protein deficiency and mitochondrial dysfunction. While gene therapy approaches are promising, consistent induction of therapeutic FXN protein expression that is sub-toxic has proven challenging, and numerous therapeutic approaches are being tested in animal models. FXN (hFXN in humans, mFXN in mice) is proteolytically modified in mitochondria to produce mature FXN. However, unlike endogenous hFXN, endogenous mFXN is further processed into N-terminally truncated, extra-mitochondrial mFXN forms of unknown function. This study assessed mature exogenous hFXN expression levels in the heart and liver of C57Bl/6 mice 7-10 months after intravenous administration of a recombinant adeno-associated virus encoding hFXN (AAVrh.10hFXN) and examined the potential for hFXN truncation in mice. AAVrh.10hFXN induced dose-dependent expression of hFXN in the heart and liver. Interestingly, hFXN was processed into truncated forms, but found at lower levels than mature hFXN. However, the truncations were at different positions than mFXN. AAVrh.10hFXN induced mature hFXN expression in mouse heart and liver at levels that approximated endogenous mFXN levels. These results demonstrate that AAVrh.10hFXN may induce expression of therapeutic levels of mature hFXN in mice., Competing Interests: Competing interests N.S., P.J., and R.K. are full-time employees of LEXEO Therapeutics, Inc, S.M.K. and D.S. have equity in LEXEO Therapeutics, Inc, and R.G.C. has equity in and is a consultant for LEXEO Therapeutics, Inc. I.A.B. has paid service/sponsored research agreements with LEXEO Therapeutics, Inc and Design Therapeutics, Inc. The other authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
22. Positron Emission Tomography Quantitative Assessment of Off-Target Whole-Body Biodistribution of I-124-Labeled Adeno-Associated Virus Capsids Administered to Cerebral Spinal Fluid.
- Author
-
Rosenberg JB, Fung EK, Dyke JP, De BP, Lou H, Kelly JM, Reejhsinghani L, Ricart Arbona RJ, Sondhi D, Kaminsky SM, Cartier N, Hinderer C, Hordeaux J, Wilson JM, Ballon DJ, and Crystal RG
- Subjects
- Animals, Iodine Radioisotopes, Capsid, Tissue Distribution, Transduction, Genetic, Genetic Therapy methods, Positron-Emission Tomography, Genetic Vectors genetics, Gene Transfer Techniques, Dependovirus genetics, Nervous System Diseases
- Abstract
Based on studies in experimental animals demonstrating that administration of adeno-associated virus (AAV) vectors to the cerebrospinal fluid (CSF) is an effective route to transfer genes to the nervous system, there are increasing number of clinical trials using the CSF route to treat nervous system disorders. With the knowledge that the CSF turns over four to five times daily, and evidence in experimental animals that at least some of CSF administered AAV vectors are distributed to systemic organs, we asked: with AAV administration to the CSF, what fraction of the total dose remains in the nervous system and what fraction goes off target and is delivered systemically? To quantify the biodistribution of AAV capsids immediately after administration, we covalently labeled AAV capsids with iodine 124 (I-124), a cyclotron generated positron emitter, enabling quantitative positron emission tomography scanning of capsid distribution for up to 96 h after AAV vector administration. We assessed the biodistribution to nonhuman primates of I-124-labeled capsids from different AAV clades, including 9 (clade F), rh.10 (E), PHP.eB (F), hu68 (F), and rh91(A). The analysis demonstrated that 60-90% of AAV vectors administered to the CSF through either the intracisternal or intrathecal (lumbar) routes distributed systemically to major organs. These observations have potentially significant clinical implications regarding accuracy of AAV vector dosing to the nervous system, evoking systemic immunity at levels similar to that with systemic administration, and potential toxicity of genes designed to treat nervous system disorders being expressed in non-nervous system organs. Based on these data, individuals in clinical trials using AAV vectors administered to the CSF should be monitored for systemic as well as nervous system adverse events and CNS dosing considerations should account for a significant AAV systemic distribution.
- Published
- 2023
- Full Text
- View/download PDF
23. Assessment of Safety and Biodistribution of AAVrh.10hCLN2 Following Intracisternal Administration in Nonhuman Primates for the Treatment of CLN2 Batten Disease.
- Author
-
De BP, Rosenberg JB, Selvan N, Wilson I, Yusufzai N, Greco A, Kaminsky SM, Heier LA, Ricart Arbona RJ, Miranda IC, Monette S, Nair A, Khanna R, Crystal RG, and Sondhi D
- Subjects
- Humans, Animals, Child, Tissue Distribution, Central Nervous System, Brain diagnostic imaging, Primates, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses therapy
- Abstract
CLN2 disease is a fatal, childhood autosomal recessive disorder caused by mutations in ceroid lipofuscinosis type 2 (CLN2) gene, encoding tripeptidyl peptidase 1 (TPP-1). Loss of TPP-1 activity leads to accumulation of storage material in lysosomes and resultant neuronal cell death with neurodegeneration. Genotype/phenotype comparisons suggest that the phenotype should be ameliorated with increase of TPP-1 levels to 5-10% of normal with wide central nervous system (CNS) distribution. Our previous clinical study showed that intraparenchymal (IPC) administration of AAVrh.10hCLN2, an adeno-associated vector serotype rh.10 encoding human CLN2, slowed, but did not stop disease progression, suggesting that this may be insufficient to distribute the therapy throughout the CNS (Sondhi 2020). In this study, we assessed whether the less invasive intracisternal delivery route would be safe and provide a wider distribution of TPP-1. A study was conducted in nonhuman primates (NHPs) with intracisternal delivery to cerebrospinal fluid (CSF) of AAVrh.10hCLN2 (5 × 10
13 genome copies) or phosphate buffered saline (PBS). No abnormal behavior was noted. CNS magnetic resonance imaging and clinical chemistry data were all unremarkable. Histopathology of major organs had no abnormal finding attributable to the intervention or the vector, except that in one out of two animals treated with AAVrh.10hCLN2, dorsal root ganglia showed mild-to-moderate mononuclear cell infiltrates and neuronal degeneration. In contrast to our previous NHP study (Sondhi 2012) with IPC administration where TPP-1 activity was >2 × above controls in 30% of treated brains, in the two intracisternal treated NHPs, the TPP-1 activity was >2 × above controls in 50% and 41% of treated brains, and 52% and 84% of brain had >1,000 vector genomes/μg DNA, compared to 0% in the two PBS NHP. CSF TPP1 levels in treated animals were 43-62% of normal human levels. Collectively, these data indicate that AAVrh.10hCLN2 delivered by intracisternal route is safe and widely distributes TPP-1 in brain and CSF at levels that are potentially therapeutic. Clinical Trial Registration: NCT02893826, NCT04669535, NCT04273269, NCT03580083, NCT04408625, NCT04127578, and NCT04792944.- Published
- 2023
- Full Text
- View/download PDF
24. Assessment of Residual Full-Length SV40 Large T Antigen in Clinical-Grade Adeno-Associated Virus Vectors Produced in 293T Cells.
- Author
-
De BP, Cram S, Lee H, Rosenberg JB, Sondhi D, Crystal RG, and Kaminsky SM
- Subjects
- Mice, Animals, Humans, HEK293 Cells, Dependovirus genetics, Dependovirus metabolism, Antigens, Viral, Tumor genetics, Antigens, Viral, Tumor metabolism, Genetic Vectors genetics, DNA, Herpesvirus 1, Human genetics, Herpesvirus 1, Human metabolism
- Abstract
Efficient production of adeno-associated virus (AAV) vectors is a significant challenge. Human embryonic kidney HEK293T cells are widely used in good manufacturing practice facilities, producing higher yield of AAV vectors for clinical applications than HEK293 through the addition of a constitutive expression of SV40 large T antigen (SV40T), which stimulates Rep expression. However, the theoretical potential for tumorigenic consequences of a clinical AAV product containing residual DNA encoding SV40T, which may inhibit p53 growth suppressive functions is a safety concern. Although the risk is theoretical, to assure a low risk/high confidence of safety for clinical drug development, we have established a sensitive assay for assessment of functional full-length transcription competent SV40T DNA in HEK293T cell-produced AAV vectors. Using HEK293T generated 8, 9, and rh.10 serotype AAV vectors, the presence of SV40T in purified vector was assessed in vitro using quantitative polymerase chain reaction (qPCR) targeting a 129 bp amplicon combined with nested PCR targeting full-length SV40T DNA. Although low levels of the smaller amplicon were present in each AAV serotype, the full-length SV40T was undetectable. No transcription competent full-length SV40T DNA was observed by reverse transcription-quantitative polymerase chain reaction using an in vivo amplification of signal in mouse liver administered (2-10 × 10
10 gc) 129 bp amplicon-positive AAV vectors. As a control for gene transfer, high levels of expressed transgene mRNAs were observed from each serotype AAV vector, yet, SV40T mRNA was undetectable. In vivo assessment of these three liver-tropic AAV serotypes, each with amplicon-positive qPCR SV40T DNA, demonstrated high transgene mRNA expression but no SV40T mRNA, that is, detection of small segments of SV40T DNA in 293T cell produced AAV inappropriately leads to the conclusion of residuals with the potential to express SV40T. This sensitive assay can be used to assess the level, if any, of SV40T antigen contaminating AAV vectors generated by HEK293T cells. ClinicalTrials.gov identifier: NCT03634007; NCT05302271; NCT01414985; NCT01161576.- Published
- 2023
- Full Text
- View/download PDF
25. Identification of Safe and Effective Intravenous Dose of AAVrh.10hFXN to Treat the Cardiac Manifestations of Friedreich's Ataxia.
- Author
-
Munoz-Zuluaga C, Gertz M, Yost-Bido M, Greco A, Gorman N, Chen A, Kooner V, Rosenberg JB, De BP, Kaminsky SM, Borczuk A, Ricart Arbona RJ, Martin HR, Monette S, Khanna R, Barth JA, Crystal RG, and Sondhi D
- Subjects
- Humans, Mice, Animals, Heart, Iron-Binding Proteins genetics, Mice, Knockout, Friedreich Ataxia genetics, Friedreich Ataxia therapy, Heart Failure
- Abstract
Friedreich's ataxia (FA) is a life-threatening autosomal recessive disorder characterized by neurological and cardiac dysfunction. Arrhythmias and heart failure are the main cause of premature death. From prior studies in murine models of FA, adeno-associated virus encoding the normal human frataxin gene (AAVrh.10hFXN) effectively treated the cardiac manifestations of the disease. However, the therapeutic dose window is limited by high level of human frataxin (hFXN) gene expression associated with toxicity. As a therapeutic goal, since FA heterozygotes have no clinical manifestations of FA, we estimated the level of frataxin (FXN) necessary to convert the heart of a homozygote to that of a heterozygote. In noncardiac cells, FA heterozygotes have 30-80% of normal FXN levels (17.7-47.2 ng/mg, average 32.5 ng/mg) and FA homozygotes 2-30% normal levels (1.2-17.7 ng/mg, average 9.4 ng/mg). Therefore, an AAV vector would need to augment endogenous in an FA homozygote by >8.3 ng/mg. To determine the required dose of AAVrh.10hFXN, we administered 1.8 × 10
11 , 5.7 × 1011 , or 1.8 × 1012 gc/kg of AAVrh.10hFXN intravenously (IV) to muscle creatine kinase (mck)-Cre conditional knockout Fxn mice, a cardiac and skeletal FXN knockout model. The minimally effective dose was 5.7 × 1011 gc/kg, resulting in cardiac hFXN levels of 6.1 ± 4.2 ng/mg and a mild ( p < 0.01 compared with phosphate-buffered saline controls) improvement in mortality. A dose of 1.8 × 1012 gc/kg resulted in cardiac hFXN levels of 33.7 ± 6.4 ng/mg, a significant improvement in ejection fraction and fractional shortening ( p < 0.05, both comparisons) and a 21.5% improvement in mortality ( p < 0.001). To determine if the significantly effective dose of 1.8 × 1012 gc/kg could achieve human FA heterozygote levels in a large animal, this dose was administered IV to nonhuman primates. After 12 weeks, the vector-expressed FXN in the heart was 17.8 ± 4.9 ng/mg, comparable to the target human levels. These data identify both minimally and significantly effective therapeutic doses that are clinically relevant for the treatment of the cardiac manifestations of FA.- Published
- 2023
- Full Text
- View/download PDF
26. Safety of Intravenous Administration of an AAV8 Vector Coding for an Oxidation-Resistant Human α1-Antitrypsin for the Treatment of α1-Antitrypsin Deficiency.
- Author
-
Rosenberg JB, De BP, Greco A, Gorman N, Kooner V, Chen A, Yost-Bido M, Munoz-Zuluaga C, Kaminsky SM, Rostami M, Monette S, Crystal RG, and Sondhi D
- Subjects
- Humans, Mice, Animals, alpha 1-Antitrypsin genetics, alpha 1-Antitrypsin metabolism, Lung metabolism, Antibodies, Administration, Intravenous, alpha 1-Antitrypsin Deficiency genetics, alpha 1-Antitrypsin Deficiency therapy, Pulmonary Emphysema drug therapy, Pulmonary Emphysema genetics
- Abstract
α1-antitrypsin (AAT) deficiency is a common autosomal recessive hereditary disorder, with a high risk for the development of early-onset panacinar emphysema. AAT, produced primarily in the liver, functions to protect the lung from neutrophil protease; with AAT deficiency, unimpeded neutrophil proteases destroy the lung parenchyma. AAT is susceptible to oxidative damage resulting in an inability to inhibit its target proteases, neutrophil elastase, and cathepsin G. The major sites of oxidative modification on the AAT molecule are methionine residues 351 and 358. We have previously demonstrated that an engineered variant of AAT that resists oxidation by modifying both protein surface methionines (M351V and M358L) provides antiprotease protection, despite oxidative stress. In mice, intravenous delivery of the modified AAT(AVL) variant by AAV serotype 8, AAV8hAAT(AVL), primarily to the liver resulted in long-term expression of an AAT that resists oxidative inactivation. In this study, we evaluated the safety of intravenous administration of AAV8hAAT(AVL) in a dose-escalating, blinded, placebo-controlled toxicology study in wild-type mice. The study assessed organ histology and clinical pathology findings of mice, intravenously administered AAV8hAAT(AVL) at three doses (5.0 × 10
11 , 5.0 × 1012 , and 5.0 × 1013 genome copies [gc]/kg), compared to control mice injected intravenously with phosphate-buffered saline. As previously demonstrated, administration of AAV8hAAT(AVL) resulted in dose-dependent expression of high, potentially therapeutic, levels of serum human AAT protein that persist for at least 6 months. Antibodies against the AAV8 capsid were elicited as expected, but there was no antibody detected against the AAT(AVL) protein generated by the AAV8hAAT(AVL) vector. There was no morbidity or mortality observed in the study. The data demonstrate that intravenous administration of AAV8hAAT(AVL) is safe with no significant adverse effect attributed to AAV8hAAT(AVL) vector at any dose. This study demonstrates that AAV8hAAT(AVL) has a safety profile consistent with the requirements for proceeding to a clinical study.- Published
- 2023
- Full Text
- View/download PDF
27. Long-term functional correction of cystathionine β-synthase deficiency in mice by adeno-associated viral gene therapy.
- Author
-
Lee HO, Salami CO, Sondhi D, Kaminsky SM, Crystal RG, and Kruger WD
- Subjects
- Animals, Cystathionine beta-Synthase blood, Cystathionine beta-Synthase deficiency, Disease Models, Animal, Female, Gene Expression, Gene Transfer Techniques, Genetic Vectors administration & dosage, Homocystinuria metabolism, Liver metabolism, Liver pathology, Male, Mice, Mice, Knockout, Phenotype, Cystathionine beta-Synthase genetics, Dependovirus genetics, Genetic Therapy, Genetic Vectors genetics, Homocystinuria genetics, Homocystinuria therapy
- Abstract
Cystathionine β-synthase (CBS) deficiency is a recessive inborn error of sulfur metabolism characterized by elevated blood levels of total homocysteine (tHcy). Patients diagnosed with CBS deficiency are currently treated by a combination of vitamin supplementation and restriction of foods containing the homocysteine precursor methionine, but the effectiveness of this therapy is limited due to poor compliance. A mouse model for CBS deficiency (Tg-I278T Cbs
-/- ) was used to evaluate a potential gene therapy approach to treat CBS deficiency utilizing an AAVrh.10-based vector containing the human CBS cDNA downstream of the constitutive, strong CAG promoter (AAVrh.10hCBS). Mice were administered a single dose of virus and followed for up to 1 year. The data demonstrated a dose-dependent increase in liver CBS activity and a dose-dependent decrease in serum tHcy. Liver CBS enzyme activity at 1 year was similar to Cbs+/- control mice. Mice given the highest dose (5.6 × 1011 genomes/mouse) had mean serum tHcy decrease of 97% 1 week after injection and an 81% reduction 1 year after injection. Treated mice had either full- or substantial correction of alopecia, bone loss, and fat mass phenotypes associated with Cbs deficiency in mice. Our findings show that AAVrh.10-based gene therapy is highly effective in treating CBS deficiency in mice and supports additional pre-clinical testing for eventual use human trials., (© 2021 SSIEM.)- Published
- 2021
- Full Text
- View/download PDF
28. Automated Retinal Layer Segmentation in CLN2-Associated Disease: Commercially Available Software Characterizing a Progressive Maculopathy.
- Author
-
Kovacs KD, Orlin A, Sondhi D, Kaminsky SM, D'Amico DJ, Crystal RG, and Kiss S
- Subjects
- Child, Preschool, Humans, Retrospective Studies, Software, Tomography, Optical Coherence, Tripeptidyl-Peptidase 1, Macular Degeneration, Retina diagnostic imaging
- Abstract
Purpose: CLN2-associated disease is a hereditary, fatal lysosomal storage disorder characterized by progressive brain and retinal deterioration. Here, we characterize the inner and outer retinal degeneration using automated segmentation software in optical coherence tomography scans, providing an objective, quantifiable metric for monitoring subtle changes previously identified with a validated disease classification scale (the Weill Cornell Batten Scale)., Methods: This study is a retrospective, single-center cohort review of images from examinations under anesthesia in treatment-naïve patients with CLN2-associated disease. Automated segmentation software was used to delineate retinal nerve fiber, ganglion cell layer (GCL), and outer nuclear layer (ONL) thickness measurements in the fovea, parafovea, and perifovea based on age groups (months): 30 to 38, 39 to 45, 46 to 52, 53 to 59, 60 to 66, and 67 or older., Results: Twenty-seven eyes from 14 patients were included, with 8 serial images yielding 36 interpretable optical coherence tomography scans. There was a significant difference in parafoveal ONL thickness between 39 to 45 and 46 to 52 months of age (P = 0.032) not seen in other regions or retinal layers. Perifoveal ONL demonstrated a difference in thickness between the 60 to 66 and greater than 67 months age cohorts (P = 0.047). There was strong symmetry between eyes, and high segmentation repeatability., Conclusions: Parafoveal ONL thickness represents a sensitive, early age indicator of CLN2-associated degeneration. Outer retinal degeneration is apparent at younger ages than inner retinal changes though in treatment-naïve patients all retinal layers showed significant differences between 60 to 66 and more than 67 months of age., Translational Relevance: This study establishes sensitive, quantitative biomarkers for assessing retinal degeneration in a large cohort natural history study in anticipation of future clinical trials.
- Published
- 2021
- Full Text
- View/download PDF
29. Safety of Direct Intraparenchymal AAVrh.10-Mediated Central Nervous System Gene Therapy for Metachromatic Leukodystrophy.
- Author
-
Rosenberg JB, Chen A, De BP, Dyke JP, Ballon DJ, Monette S, Ricart Arbona RJ, Kaminsky SM, Crystal RG, and Sondhi D
- Subjects
- Animals, Central Nervous System, Cerebroside-Sulfatase genetics, Child, Dependovirus genetics, Genetic Therapy, Genetic Vectors genetics, Humans, Mice, Leukodystrophy, Metachromatic genetics, Leukodystrophy, Metachromatic therapy
- Abstract
Metachromatic leukodystrophy, a fatal pediatric neurodegenerative lysosomal storage disease caused by mutations in the arylsulfatase A ( ARSA ) gene, is characterized by intracellular accumulation of sulfatides in the lysosomes of cells of the central nervous system (CNS). In previous studies, we have demonstrated efficacy of AAVrh.10hARSA, an adeno-associated virus (AAV) serotype rh.10 vector coding for the human ARSA gene to the CNS of a mouse model of the disease, and that catheter-based intraparenchymal administration of AAVrh.10hARSA to the CNS of nonhuman primates (NHPs) white matter results in widespread expression of ARSA. As a formal dose-escalating safety/toxicology study, we assessed the safety of intraparenchymal delivery of AAVrh.10hARSA vector to 12 sites in the white matter of the CNS of NHPs at 2.85 × 10
10 (total low dose, 2.4 × 109 genome copies [gc]/site) and 1.5 × 1012 (total high dose, 1.3 × 1011 gc/site) gc, compared to AAVrh.10Null (1.5 × 1012 gc total, 1.3 × 1011 gc/site) as a vector control, and phosphate buffered saline for a sham surgical control. No significant adverse effects were observed in animals treated with low dose AAVrh.10hARSA. However, animals treated with the high dose AAVrh.10ARSA and the high dose Null vector had highly localized CNS abnormalities on magnetic resonance imaging scans at the sites of catheter infusions, and histopathology demonstrated that these sites were associated with infiltrates of T cells, B cells, microglial cells, and/or macrophages. Although these findings had no clinical consequences, these safety data contribute to understanding the dose limits for CNS white matter direct intraparenchymal administration of AAVrh.10 vectors for treatment of CNS disorders.- Published
- 2021
- Full Text
- View/download PDF
30. Slowing late infantile Batten disease by direct brain parenchymal administration of a rh.10 adeno-associated virus expressing CLN2 .
- Author
-
Sondhi D, Kaminsky SM, Hackett NR, Pagovich OE, Rosenberg JB, De BP, Chen A, Van de Graaf B, Mezey JG, Mammen GW, Mancenido D, Xu F, Kosofsky B, Yohay K, Worgall S, Kaner RJ, Souwedaine M, Greenwald BM, Kaplitt M, Dyke JP, Ballon DJ, Heier LA, Kiss S, and Crystal RG
- Subjects
- Aminopeptidases genetics, Brain, Child, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases genetics, Genetic Therapy, Humans, Magnetic Resonance Imaging, Tripeptidyl-Peptidase 1, Dependovirus genetics, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses therapy
- Abstract
Late infantile Batten disease (CLN2 disease) is an autosomal recessive, neurodegenerative lysosomal storage disease caused by mutations in the CLN2 gene encoding tripeptidyl peptidase 1 (TPP1). We tested intraparenchymal delivery of AAVrh.10hCLN2, a nonhuman serotype rh.10 adeno-associated virus vector encoding human CLN2 , in a nonrandomized trial consisting of two arms assessed over 18 months: AAVrh.10hCLN2-treated cohort of 8 children with mild to moderate disease and an untreated, Weill Cornell natural history cohort consisting of 12 children. The treated cohort was also compared to an untreated European natural history cohort of CLN2 disease. The vector was administered through six burr holes directly to 12 sites in the brain without immunosuppression. In an additional safety assessment under a separate protocol, five children with severe CLN2 disease were treated with AAVrh.10hCLN2. The therapy was associated with a variety of expected adverse events, none causing long-term disability. Induction of systemic anti-AAVrh.10 immunity was mild. After therapy, the treated cohort had a 1.3- to 2.6-fold increase in cerebral spinal fluid TPP1. There was a slower loss of gray matter volume in four of seven children by MRI and a 42.4 and 47.5% reduction in the rate of decline of motor and language function, compared to Weill Cornell natural history cohort ( P < 0.04) and European natural history cohort ( P < 0.0001), respectively. Intraparenchymal brain administration of AAVrh.10hCLN2 slowed the progression of disease in children with CLN2 disease. However, improvements in vector design and delivery strategies will be necessary to halt disease progression using gene therapy., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
31. Quantitative Whole-Body Imaging of I-124-Labeled Adeno-Associated Viral Vector Biodistribution in Nonhuman Primates.
- Author
-
Ballon DJ, Rosenberg JB, Fung EK, Nikolopoulou A, Kothari P, De BP, He B, Chen A, Heier LA, Sondhi D, Kaminsky SM, Mozley PD, Babich JW, and Crystal RG
- Subjects
- Animals, Brain metabolism, Brain pathology, Brain virology, Dependovirus chemistry, Genetic Vectors genetics, Humans, Iodine Radioisotopes chemistry, Primates, Tissue Distribution drug effects, Brain diagnostic imaging, Dependovirus genetics, Iodine Radioisotopes pharmacology, Whole Body Imaging methods
- Abstract
A method is presented for quantitative analysis of the biodistribution of adeno-associated virus (AAV) gene transfer vectors following in vivo administration. We used iodine-124 (I-124) radiolabeling of the AAV capsid and positron emission tomography combined with compartmental modeling to quantify whole-body and organ-specific biodistribution of AAV capsids from 1 to 72 h following administration. Using intravenous (IV) and intracisternal (IC) routes of administration of AAVrh.10 and AAV9 vectors to nonhuman primates in the absence or presence of anticapsid immunity, we have identified novel insights into initial capsid biodistribution and organ-specific capsid half-life. Neither I-124-labeled AAVrh.10 nor AAV9 administered intravenously was detected at significant levels in the brain relative to the administered vector dose. Approximately 50% of the intravenously administered labeled capsids were dispersed throughout the body, independent of the liver, heart, and spleen. When administered by the IC route, the labeled capsid had a half-life of ∼10 h in the cerebral spinal fluid (CSF), suggesting that by this route, the CSF serves as a source with slow diffusion into the brain. For both IV and IC administration, there was significant influence of pre-existing anticapsid immunity on I-124-capsid biodistribution. The methodology facilitates quantitative in vivo viral vector dosimetry, which can serve as a technique for evaluation of both on- and off-target organ biodistribution, and potentially accelerate gene therapy development through rapid prototyping of novel vector designs.
- Published
- 2020
- Full Text
- View/download PDF
32. Cocaine vaccine dAd5GNE protects against moderate daily and high-dose "binge" cocaine use.
- Author
-
Havlicek DF, Rosenberg JB, De BP, Hicks MJ, Sondhi D, Kaminsky SM, and Crystal RG
- Subjects
- Adenoviridae genetics, Animals, Antibodies blood, Antibodies immunology, Chlorocebus aethiops, Cocaine administration & dosage, Cocaine adverse effects, Cocaine therapeutic use, Female, Mice, Mice, Inbred BALB C, Receptors, Drug metabolism, Vaccination, Vaccines therapeutic use, Cocaine analogs & derivatives, Cocaine immunology, Cocaine-Related Disorders prevention & control
- Abstract
The cocaine vaccine dAd5GNE is comprised of a disrupted serotype 5 adenovirus gene therapy vector covalently conjugated to the cocaine analog GNE. The vaccine evokes a high titer of circulating anti-cocaine antibodies that prevent cocaine from reaching its cognate receptors in the central nervous system. Prior studies have demonstrated the efficacy of dAd5GNE in models of occasional, moderate cocaine use. However, previous studies have not sufficiently evaluated the efficacy of dAd5GNE in models of the repetitive and high-dose "binge" use patterns common in human addicts. In the present study, we evaluated the capacity of dAd5GNE vaccination to protect against "binge" cocaine use and circumstances where vaccinated addicts attempt to override the vaccine. We modeled repetitive daily cocaine use in vaccinated Balb/c mice and African green monkeys, and evaluated high-dose "binge" scenarios in Balb/c mice. In each model of daily use the dAd5GNE vaccine prevented cocaine from reaching the central nervous system. In the high-dose "binge" model, vaccination decreased cocaine-induced hyperactivity and reduced the number of cocaine-induced seizures. Based on this data and our prior data in rodents and nonhuman primates, we have initiated a clinical trial evaluating the dAd5GNE anti-cocaine vaccine as a potential therapy for cocaine addicts who wish to stop cocaine use. If dAd5GNE vaccination is safe and produces high anti-cocaine antibody titers in the clinic, we hypothesize that the vaccine will restrict the access of cocaine to the central nervous system and inhibit cocaine-induced "highs" even in the context of moderate daily and high-dose "binge" use that might otherwise cause a drug-induced overdose., Competing Interests: The authors have read the journal's policy and have the following competing interests: SMK, JBR, BPD, MJH, RGC are listed on Cornell University intellectual property regarding the dAd5GNE vaccine. RGC, SMK and DS are consultant to, and have equity in, LEXEO Therapeutics which has an option to license this intellectual property and data from Cornell University. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2020
- Full Text
- View/download PDF
33. Stress-Induced Mouse Model of the Cardiac Manifestations of Friedreich's Ataxia Corrected by AAV-mediated Gene Therapy.
- Author
-
Salami CO, Jackson K, Jose C, Alyass L, Cisse GI, De BP, Stiles KM, Chiuchiolo MJ, Sondhi D, Crystal RG, and Kaminsky SM
- Subjects
- Animals, Cardiomyopathies etiology, Cardiomyopathies metabolism, Cardiomyopathies pathology, Disease Models, Animal, Female, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Phenotype, Frataxin, Cardiomyopathies therapy, Dependovirus genetics, Friedreich Ataxia complications, Genetic Therapy, Genetic Vectors administration & dosage, Iron-Binding Proteins genetics, Stress, Physiological
- Abstract
Friedreich's ataxia (FA), an autosomal recessive disorder caused by a deficiency in the expression of frataxin (FXN), is characterized by progressive ataxia and hypertrophic cardiomyopathy. Although cardiac dysfunction is the most common cause of mortality in FA, the cardiac disease remains subclinical for most of the clinical course because the neurologic disease limits muscle oxygen demands. Previous FXN knockout mouse models exhibit fatal cardiomyopathy similar to human FA, but in contrast to the human condition, untreated mice become moribund by 2 months of age, unlike humans where the cardiac disease often does not manifest until the third decade. The study was designed to create a mouse model for early FA disease relevant to the time for which a gene therapy would likely be most effective. To generate a cardiac-specific mouse model of FA cardiomyopathy similar to the human disease, we used a cardiac promoter (αMyhc) driving CRE recombinase cardiac-specific excision of FXN exon 4 to generate a mild, cardiac-specific FA model that is normal at rest, but exhibits the cardiac phenotype with stress. The hearts of αMyhc mice had decreased levels of FXN and activity of the mitochondrial complex II/complex IV respiratory chain. At rest, αMyhc mice exhibited normal cardiac function as assessed by echocardiographic assessment of ejection fraction and fractional shortening, but when the heart was stressed chemically with dobutamine, αMyhc mice compared with littermate control mice had a 62% reduction in the stress ejection fraction ( p < 2 × 10
-4 ) and 71% reduction in stress-related fractional shortening ( p < 10-5 ). When assessing functional cardiac performance using running on an inclined treadmill, αMyhc mice stayed above the midline threefold less than littermate controls ( p < 0.02). A one-time intravenous administration of 1011 genome copies of AAVrh.10hFXN, an adeno-associated virus (AAV) serotype rh10 gene transfer vector expressing human FXN , corrected the stress-induced ejection fraction and fractional shortening phenotypes. Treated αMyhc mice exhibited exercise performance on a treadmill indistinguishable from littermate controls ( p > 0.07). These αMyhc mice provide an ideal model to study long-term cardiac complications due to FA and AAV-mediated gene therapy correction of stress-induced cardiac phenotypes typical of human FA.- Published
- 2020
- Full Text
- View/download PDF
34. Symmetric Age Association of Retinal Degeneration in Patients with CLN2-Associated Batten Disease.
- Author
-
Kovacs KD, Patel S, Orlin A, Kim K, Van Everen S, Conner T, Sondhi D, Kaminsky SM, D'Amico DJ, Crystal RG, and Kiss S
- Subjects
- Aminopeptidases metabolism, Child, Preschool, DNA Mutational Analysis, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases metabolism, Fluorescein Angiography, Fundus Oculi, Humans, Infant, Macular Degeneration diagnosis, Neuronal Ceroid-Lipofuscinoses complications, Neuronal Ceroid-Lipofuscinoses diagnosis, Retrospective Studies, Serine Proteases metabolism, Tomography, Optical Coherence methods, Tripeptidyl-Peptidase 1, Aminopeptidases genetics, DNA genetics, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases genetics, Macular Degeneration etiology, Mutation, Neuronal Ceroid-Lipofuscinoses genetics, Retina pathology, Serine Proteases genetics, Visual Acuity
- Abstract
Purpose: Mutations in the CLN2 gene lead to a neurodegenerative and blinding lysosomal storage disorder: late infantile neuronal ceroid lipofucinosis, also known as "CLN2 disease." The purpose of the current study was to characterize the evolution of CLN2-associated retinal manifestations using the Weill Cornell Batten Scale (WCBS) and the age association of the retinal degeneration using central subfield thickness (CST) measurements and then correlate these findings with fundus photography and OCT to determine a critical period for retinal intervention., Design: Retrospective, single-center cohort., Participants: Eighty-four eyes of 42 treatment-naïve patients with CLN2 disease., Methods: Clinical records, fundus photographs, and OCT imaging for patients with CLN2 disease collected during examinations under anesthesia were reviewed. Imaging was categorized per WCBS criteria by 3 masked graders., Main Outcome Measures: CLN2-associated retinopathy assessed using WCBS scores, fundus photographs, and OCT imaging, correlated with patient age., Results: Eighty-four eyes of 42 patients had baseline fundus photographs, with baseline OCT in 31 eyes of 16 patients. Fundus photographs were obtained serially for 26 eyes of 13 patients, and serial OCT scans were obtained in 10 eyes of 5 patients. At baseline, bilateral WCBS scores were highly correlated for OCT and fundus photographs (r = 0.96 and 0.82, respectively). Central subfield thickness was negatively correlated with left and right eye WCBS OCT scores (r = -0.92 and -0.83, respectively; P < 0.001) and fundus photograph scores (r = -0.80 and -0.83, respectively; P < 0.001). OCT thickness was symmetrical between each eye. Baseline OCT data with age fit using a sigmoid function demonstrated a period of accelerated loss between 48 and 72 months of age., Conclusions: Retinal degeneration associated with CLN2 disease manifests as a progressive, symmetrical decline, which appears to accelerate during a critical period at 48 to 72 months of age, suggesting intervention with retina-specific CLN2 gene therapy should occur ideally before or as early as possible within this critical period. The WCBS is a valuable tool and is highly correlated with the extent of retinal degeneration observed in OCT or fundus photographs; by using the fellow eye as a control, this grading scale can be used to monitor the effect of CLN2 gene therapy in future trials., (Copyright © 2020 American Academy of Ophthalmology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. Anti-Phospho-Tau Gene Therapy for Chronic Traumatic Encephalopathy.
- Author
-
Sacramento CB, Sondhi D, Rosenberg JB, Chen A, Giordano S, Pey E, Lee V, Stiles KM, Havlicek DF, Leopold PL, Kaminsky SM, and Crystal RG
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Brain metabolism, Brain pathology, Dependovirus genetics, Disease Management, Disease Models, Animal, Disease Susceptibility, Genetic Vectors genetics, Humans, Immunohistochemistry, Male, Mice, Mice, Transgenic, Neurons metabolism, Protein Binding, tau Proteins antagonists & inhibitors, tau Proteins metabolism, Chronic Traumatic Encephalopathy genetics, Chronic Traumatic Encephalopathy therapy, Genetic Therapy adverse effects, Genetic Therapy methods, tau Proteins genetics
- Abstract
Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disorder caused by repetitive trauma to the central nervous system (CNS) suffered by soldiers, contact sport athletes, and civilians following accident-related trauma. CTE is a CNS tauopathy, with trauma-induced inflammation leading to accumulation of hyperphosphorylated forms of the microtubule-binding protein Tau (pTau), resulting in neurofibrillary tangles and progressive loss of neurons. At present, there are no therapies to treat CTE. We hypothesized that direct CNS administration of an adeno-associated virus (AAV) vector coding for an anti-pTau antibody would generate sufficient levels of anti-pTau in the CNS to suppress pTau accumulation thus interrupting the pathogenic process. Using a serotype AAVrh.10 gene transfer vector coding for a monoclonal antibody directed against pTau, we demonstrate the feasibility of this strategy in a murine CTE model in which pTau accumulation was elicited by repeated traumatic brain injury (TBI) using a closed cortical impact procedure over 5 days. Direct delivery of AAVrh.10 expression vectors coding for either of the two different anti-pTau antibodies to the hippocampus of these TBI mice significantly reduced pTau levels across the CNS. Using doses that can be safely scaled to humans, the data demonstrate that CNS administration of AAVrh.10anti-pTau is effective, providing a new strategy to interrupt the CTE consequences of TBI.
- Published
- 2020
- Full Text
- View/download PDF
36. Gene therapy for C1 esterase inhibitor deficiency in a Murine Model of Hereditary angioedema.
- Author
-
Qiu T, Chiuchiolo MJ, Whaley AS, Russo AR, Sondhi D, Kaminsky SM, Crystal RG, and Pagovich OE
- Subjects
- Animals, CRISPR-Cas Systems, Capillary Permeability genetics, Dependovirus genetics, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Transgenic, Mutation, Phenotype, Transgenes, Angioedemas, Hereditary therapy, Complement C1 Inhibitor Protein genetics, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors therapeutic use
- Abstract
Background: Hereditary angioedema (HAE) is a life-threatening, autosomal dominant disorder characterized by unpredictable, episodic swelling of the face, upper airway, oropharynx, extremities, genitalia, and gastrointestinal tract. Almost all cases of HAE are caused by mutations in the SERPING1 gene resulting in a deficiency in functional plasma C1 esterase inhibitor (C1EI), a serine protease inhibitor that normally inhibits proteases in the contact, complement, and fibrinolytic systems. Current treatment of HAE includes long-term prophylaxis with attenuated androgens or human plasma-derived C1EI and management of acute attacks with human plasma-derived or recombinant C1EI, bradykinin, and kallikrein inhibitors, each of which requires repeated administration. As an approach to effectively treat HAE with a single treatment, we hypothesized that a one-time intravenous administration of an adeno-associated virus (AAV) gene transfer vector expressing the genetic sequence of the normal human C1 esterase inhibitor (AAVrh.10hC1EI) would provide sustained circulating C1EI levels sufficient to prevent angioedema episodes., Methods: To study the efficacy of AAVrh.10hC1EI, we used CRISPR/Cas9 technology to create a heterozygote C1EI-deficient mouse model (S63±) that shares characteristics associated with HAE in humans including decreased plasma C1EI and C4 levels. Phenotypically, these mice have increased vascular permeability of skin and internal organs., Results: Systemic administration of AAVrh.10hC1EI to the S63± mice resulted in sustained human C1EI activity levels above the predicted therapeutic levels and correction of the vascular leak in skin and internal organs., Conclusion: A single treatment with AAVrh.10hC1EI has the potential to provide long-term protection from angioedema attacks in affected individuals., (© 2018 EAACI and John Wiley and Sons A/S. Published by John Wiley and Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
37. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis.
- Author
-
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, and Sondhi D
- Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura
® ) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL., Areas Covered: The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal., Expert Opinion: Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older., Competing Interests: Declaration of Interest. RGC is a consultant to BioMarin. All other authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
38. Untargeted Metabolite Profiling of Cerebrospinal Fluid Uncovers Biomarkers for Severity of Late Infantile Neuronal Ceroid Lipofuscinosis (CLN2, Batten Disease).
- Author
-
Sindelar M, Dyke JP, Deeb RS, Sondhi D, Kaminsky SM, Kosofsky BE, Ballon DJ, Crystal RG, and Gross SS
- Subjects
- Acetates metabolism, Adolescent, Adult, Aged, Aminopeptidases cerebrospinal fluid, Aminopeptidases genetics, Animals, Brain pathology, Child, Child, Preschool, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases cerebrospinal fluid, Dipeptidyl-Peptidases and Tripeptidyl-Peptidases genetics, Disease Models, Animal, Female, Humans, Male, Metabolomics, Middle Aged, Mitochondria metabolism, Mitochondria pathology, Neuronal Ceroid-Lipofuscinoses cerebrospinal fluid, Neuronal Ceroid-Lipofuscinoses pathology, Neurons metabolism, Neurons pathology, Serine Proteases cerebrospinal fluid, Serine Proteases genetics, Severity of Illness Index, Tripeptidyl-Peptidase 1, Young Adult, Biomarkers cerebrospinal fluid, Brain metabolism, Metabolome genetics, Neuronal Ceroid-Lipofuscinoses genetics, Neuronal Ceroid-Lipofuscinoses metabolism
- Abstract
Late infantile neuronal ceroid lipofuscinosis (CLN2 disease) is a rare lysosomal storage disorder caused by a monogenetic deficiency of tripeptidyl peptidase-1 (TPP1). Despite knowledge that lipofuscin is the hallmark disease product, the relevant TPP1 substrate and its role in neuronal physiology/pathology is unknown. We hypothesized that untargeted metabolite profiling of cerebrospinal fluid (CSF) could be used as an effective tool to identify disease-associated metabolic disruptions in CLN2 disease, offering the potential to identify biomarkers that inform on disease severity and progression. Accordingly, a mass spectrometry-based untargeted metabolite profiling approach was employed to differentiate CSF from normal vs. CLN2 deficient individuals. Of 1,433 metabolite features surveyed, 29 linearly correlated with currently employed disease severity scores. With tandem mass spectrometry 8 distinct metabolite identities were structurally confirmed based on retention time and fragmentation pattern matches, vs. standards. These putative CLN2 biomarkers include 7 acetylated species - all attenuated in CLN2 compared to controls. Because acetate is the major bioenergetic fuel for support of mitochondrial respiration, deficient acetylated species in CSF suggests a brain energy defect that may drive neurodegeneration. Targeted analysis of these metabolites in CSF of CLN2 patients offers a powerful new approach for monitoring CLN2 disease progression and response to therapy.
- Published
- 2018
- Full Text
- View/download PDF
39. Intrapleural Gene Therapy for Alpha-1 Antitrypsin Deficiency-Related Lung Disease.
- Author
-
Stiles KM, Sondhi D, Kaminsky SM, De BP, Rosenberg JB, and Crystal RG
- Abstract
Alpha-1 antitrypsin deficiency (AATD) manifests primarily as early-onset emphysema caused by the destruction of the lung by neutrophil elastase due to low amounts of the serine protease inhibitor alpha-1 antitrypsin (AAT). The current therapy involves weekly intravenous infusions of AAT-derived from pooled human plasma that is efficacious, yet costly. Gene therapy applications designed to provide constant levels of the AAT protein are currently under development. The challenge is for gene therapy to provide sufficient amounts of AAT to normalize the inhibitor level and anti-neutrophil elastase capacity in the lung. One strategy involves administration of an adeno-associated virus (AAV) gene therapy vector to the pleural space providing both local and systemic production of AAT to reach consistent therapeutic levels. This review focuses on the strategy, advantages, challenges, and updates for intrapleural administration of gene therapy vectors for the treatment of AATD., Competing Interests: Adverum Biotechnologies has licensed the AAVrh.10hAAT intrapleural technology and investigational new drug from Cornell University. RGC is a consultant and holds equity in Adverum. RGC, DS, SK share licensing fees paid to Cornell University.
- Published
- 2018
- Full Text
- View/download PDF
40. Disease characteristics and progression in patients with late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease: an observational cohort study.
- Author
-
Nickel M, Simonati A, Jacoby D, Lezius S, Kilian D, Van de Graaf B, Pagovich OE, Kosofsky B, Yohay K, Downs M, Slasor P, Ajayi T, Crystal RG, Kohlschütter A, Sondhi D, and Schulz A
- Subjects
- Adolescent, Child, Child, Preschool, Cohort Studies, Disease Progression, Female, Humans, Infant, Longitudinal Studies, Male, Tripeptidyl-Peptidase 1, Neuronal Ceroid-Lipofuscinoses diagnosis
- Abstract
Background: Late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease, characterised by rapid psychomotor decline and epilepsy, is caused by deficiency of the lysosomal enzyme tripeptidyl peptidase 1. We aimed to analyse the characteristics and rate of progression of CLN2 disease in an international cohort of patients., Methods: We did an observational cohort study using data from two independent, international datasets of patients with untreated genotypically confirmed CLN2 disease: the DEM-CHILD dataset (n=74) and the Weill Cornell Medical College (WCMC) dataset (n=66). Both datasets included quantitative rating assessments with disease-specific clinical domain scores, and disease course was measured longitudinally in 67 patients in the DEM-CHILD cohort. We analysed these data to determine age of disease onset and diagnosis, as well as disease progression-measured by the rate of decline in motor and language summary scores (on a scale of 0-6 points)-and time from first symptom to death., Findings: In the combined DEM-CHILD and WCMC dataset, median age was 35·0 months (IQR 24·0-38·5) at first clinical symptom, 37·0 months (IQR 35·0 -42·0) at first seizure, and 54·0 months (IQR 47·5-60·0) at diagnosis. Of 74 patients in the DEM-CHILD dataset, the most common first symptoms of disease were seizures (52 [70%]), language difficulty (42 [57%]), motor difficulty (30 [41%]), behavioural abnormality (12 [16%]), and dementia (seven [9%]). Among the 41 patients in the DEM-CHILD dataset for whom longitudinal assessments spanning the entire disease course were available, a rapid annual decline of 1·81 score units (95% CI 1·50-2·12) was seen in motor-language summary scores from normal (score of 6) to no function (score of 0), which occurred over approximately 30 months. Among 53 patients in the DEM-CHILD cohort with available data, the median time between onset of first disease symptom and death was 7·8 years (SE 0·9) years., Interpretation: In view of its natural history, late-infantile CLN2 disease should be considered in young children with delayed language acquisition and new onset of seizures. CLN2 disease has a largely predictable time course with regard to the loss of language and motor function, and these data might serve as historical controls for the assessment of current and future therapies., Funding: EU Seventh Framework Program, German Ministry of Education and Research, EU Horizon2020 Program, National Institutes of Health, Nathan's Battle Foundation, Cures Within Reach Foundation, Noah's Hope Foundation, Hope4Bridget Foundation., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
41. Attenuation of the Niemann-Pick type C2 disease phenotype by intracisternal administration of an AAVrh.10 vector expressing Npc2.
- Author
-
Markmann S, J Christie-Reid J, Rosenberg JB, De BP, Kaminsky SM, Crystal RG, and Sondhi D
- Subjects
- Animals, Cisterna Magna, Dependovirus genetics, Gene Expression, Genetic Vectors, Life Expectancy, Liver pathology, Mice, Mice, Inbred BALB C, Mice, Knockout, Microinjections, Motor Activity, Niemann-Pick Disease, Type C pathology, Niemann-Pick Disease, Type C psychology, Phenotype, Psychomotor Performance, Spleen pathology, Genetic Therapy methods, Niemann-Pick Disease, Type C therapy, Vesicular Transport Proteins genetics
- Abstract
Niemann-Pick type C2 (NPC2) disease is a rare, neurodegenerative disorder caused by mutations in the NPC2 gene, leading to lysosomal accumulation of unesterified cholesterol and other lipids. It is characterized by hepatosplenomegaly, liver dysfunction and severe neurological manifestations, resulting in early death. There is no effective therapy for NPC2 disease. Here, we evaluated the effectiveness of an adeno-associated virus (AAV), serotype rh.10 gene transfer vector expressing the mouse Npc2 gene (AAVrh.10-mNpc2-HA, HA tagged to facilitate analysis) to treat the disease in an Npc2-/- mouse model. A single intracisternal administration of the AAVrh.10-mNpc2-HA to 6 week old Npc2-/- mice mediated vector DNA, transgene mRNA and protein expression in brain and other organs. Compared to untreated Npc2-/- mice, AAV-treated Npc2-/- mice demonstrated amelioration of disease pathology in the brain, reduced lysosomal storage, reduced Purkinje cell death, decreased gliosis, and improved performance in behavioral tasks. Treatment-related reduction in serum disease markers was detected early and this effect persisted. Liver and spleen pathology were improved with significant reduction of liver cholesterol and sphingomyelin levels in treated Npc2-/- mice. Finally, administration of AAVrh.10-mNpc2-HA significantly extended life-span. Taken together, these data demonstrate the benefit of a one-time intracisternal administration of AAVrh.10-mNpc2-HA as a life-long treatment for NPC2 disease., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
42. In Vivo Potency Assay for Adeno-Associated Virus-Based Gene Therapy Vectors Using AAVrh.10 as an Example.
- Author
-
De BP, Chen A, Salami CO, Van de Graaf B, Rosenberg JB, Pagovich OE, Sondhi D, Crystal RG, and Kaminsky SM
- Subjects
- Animals, Dependovirus metabolism, Genetic Therapy adverse effects, Genetic Therapy standards, Genetic Vectors metabolism, Humans, Liver metabolism, Male, Mice, Mice, Inbred BALB C, RNA, Messenger genetics, RNA, Messenger metabolism, Dependovirus genetics, Genetic Therapy methods, Genetic Vectors genetics
- Abstract
The development of a drug product requires rigorous methods of characterization and quality control to assure drug potency. Gene therapy products, a relatively new strategy for drug design with very few licensed examples, represent a unique challenge for the measure of potency. Unlike traditional drugs, potency for a gene therapeutic is a tally of the measures of multiple steps, including infectivity, transcription, translation, protein modifications, proper localization of the protein product, and protein function. This is particularly challenging for products based on the adeno-associated virus (AAV) platform, which has poor in vitro infectivity, limiting the sensitivity and thus the usefulness of cell-based assays. A rigorous in vivo assay has been established that separately evaluates infection, transcription, and resulting protein levels with specifications for each based on real time polymerase chain reaction (DNA and RNA) and standard protein assays. For an acceptance criterion, an administered vector must have vector DNA, transgene mRNA, and transgene expressed protein each concurrently meet individual specifications or the production lot fails. Using the AAVrh.10 serotype as a model vector and three different transgenes as examples, the assay is based on intravenous administration of the vector to male mice. At 2 weeks, the harvested liver is homogenized and assessed for vector genome levels (to assess for vector delivery), mRNA (to assess vector infectivity and transcription), and protein in the liver or serum (to assess protein expression). For all AAV vectors, the assay is robust and reproducible: vector DNA (linearity 10
2 -109 copies, coefficient of variation) intra-assay <0.8%, inter-assay <0.5%; mRNA intra-assay <3.3%, inter-assay <3.4%. The reproducibility of the assay for transgene expressed protein is product specific. This in vivo potency assay is a strategy for characterization and a quantitative lot release test, providing a path forward to meet regulatory drug requirements for any AAV gene therapy vectors.- Published
- 2018
- Full Text
- View/download PDF
43. Biology of the Adrenal Gland Cortex Obviates Effective Use of Adeno-Associated Virus Vectors to Treat Hereditary Adrenal Disorders.
- Author
-
Markmann S, De BP, Reid J, Jose CL, Rosenberg JB, Leopold PL, Kaminsky SM, Sondhi D, Pagovich O, and Crystal RG
- Subjects
- Adrenal Glands metabolism, Adrenal Glands pathology, Adrenal Hyperplasia, Congenital pathology, Adrenal Hyperplasia, Congenital therapy, Adrenal Medulla metabolism, Animals, Apoptosis genetics, Dependovirus genetics, Disease Models, Animal, Female, Humans, Mice, Mutation, Adrenal Hyperplasia, Congenital genetics, Genetic Therapy, Steroid 21-Hydroxylase genetics
- Abstract
Congenital adrenal hyperplasia (CAH) is an autosomal recessive disorder occurring in 1:10,000 to 1:20,000 live births. In >95% of the cases, CAH results from mutations in the CYP21A2 gene, encoding the adrenal steroid enzyme 21-hydroxylase (21OH). Cardinal phenotypic features of CAH include genital ambiguity and sexual precocity, and in severe cases, neonatal salt loss and death. Current standard of care consists of lifelong oral steroid replacement to reverse the cortisol deficiency. Although significant advances in the treatment of CAH have been made, the burden of a lifelong therapeutic intervention is not ideal for quality of life. Gene therapy for CAH by adeno-associated virus (AAV) vectors has been shown to efficiently transduce the adrenal cortex, restoring normal steroidogenesis in the short term. However, adrenocortical cells are continuously renewed by stem cells located at the adrenal capsule, which differentiate as they centripetally migrate towards the adrenal medulla where they undergo apoptosis. In this context, we hypothesized that AAV-mediated genetic correction of the adrenal cortex will work short term but will eventually lead to a loss of correction. To test this hypothesis, we administered intravenously an AAV serotype rh.10 gene transfer vector (AAVrh.10-21OH-HA) to 21-hydroxylase deficient mice (21OH
-/- ). The data demonstrates that a single intravenous administration efficiently transduces adrenocortical cells leading to 21OH-HA expression and restoration of normal steroidogenesis. However, the duration of therapeutic efficacy lasted for only 8 weeks, accompanied by loss of 21OH-HA expression in the adrenal gland. Analysis in immunodeficient mice confirmed that the disappearance of transgene expression was not due to an antiviral/transgene immune response. Taken together, these results demonstrate that a single treatment with an adeno-associated viral vector expressing a functional copy of the mutated gene can only transiently treat adrenocortical hereditary disorders and that strategies to genetically modify the adrenocortical stem cells population will likely be required.- Published
- 2018
- Full Text
- View/download PDF
44. AAVrh.10-Mediated APOE2 Central Nervous System Gene Therapy for APOE4-Associated Alzheimer's Disease.
- Author
-
Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, and Crystal RG
- Subjects
- Alzheimer Disease genetics, Animals, Apolipoprotein E2 metabolism, Apolipoprotein E4 genetics, Chlorocebus aethiops, Dependovirus genetics, Genetic Therapy adverse effects, Genetic Vectors genetics, HEK293 Cells, Humans, Male, Alzheimer Disease therapy, Apolipoprotein E2 genetics, Central Nervous System metabolism, Genetic Therapy methods
- Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological disorder affecting nearly one in nine elderly people in the United States. Population studies have shown that an inheritance of the apolipoprotein E (APOE) variant APOE4 allele increases the risk of developing AD, whereas APOE2 homozygotes are protected from late-onset AD. It was hypothesized that expression of the "protective" APOE2 variant by genetic modification of the central nervous system (CNS) of APOE4 homozygotes could reverse or prevent progressive neurologic damage. To assess the CNS distribution and safety of APOE2 gene therapy for AD in a large-animal model, intraparenchymal, intracisternal, and intraventricular routes of delivery to the CNS of nonhuman primates of AAVrh.10hAPOE2-HA, an AAVrh.10 serotype coding for an HA-tagged human APOE2 cDNA sequence, were evaluated. To evaluate the route of delivery that achieves the widest extent of APOE2 expression in the CNS, the expression of APOE2 in the CNS was evaluated 2 months following vector administration for APOE2 DNA, mRNA, and protein. Finally, using conventional toxicology assays, the safety of the best route of delivery was assessed. The data demonstrated that while all three routes are capable of mediating ApoE2 expression in AD relevant regions, intracisternal delivery of AAVrh.10hAPOE2-HA safely mediated wide distribution of ApoE2 with the least invasive surgical intervention, thus providing the optimal strategy to deliver vector-mediated human APOE2 to the CNS.
- Published
- 2018
- Full Text
- View/download PDF
45. Genetic Modification of the Lung Directed Toward Treatment of Human Disease.
- Author
-
Sondhi D, Stiles KM, De BP, and Crystal RG
- Subjects
- Animals, Humans, Lung Diseases genetics, Genetic Therapy, Genetic Vectors administration & dosage, Lung Diseases therapy, Viruses genetics
- Abstract
Genetic modification therapy is a promising therapeutic strategy for many diseases of the lung intractable to other treatments. Lung gene therapy has been the subject of numerous preclinical animal experiments and human clinical trials, for targets including genetic diseases such as cystic fibrosis and α1-antitrypsin deficiency, complex disorders such as asthma, allergy, and lung cancer, infections such as respiratory syncytial virus (RSV) and Pseudomonas, as well as pulmonary arterial hypertension, transplant rejection, and lung injury. A variety of viral and non-viral vectors have been employed to overcome the many physical barriers to gene transfer imposed by lung anatomy and natural defenses. Beyond the treatment of lung diseases, the lung has the potential to be used as a metabolic factory for generating proteins for delivery to the circulation for treatment of systemic diseases. Although much has been learned through a myriad of experiments about the development of genetic modification of the lung, more work is still needed to improve the delivery vehicles and to overcome challenges such as entry barriers, persistent expression, specific cell targeting, and circumventing host anti-vector responses.
- Published
- 2017
- Full Text
- View/download PDF
46. Vectored Intracerebral Immunization with the Anti-Tau Monoclonal Antibody PHF1 Markedly Reduces Tau Pathology in Mutant Tau Transgenic Mice.
- Author
-
Liu W, Zhao L, Blackman B, Parmar M, Wong MY, Woo T, Yu F, Chiuchiolo MJ, Sondhi D, Kaminsky SM, Crystal RG, and Paul SM
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, DNA-Binding Proteins, Dependovirus immunology, Female, Hippocampus, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microinjections, Mutation genetics, Neurofibrillary Tangles pathology, Polycomb-Group Proteins, Tissue Distribution, Antibodies, Monoclonal immunology, Immunization, Passive methods, Tauopathies immunology, Tauopathies prevention & control, Transcription Factors immunology, tau Proteins genetics
- Abstract
Passive immunization with anti-tau monoclonal antibodies has been shown by several laboratories to reduce age-dependent tau pathology and neurodegeneration in mutant tau transgenic mice. These studies have used repeated high weekly doses of various tau antibodies administered systemically for several months and have reported reduced tau pathology of ∼40-50% in various brain regions. Here we show that direct intrahippocampal administration of the adeno-associated virus (AAV)-vectored anti-phospho-tau antibody PHF1 to P301S tau transgenic mice results in high and durable antibody expression, primarily in neurons. Hippocampal antibody levels achieved after AAV delivery were ∼50-fold more than those reported following repeated systemic administration. In contrast to systemic passive immunization, we observed markedly reduced (≥80-90%) hippocampal insoluble pathological tau species and neurofibrillary tangles following a single dose of AAV-vectored PHF1 compared with mice treated with an AAV-IgG control vector. Moreover, the hippocampal atrophy observed in untreated P301S mice was fully rescued by treatment with the AAV-vectored PHF1 antibody. Vectored passive immunotherapy with an anti-tau monoclonal antibody may represent a viable therapeutic strategy for treating or preventing such tauopathies as frontotemporal dementia, progressive supranuclear palsy, or Alzheimer's disease., Significance Statement: We have used an adeno-associated viral (AAV) vector to deliver the genes encoding an anti-phospho-tau monoclonal antibody, PHF1, directly to the brain of mice that develop neurodegeneration due to a tau mutation that causes frontotemporal dementia (FTD). When administered systemically, PHF1 has been shown to modestly reduce tau pathology and neurodegeneration. Since such antibodies do not readily cross the blood-brain barrier, we used an AAV vector to deliver antibody directly to the hippocampus and observed much higher antibody levels and a much greater reduction in tau pathology. Using AAV vectors to deliver antibodies like PHF1 directly to brain may constitute a novel approach to treating various neurodegenerative disorders, such as FTD and Alzheimer's disease., (Copyright © 2016 the authors 0270-6474/16/3612425-11$15.00/0.)
- Published
- 2016
- Full Text
- View/download PDF
47. Anti-hIgE gene therapy of peanut-induced anaphylaxis in a humanized murine model of peanut allergy.
- Author
-
Pagovich OE, Wang B, Chiuchiolo MJ, Kaminsky SM, Sondhi D, Jose CL, Price CC, Brooks SF, Mezey JG, and Crystal RG
- Subjects
- Animals, Cytokines genetics, Disease Models, Animal, Female, Histamine Release drug effects, Humans, Immunoglobulin E blood, Male, Mice, Mice, Inbred NOD, Plant Extracts therapeutic use, Allergens immunology, Anaphylaxis immunology, Arachis immunology, Genetic Therapy, Peanut Hypersensitivity immunology, Plant Extracts immunology, Th2 Cells immunology
- Abstract
Background: Peanuts are the most common food to provoke fatal or near-fatal anaphylactic reactions. Treatment with an anti-hIgE mAb is efficacious but requires frequent parenteral administration., Objective: Based on the knowledge that peanut allergy is mediated by peanut-specific IgE, we hypothesized that a single administration of an adeno-associated virus (AAV) gene transfer vector encoding for anti-hIgE would protect against repeated peanut exposure in the host with peanut allergy., Methods: We developed a novel humanized murine model of peanut allergy that recapitulates the human anaphylactic response to peanuts in NOD-scid IL2Rgamma
null mice transferred with blood mononuclear cells from donors with peanut allergy and then sensitized with peanut extract. As therapy, we constructed an adeno-associated rh.10 serotype vector coding for a full-length, high-affinity, anti-hIgE antibody derived from the Fab fragment of the anti-hIgE mAb omalizumab (AAVrh.10anti-hIgE). In the reconstituted mice peanut-specific IgE was induced by peanut sensitization and hypersensitivity, and reactions were provoked by feeding peanuts to mice with symptoms similar to those of human subjects with peanut allergy., Results: A single administration of AAVrh.10anti-hIgE vector expressed persistent levels of anti-hIgE. The anti-hIgE vector, administered either before sensitization or after peanut sensitization and manifestation of the peanut-induced phenotype, blocked IgE-mediated alterations in peanut-induced histamine release, anaphylaxis scores, locomotor activity, and free IgE levels and protected animals from death caused by anaphylaxis., Conclusion: If this degree of persistent efficacy translates to human subjects, AAVrh.10anti-hIgE could be an effective 1-time preventative therapy for peanut allergy and possibly other severe, IgE-mediated allergies., (Copyright © 2016 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
48. Gene therapy for metachromatic leukodystrophy.
- Author
-
Rosenberg JB, Kaminsky SM, Aubourg P, Crystal RG, and Sondhi D
- Subjects
- Animals, Cerebroside-Sulfatase deficiency, Cerebroside-Sulfatase genetics, Disease Models, Animal, Humans, Leukodystrophy, Metachromatic genetics, Genetic Therapy methods, Leukodystrophy, Metachromatic therapy
- Abstract
Leukodystrophies (LDs) are rare, often devastating genetic disorders with neurologic symptoms. There are currently no disease-specific therapeutic approaches for these diseases. In this review we use metachromatic leukodystrophy as an example to outline in the brief the therapeutic approaches to MLD that have been tested in animal models and in clinical trials, such as enzyme-replacement therapy, bone marrow/umbilical cord blood transplants, ex vivo transplantation of genetically modified hematopoietic stem cells, and gene therapy. These studies suggest that to be successful the ideal therapy for MLD must provide persistent and high level expression of the deficient gene, arylsulfatase A in the CNS. Gene therapy using adeno-associated viruses is therefore the ideal choice for clinical development as it provides the best balance of potential for efficacy with reduced safety risk. Here we have summarized the published preclinical data from our group and from others that support the use of a gene therapy with AAVrh.10 serotype for clinical development as a treatment for MLD, and as an example of the potential of gene therapy for LDs especially for Krabbe disease, which is the focus of this special issue. © 2016 Wiley Periodicals, Inc., Competing Interests: None for all authors, (© 2016 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
49. Anti-Epidermal Growth Factor Receptor Gene Therapy for Glioblastoma.
- Author
-
Hicks MJ, Chiuchiolo MJ, Ballon D, Dyke JP, Aronowitz E, Funato K, Tabar V, Havlicek D, Fan F, Sondhi D, Kaminsky SM, and Crystal RG
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation genetics, Cell Transformation, Neoplastic, Cetuximab therapeutic use, Dependovirus genetics, Gene Expression Regulation, Neoplastic genetics, Genetic Vectors genetics, Glioblastoma pathology, Humans, Male, Mice, Survival Analysis, Cetuximab genetics, Cetuximab immunology, ErbB Receptors immunology, Genetic Therapy methods, Glioblastoma genetics, Glioblastoma therapy
- Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary intracranial brain tumor in adults with a mean survival of 14 to 15 months. Aberrant activation of the epidermal growth factor receptor (EGFR) plays a significant role in GBM progression, with amplification or overexpression of EGFR in 60% of GBM tumors. To target EGFR expressed by GBM, we have developed a strategy to deliver the coding sequence for cetuximab, an anti-EGFR antibody, directly to the CNS using an adeno-associated virus serotype rh.10 gene transfer vector. The data demonstrates that single, local delivery of an anti-EGFR antibody by an AAVrh.10 vector coding for cetuximab (AAVrh.10Cetmab) reduces GBM tumor growth and increases survival in xenograft mouse models of a human GBM EGFR-expressing cell line and patient-derived GBM. AAVrh10.CetMab-treated mice displayed a reduction in cachexia, a significant decrease in tumor volume and a prolonged survival following therapy. Adeno-associated-directed delivery of a gene encoding a therapeutic anti-EGFR monoclonal antibody may be an effective strategy to treat GBM., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
50. Intracerebral adeno-associated virus gene delivery of apolipoprotein E2 markedly reduces brain amyloid pathology in Alzheimer's disease mouse models.
- Author
-
Zhao L, Gottesdiener AJ, Parmar M, Li M, Kaminsky SM, Chiuchiolo MJ, Sondhi D, Sullivan PM, Holtzman DM, Crystal RG, and Paul SM
- Subjects
- Age of Onset, Alleles, Alzheimer Disease pathology, Alzheimer Disease therapy, Animals, Brain pathology, Disease Models, Animal, Genotype, Mice, Knockout, Molecular Targeted Therapy, Risk Factors, Alzheimer Disease metabolism, Alzheimer Disease prevention & control, Amyloid beta-Peptides metabolism, Apolipoprotein E2 genetics, Brain metabolism, Dependovirus, Gene Transfer Techniques, Genetic Therapy methods, Genetic Vectors
- Abstract
The common apolipoprotein E alleles (ε4, ε3, and ε2) are important genetic risk factors for late-onset Alzheimer's disease, with the ε4 allele increasing risk and reducing the age of onset and the ε2 allele decreasing risk and markedly delaying the age of onset. Preclinical and clinical studies have shown that apolipoprotein E (APOE) genotype also predicts the timing and amount of brain amyloid-β (Aβ) peptide deposition and amyloid burden (ε4 >ε3 >ε2). Using several administration protocols, we now report that direct intracerebral adeno-associated virus (AAV)-mediated delivery of APOE2 markedly reduces brain soluble (including oligomeric) and insoluble Aβ levels as well as amyloid burden in 2 mouse models of brain amyloidosis whose pathology is dependent on either the expression of murine Apoe or more importantly on human APOE4. The efficacy of APOE2 to reduce brain Aβ burden in either model, however, was highly dependent on brain APOE2 levels and the amount of pre-existing Aβ and amyloid deposition. We further demonstrate that a widespread reduction of brain Aβ burden can be achieved through a single injection of vector via intrathalamic delivery of AAV expressing APOE2 gene. Our results demonstrate that AAV gene delivery of APOE2 using an AAV vector rescues the detrimental effects of APOE4 on brain amyloid pathology and may represent a viable therapeutic approach for treating or preventing Alzheimer's disease especially if sufficient brain APOE2 levels can be achieved early in the course of the disease., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.