418 results on '"Shankar, Suma"'
Search Results
2. Prader-Willi and Angelman Syndromes: Mechanisms and Management
- Author
-
Ma, Van K, Mao, Rong, Toth, Jessica N, Fulmer, Makenzie L, Egense, Alena S, and Shankar, Suma P
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Clinical Research ,Brain Disorders ,Rare Diseases ,Congenital Structural Anomalies ,Intellectual and Developmental Disabilities (IDD) ,Pediatric ,Congenital ,Quality Education ,chromosome 15 ,developmental delay ,hyperphagia ,imprinting disorders ,obesity ,uniparental disomy ,Clinical Sciences ,Oncology and Carcinogenesis ,Clinical sciences ,Oncology and carcinogenesis - Abstract
Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are genetic imprinting disorders resulting from absent or reduced expression of paternal or maternal genes in chromosome 15q11q13 region, respectively. The most common etiology is deletion of the maternal or paternal 15q11q13 region. Methylation is the first line for molecular diagnostic testing; MS-MLPA is the most sensitive test. The molecular subtype of PWS/AS provides more accurate recurrence risk information for parents and for the individual affected with the condition. Management should include a multidisciplinary team by various medical subspecialists and therapists. Developmental and behavioral management of PWS and AS in infancy and early childhood includes early intervention services and individualized education programs for school-aged children. Here, we compare and discuss the mechanisms, pathophysiology, clinical features, and management of the two imprinting disorders, PWS and AS.
- Published
- 2023
3. Contributors
- Author
-
Albokhari, Daniah, primary, Ayoubieh, Houriya, additional, Balwani, Manisha, additional, Barry, Jessica C., additional, Blagowidow, Natalie, additional, Bodurtha, Joann, additional, Bottini, Alexander, additional, Brewer, Takae M., additional, Brodie, Scott, additional, Brown, Emily E., additional, Bush, Lynn Wein, additional, Butterfield, Russell J., additional, Campbell, Kirk, additional, Clemens, Douglas K., additional, Corson, Virginia L., additional, Cytrynbaum, Cheryl, additional, Dedania, Vaidehi, additional, Diaz, George A., additional, Dietz, Harry C., additional, Dinulos, Mary Beth Palko, additional, Eng, Christine M., additional, Eng, Charis, additional, Fan, Audrey L., additional, Francomano, Clair A., additional, Frucht, Steven J., additional, Ganesh, Jaya, additional, Gelb, Bruce D., additional, Goduni, Lediana, additional, Gu, Shen, additional, Gupta, Isha, additional, Hagerman, Randi J., additional, Hall, Judith G., additional, Hoover-Fong, Julie, additional, Hudgins, Louanne, additional, Iverson, Ayuko, additional, Jabs, Ethylin Wang, additional, James, Cynthia A., additional, Jari, Shama, additional, Keppler-Noreuil, Kim M., additional, Kerr, Lynne M., additional, Kimball, Amy, additional, Kline, Antonie D., additional, Kline, Joel N., additional, Kritzer, Amy, additional, Lambert, Michele P., additional, Lew, Cheryl D., additional, Li, Shao-Tzu, additional, MacCarrick, Gretchen, additional, Matalon, Dena R., additional, McDonald-McGinn, Donna M., additional, McMahon, Francis J., additional, Meliambro, Kristin, additional, Moore, Rebekah A., additional, Murray, Brittney, additional, Newcomb, Tara, additional, Ngeow, Joanne, additional, Ogawa, Jessica, additional, Patel, Dhruv K., additional, Pollin, Toni I., additional, Prasun, Pankaj, additional, Puliaiev, Maksym, additional, Pyeritz, Reed E., additional, Rasmussen, Sonja A., additional, Riboldi, Giulietta Maria, additional, Schecter, Scott M., additional, Scheuerle, Angela E., additional, Scott, Stuart A., additional, Shankar, Suma, additional, Slavotinek, Anne, additional, Smith-Hicks, Constance L., additional, Stewart, Rosalyn W., additional, Trandafir, Cristina, additional, Triano, Vivian Narcisa, additional, Vernon, Hilary J., additional, Wasserstein, Melissa P., additional, Webb, Bryn D., additional, Weksberg, Rosanna, additional, and Yuan, Bo, additional
- Published
- 2024
- Full Text
- View/download PDF
4. Neurodevelopmental Cases: Autism Spectrum Disorder
- Author
-
Triano, Vivien Narcisa, primary and Shankar, Suma, additional
- Published
- 2024
- Full Text
- View/download PDF
5. Retinal dystrophies: A look beyond the eyes.
- Author
-
Tang, Vincent Duong, Egense, Alena, Yiu, Glenn, Meyers, Elijah, Moshiri, Ala, and Shankar, Suma P
- Subjects
Alstrom syndrome ,Bardet Biedl syndrome ,Genetic testing ,Genome sequencing ,Refsum disease ,Retinal dystrophies ,Neurosciences ,Clinical Research ,Intellectual and Developmental Disabilities (IDD) ,Rare Diseases ,Brain Disorders ,Eye Disease and Disorders of Vision ,Genetics ,Detection ,screening and diagnosis ,Aetiology ,4.2 Evaluation of markers and technologies ,2.1 Biological and endogenous factors ,Eye - Abstract
PurposeTo illustrate the importance of systemic evaluation in retinal dystrophies through examples of Alstrom syndrome, Bardet Biedl syndrome, and Refsum disease.ObservationsDetailed eye evaluations, including visual acuity, visual field, slit lamp examination, and indirect ophthalmoscopy were performed. Retinal imaging included fundus photography and spectral domain optical coherence tomography (SD-OCT). Functional testing of the retina was done using full field electroretinography (ffERG). In addition, molecular genetic testing was performed using a ciliopathy panel, a retinal dystrophy panel, and whole genome sequencing (WGS).We report three individuals who presented with vision concerns first to ophthalmology, noted to have retinal dystrophy, and then referred to genomic medicine for genetic testing. Additional evaluation led to suspicion of specific groups of systemic disorders and guided appropriate genetic testing. The first individual presented with retinal dystrophy, obesity, and short stature with no reported neurocognitive deficits. Genetic testing included a ciliopathy panel that was negative followed by WGS that identified biallelic variants in ALMS: a novel frame-shift pathogenic variant c.6525dupT (p.Gln2176Serfs*17) and a rare nonsense pathogenic variant c.2035C > T (p.Arg679Ter) consistent with Alstrom syndrome. The second individual presented with retinal dystrophy, central obesity, and mild neurocognitive deficits. A ciliopathy genetic testing panel identified a homozygous pathogenic variant in BBS7: c.389_390del (p.Asn130Thrfs*4), confirming the diagnosis of Bardet Biedl syndrome. The third individual presented with progressive vision loss due to retinitis pigmentosa, anosmia, hearing loss, and shortened metatarsals and digits. Genetic testing identified two variants in PHYH: c.375_375del (p.Glu126Argfs*2) a pathogenic variant and c.536A > G (p.His179Arg), a variant of uncertain significance (VUS), suggestive of Refsum disease. Additional biochemical testing revealed markedly elevated phytanic acid with a low concentration of pristanic acid and normal concentrations of very long-chain fatty acids (C22:0, C24:0, C26:0), a pattern consistent with a diagnosis of Refsum disease.Conclusions and importanceIn individuals who present with retinal dystrophy to ophthalmologists, additional systemic manifestations such as sensorineural hearing loss, anosmia, or polydactyly, should be sought and a positive history or examination finding should prompt an immediate referral to a clinical geneticist for additional evaluation and appropriate genetic testing. This facilitates pre-test genetic counseling and allows for more accurate diagnosis, prognosis, and management of affected individuals along with better recurrence risk estimates for family members. Identification of an underlying etiology also enhances the understanding of the pathophysiology of disease and expands the genotypic and phenotypic spectrum. Ultimately, successful recognition of these diseases facilitates development of targeted therapies and surveillance of affected individuals.
- Published
- 2022
6. Fragile X syndrome in a girl with variant Turner syndrome and an isodicentric X chromosome
- Author
-
Tassanakijpanich, Nattaporn, Wright, Rachel, Tassone, Flora, Shankar, Suma P, and Hagerman, Randi
- Subjects
Biomedical and Clinical Sciences ,Fragile X Syndrome ,Genetics ,Intellectual and Developmental Disabilities (IDD) ,Brain Disorders ,Rare Diseases ,Pediatric ,Mental Health ,2.1 Biological and endogenous factors ,Child ,Chromosome Aberrations ,Chromosome Disorders ,Female ,Fragile X Mental Retardation Protein ,Guanine ,Humans ,Turner Syndrome ,X Chromosome ,Paediatrics ,Developmental paediatrocs ,Clinical Sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Fragile X (FXS) and Turner (TS) syndromes are X-chromosome-associated disorders. Herein, we report the case of a girl in middle childhood with bicuspid aortic valve in infancy, growth failure, global developmental delay (GDD), visual problems, and coexisting attention-deficit/hyperactivity and anxiety disorders. A high-resolution karyotype in 20 cells revealed 46,X,Idic(X)(p11.21)[19]/45,X[1], suggestive of variant TS. Given her atypical phenotype, subsequent DNA testing was performed. Four FMR1 cytosine-guanine-guanine repeats (30, 410, 580 and 800) were identified, confirming the additional FXS diagnosis. This case study highlights the importance of additional genetic testing in individuals with atypical variant TS, such as unexplained GDD and distinct facial characteristics. The additional FXS diagnosis prompted new therapeutic development for the patient to advance precision healthcare.
- Published
- 2022
7. A novel DPH5-related diphthamide-deficiency syndrome causing embryonic lethality or profound neurodevelopmental disorder
- Author
-
Shankar, Suma P, Grimsrud, Kristin, Lanoue, Louise, Egense, Alena, Willis, Brandon, Hörberg, Johanna, AlAbdi, Mayer, Klaus, Ütkür, Koray, Monaghan, Kristin G, Krier, Joel, Stoler, Joan, Alnemer, Maha, Shankar, Prabhu R, Schaffrath, Raffael, Alkuraya, Fowzan S, Brinkmann, Ulrich, Eriksson, Leif A, Lloyd, Kent, Rauen, Katherine A, Network, Undiagnosed Diseases, Acosta, Maria T, Adam, Margaret, Adams, David R, Alvey, Justin, Amendola, Laura, Andrews, Ashley, Ashley, Euan A, Azamian, Mahshid S, Bacino, Carlos A, Bademci, Guney, Balasubramanyam, Ashok, Baldridge, Dustin, Bale, Jim, Bamshad, Michael, Barbouth, Deborah, Bayrak-Toydemir, Pinar, Beck, Anita, Beggs, Alan H, Behrens, Edward, Bejerano, Gill, Bennet, Jimmy, Berg-Rood, Beverly, Bernstein, Jonathan A, Berry, Gerard T, Bican, Anna, Bivona, Stephanie, Blue, Elizabeth, Bohnsack, John, Bonner, Devon, Botto, Lorenzo, Boyd, Brenna, Briere, Lauren C, Brokamp, Elly, Brown, Gabrielle, Burke, Elizabeth A, Burrage, Lindsay C, Butte, Manish J, Byers, Peter, Byrd, William E, Carey, John, Carrasquillo, Olveen, Cassini, Thomas, Chang, Ta Chen Peter, Chanprasert, Sirisak, Chao, Hsiao-Tuan, Clark, Gary D, Coakley, Terra R, Cobban, Laurel A, Cogan, Joy D, Coggins, Matthew, Cole, F Sessions, Colley, Heather A, Cooper, Cynthia M, Cope, Heidi, Craigen, William J, Crouse, Andrew B, Cunningham, Michael, D'Souza, Precilla, Dai, Hongzheng, Dasari, Surendra, Davis, Joie, Dayal, Jyoti G, Deardorff, Matthew, Dell'Angelica, Esteban C, Dipple, Katrina, Doherty, Daniel, Dorrani, Naghmeh, Doss, Argenia L, Douine, Emilie D, Duncan, Laura, Earl, Dawn, Eckstein, David J, Emrick, Lisa T, Eng, Christine M, Esteves, Cecilia, Falk, Marni, Fernandez, Liliana, Fieg, Elizabeth L, and Fisher, Paul G
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Pediatric ,Human Genome ,2.1 Biological and endogenous factors ,Adenosine Diphosphate ,Animals ,Histidine ,Humans ,Methyltransferases ,Mice ,Mice ,Inbred C57BL ,Neurodevelopmental Disorders ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Syndrome ,Nonverbal neurodevelopment delays ,Novel gene discovery ,Precision animal modeling ,Precision genomics ,Translational genetics ,Undiagnosed Diseases Network ,Clinical Sciences ,Genetics & Heredity - Abstract
PurposeDiphthamide is a post-translationally modified histidine essential for messenger RNA translation and ribosomal protein synthesis. We present evidence for DPH5 as a novel cause of embryonic lethality and profound neurodevelopmental delays (NDDs).MethodsMolecular testing was performed using exome or genome sequencing. A targeted Dph5 knockin mouse (C57BL/6Ncrl-Dph5em1Mbp/Mmucd) was created for a DPH5 p.His260Arg homozygous variant identified in 1 family. Adenosine diphosphate-ribosylation assays in DPH5-knockout human and yeast cells and in silico modeling were performed for the identified DPH5 potential pathogenic variants.ResultsDPH5 variants p.His260Arg (homozygous), p.Asn110Ser and p.Arg207Ter (heterozygous), and p.Asn174LysfsTer10 (homozygous) were identified in 3 unrelated families with distinct overlapping craniofacial features, profound NDDs, multisystem abnormalities, and miscarriages. Dph5 p.His260Arg homozygous knockin was embryonically lethal with only 1 subviable mouse exhibiting impaired growth, craniofacial dysmorphology, and multisystem dysfunction recapitulating the human phenotype. Adenosine diphosphate-ribosylation assays showed absent to decreased function in DPH5-knockout human and yeast cells. In silico modeling of the variants showed altered DPH5 structure and disruption of its interaction with eEF2.ConclusionWe provide strong clinical, biochemical, and functional evidence for DPH5 as a novel cause of embryonic lethality or profound NDDs with multisystem involvement and expand diphthamide-deficiency syndromes and ribosomopathies.
- Published
- 2022
8. The seventh international RASopathies symposium: Pathways to a cure—expanding knowledge, enhancing research, and therapeutic discovery
- Author
-
Kontaridis, Maria I, Roberts, Amy E, Schill, Lisa, Schoyer, Lisa, Stronach, Beth, Andelfinger, Gregor, Aoki, Yoko, Axelrad, Marni E, Bakker, Annette, Bennett, Anton M, Broniscer, Alberto, Castel, Pau, Chang, Caitlin A, Cyganek, Lukas, Das, Tirtha K, Hertog, Jeroen, Galperin, Emilia, Garg, Shruti, Gelb, Bruce D, Gordon, Kristiana, Green, Tamar, Gripp, Karen W, Itkin, Maxim, Kiuru, Maija, Korf, Bruce R, Livingstone, Jeff R, López‐Juárez, Alejandro, Magoulas, Pilar L, Mansour, Sahar, Milner, Theresa, Parker, Elisabeth, Pierpont, Elizabeth I, Plouffe, Kevin, Rauen, Katherine A, Shankar, Suma P, Smith, Shane B, Stevenson, David A, Tartaglia, Marco, Van, Richard, Wagner, Morgan E, Ware, Stephanie M, and Zenker, Martin
- Subjects
Pediatric ,Genetics ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being ,Costello Syndrome ,Humans ,Mitogen-Activated Protein Kinases ,Noonan Syndrome ,Signal Transduction ,ras Proteins ,cardiofaciocutaneus syndrome ,Costello syndrome ,neurofibromatosis ,Noonan syndrome ,RASopathy ,signaling ,Clinical Sciences - Abstract
RASopathies are a group of genetic disorders that are caused by genes that affect the canonical Ras/mitogen-activated protein kinase (MAPK) signaling pathway. Despite tremendous progress in understanding the molecular consequences of these genetic anomalies, little movement has been made in translating these findings to the clinic. This year, the seventh International RASopathies Symposium focused on expanding the research knowledge that we have gained over the years to enhance new discoveries in the field, ones that we hope can lead to effective therapeutic treatments. Indeed, for the first time, research efforts are finally being translated to the clinic, with compassionate use of Ras/MAPK pathway inhibitors for the treatment of RASopathies. This biannual meeting, organized by the RASopathies Network, brought together basic scientists, clinicians, clinician scientists, patients, advocates, and their families, as well as representatives from pharmaceutical companies and the National Institutes of Health. A history of RASopathy gene discovery, identification of new disease genes, and the latest research, both at the bench and in the clinic, were discussed.
- Published
- 2022
9. Quantitative brain MRI morphology in severe and attenuated forms of mucopolysaccharidosis type I
- Author
-
Kovac, Victor, Shapiro, Elsa G, Rudser, Kyle D, Mueller, Bryon A, Eisengart, Julie B, Delaney, Kathleen A, Ahmed, Alia, King, Kelly E, Yund, Brianna D, Cowan, Morton J, Raiman, Julian, Mamak, Eva G, Harmatz, Paul R, Shankar, Suma P, Ali, Nadia, Cagle, Stephanie R, Wozniak, Jeffrey R, Lim, Kelvin O, Orchard, Paul J, Whitley, Chester B, and Nestrasil, Igor
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Mucopolysaccharidoses (MPS) ,Neurosciences ,Clinical Research ,Brain Disorders ,Rare Diseases ,Biomedical Imaging ,Neurological ,Brain ,Humans ,Magnetic Resonance Imaging ,Mucopolysaccharidosis I ,Neuroimaging ,White Matter ,Mucopolysaccharidosis ,Hurler Scheie syndrome ,Brain MRI ,Quantitative brain volumetry ,Ventricle ,Cortex ,Genetics & Heredity ,Genetics ,Clinical sciences - Abstract
ObjectiveTo assess our hypothesis that brain macrostructure is different in individuals with mucopolysaccharidosis type I (MPS I) and healthy controls (HC), we conducted a comprehensive multicenter study using a uniform quantitative magnetic resonance imaging (qMRI) protocol, with analyses that account for the effects of disease phenotype, age, and cognition.MethodsBrain MRIs in 23 individuals with attenuated (MPS IA) and 38 with severe MPS I (MPS IH), aged 4-25 years, enrolled under the study protocol NCT01870375, were compared to 98 healthy controls.ResultsCortical and subcortical gray matter, white matter, corpus callosum, ventricular and choroid plexus volumes in MPS I significantly differed from HC. Thicker cortex, lower white matter and corpus callosum volumes were already present at the youngest MPS I participants aged 4-5 years. Age-related differences were observed in both MPS I groups, but most markedly in MPS IH, particularly in cortical gray matter metrics. IQ scores were inversely associated with ventricular volume in both MPS I groups and were positively associated with cortical thickness only in MPS IA.ConclusionsQuantitatively-derived MRI measures distinguished MPS I participants from HC as well as severe from attenuated forms. Age-related neurodevelopmental trajectories in both MPS I forms differed from HC. The extent to which brain structure is altered by disease, potentially spared by treatment, and how it relates to neurocognitive dysfunction needs further exploration.
- Published
- 2022
10. Clinical outcomes after 4.5 years of eliglustat therapy for Gaucher disease type 1: Phase 3 ENGAGE trial final results
- Author
-
Mistry, Pramod K, Lukina, Elena, Turkia, Hadhami Ben, Shankar, Suma P, Feldman, Hagit, Ghosn, Marwan, Mehta, Atul, Packman, Seymour, Lau, Heather, Petakov, Milan, Assouline, Sarit, Balwani, Manisha, Danda, Sumita, Hadjiev, Evgueniy, Ortega, Andres, Foster, Meredith C, Gaemers, Sebastiaan JM, and Peterschmitt, M Judith
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Hematology ,Clinical Research ,Rare Diseases ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Adult ,Double-Blind Method ,Enzyme Inhibitors ,Female ,Gaucher Disease ,Humans ,Liver ,Male ,Organ Size ,Placebo Effect ,Pyrrolidines ,Spleen ,Treatment Outcome ,Young Adult ,Cardiorespiratory Medicine and Haematology ,Immunology ,Cardiovascular medicine and haematology - Abstract
Eliglustat, an oral substrate reduction therapy, is approved for eligible adults with Gaucher disease type 1. In the Phase 3 ENGAGE trial of previously untreated adults with Gaucher disease type 1, eliglustat-treated patients had statistically significant improvements in organ volumes and hematologic parameters compared with placebo in the 9-month primary analysis. We report final outcomes by time on eliglustat among all patients who participated in the ENGAGE trial and extension. No patient deteriorated clinically or withdrew due to adverse events; 39/40 patients entered the open-label extension period and 34/40 (85%) remained in the trial until completion or switching to commercial eliglustat after its approval (2.3-6 years). Clinically meaningful improvements in Gaucher disease manifestations were seen in all patients concomitant with reductions in pathological lipid substrate levels (glucosylceramide and glucosylsphingosine). Among patients with 4.5 years of eliglustat exposure, mean spleen volume decreased by 66% (from 17.1 to 5.8 multiples of normal [MN], n = 13), mean liver volume decreased by 23% (from 1.5 to 1.1 MN, n = 13), mean hemoglobin increased 1.4 g/dl (from 11.9 to 13.4 g/dl, n = 12), mean platelet count increased by 87% (from 67.6 to 122.6 × 109 /L, n = 12), median chitotriosidase decreased by 82% (from 13 394 to 2312 nmol/h/ml, n = 11), median glucosylceramide decreased by 79% (from 11.5 to 2.4 μg/ml, n = 11), median glucosylsphingosine decreased by 84% (from 518.5 to 72.1 ng/ml, n = 10), and mean spine T-score increased from -1.07 (osteopenia) to -0.53 (normal) (n = 9). The magnitude of improvement in Gaucher disease manifestations and biomarkers over time was similar among the full trial cohort. Eliglustat was well-tolerated and led to clinically significant improvements in previously untreated patients with Gaucher disease type 1 during 4.5 years of treatment.
- Published
- 2021
11. Retinal degeneration in mice and humans with neuronal ceroid lipofuscinosis type 8
- Author
-
Salpeter, Elyse M, Leonard, Brian C, Lopez, Antonio J, Murphy, Christopher J, Thomasy, Sara, Imai, Denise M, Grimsrud, Kristin, Lloyd, KC Kent, Yan, Jiong, Russo, Rossana Sanchez, Shankar, Suma P, and Moshiri, Ala
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Neurodegenerative ,Neurosciences ,Eye Disease and Disorders of Vision ,Rare Diseases ,Eye ,Neuronal ceroid lipofuscinosis ,retinal degeneration ,murine model ,Biomedical and clinical sciences ,Health sciences - Abstract
BackgroundCeroid lipofuscinosis type 8 belongs to a heterogenous group of vision and life-threatening neurodegenerative diseases, neuronal ceroid lipofuscinosis (NCL). Effective therapy is limited to a single drug for treatment of ceroid lipofuscinosis type 2, necessitating animal disease models to facilitate further therapeutic development. Murine models are advantageous for therapeutic development due to easy genetic manipulation and rapid breeding, however appropriate genetic models need to be identified and characterized before being used for therapy testing. To date, murine models of ocular disease associated with ceroid lipofuscinosis type 8 have only been characterized in motor neuron degeneration mice.MethodsCln8-/- mice were produced by CRISPR/Cas9 genome editing through the International Mouse Phenotyping Consortium. Ophthalmic examination, optical coherence tomography, electroretinography, and ocular histology was performed on Cln8-/- mice and controls at 16 weeks of age. Quantification of all retinal layers, retinal pigmented epithelium, and the choriocapillaris was performed using images acquired with ocular coherence tomography and planimetry of histologic sections. Necropsy was performed to investigate concurrent systemic abnormalities. Clinical correlation with human patients with CLN8-associated retinopathy is provided.ResultsRetinal degeneration characterized by retinal pigment epithelium mottling, scattered drusen, and retinal vascular attenuation was noted in all Cln8-/- mice. Loss of inner and outer photoreceptor segment demarcation was noted on optical coherence tomography, with significant thinning of the whole retina (P=1e-9), outer nuclear layer (P=1e-9), and combined photoreceptor segments (P=1e-9). A global reduction in scotopic and photopic electroretinographic waveforms was noted in all Cln8-/- mice. Slight thickening of the inner plexiform layer (P=0.02) and inner nuclear layer (P=0.004), with significant thinning of the whole retina (P=0.03), outer nuclear layer (P=0.01), and outer photoreceptor segments (P=0.001) was appreciated on histologic sections. Scattered lipid vacuoles were noted in splenic red pulp of all Cln8-/- mice, though no gross systemic abnormalities were detected on necropsy. Retinal findings are consistent with those seen in patients with ceroid lipofuscinosis type 8.ConclusionsThis study provides detailed clinical characterization of retinopathy in adult Cln8-/- mice. Findings suggest that Cln8-/- mice may provide a useful murine model for development of novel therapeutics needed for treating ocular disease in patients with ceroid lipofuscinosis type 8.
- Published
- 2021
12. Project Baby Bear: Rapid precision care incorporating rWGS in 5 California children’s hospitals demonstrates improved clinical outcomes and reduced costs of care
- Author
-
Dimmock, David, Caylor, Sara, Waldman, Bryce, Benson, Wendy, Ashburner, Christina, Carmichael, Jason L, Carroll, Jeanne, Cham, Elaine, Chowdhury, Shimul, Cleary, John, D'Harlingue, Arthur, Doshi, A, Ellsworth, Katarzyna, Galarreta, Carolina I, Hobbs, Charlotte, Houtchens, Kathleen, Hunt, Juliette, Joe, Priscilla, Joseph, Maries, Kaplan, Robert H, Kingsmore, Stephen F, Knight, Jason, Kochhar, Aaina, Kronick, Richard G, Limon, Jolie, Martin, Madelena, Rauen, Katherine A, Schwarz, Adam, Shankar, Suma P, Spicer, Rosanna, Rojas, Mario Augusto, Vargas-Shiraishi, Ofelia, Wigby, Kristen, Zadeh, Neda, and Farnaes, Lauge
- Subjects
Comparative Effectiveness Research ,Pediatric ,Clinical Research ,Health Services ,Clinical Trials and Supportive Activities ,Good Health and Well Being ,California ,Cohort Studies ,Cost of Illness ,Critical Care ,Critical Illness ,Female ,Hospitals ,Pediatric ,Humans ,Infant ,Infant ,Newborn ,Male ,Medicaid ,Precision Medicine ,Prospective Studies ,Treatment Outcome ,United States ,Whole Genome Sequencing ,MediCal ,QUALY ,comparative effectiveness research ,critical care ,genetic disease ,health outcomes research ,neonatal intensive care ,pediatrics ,quality improvement ,quality-adjusted life years ,rare disease ,real-world care ,Biological Sciences ,Medical and Health Sciences ,Genetics & Heredity - Abstract
Genetic disorders are a leading contributor to mortality in neonatal and pediatric intensive care units (ICUs). Rapid whole-genome sequencing (rWGS)-based rapid precision medicine (RPM) is an intervention that has demonstrated improved clinical outcomes and reduced costs of care. However, the feasibility of broad clinical deployment has not been established. The objective of this study was to implement RPM based on rWGS and evaluate the clinical and economic impact of this implementation as a first line diagnostic test in the California Medicaid (Medi-Cal) program. Project Baby Bear was a payor funded, prospective, real-world quality improvement project in the regional ICUs of five tertiary care children's hospitals. Participation was limited to acutely ill Medi-Cal beneficiaries who were admitted November 2018 to May 2020, were
- Published
- 2021
13. The evolving role of genetics in ophthalmology.
- Author
-
Couser, Natario, Brooks, Brian, Drack, Arlene, and Shankar, Suma
- Subjects
Inherited retinal disease ,Ophthalmology subspecialist ,gene therapy ,ocular genetics training program ,ophthalmic genetics fellowship ,Eye Diseases ,Genomics ,Humans ,Ophthalmology - Abstract
Advances in molecular genetics over the past three decades have helped identify a substantial number of genetic variants causing inherited eye diseases that can be identified rapidly by appropriate genetic tests in a clinically useful window. With this progression of knowledge, the roles of genetics and ophthalmology in patient care have become increasingly intertwined, and the necessity for subspecialists in the field of ophthalmic genetics is of paramount importance. As a result of continual medical specialization, technological progress in genetics and knowledge garnered by over a century and a half of cataloguing eye pathology, ophthalmic genetics has become an emerging subspecialty within ophthalmology. By virtue of its rapidly changing advances, genetics and genomics serves a large role within ophthalmology, and subspecialists with the same level of detailed and broad knowledge as any other ophthalmology subspecialty are now required in order to meet the growing needs of the expanding population.
- Published
- 2021
14. Costello syndrome: Clinical phenotype, genotype, and management guidelines.
- Author
-
Gripp, Karen W, Morse, Lindsey A, Axelrad, Marni, Chatfield, Kathryn C, Chidekel, Aaron, Dobyns, William, Doyle, Daniel, Kerr, Bronwyn, Lin, Angela E, Schwartz, David D, Sibbles, Barbara J, Siegel, Dawn, Shankar, Suma P, Stevenson, David A, Thacker, Mihir M, Weaver, K Nicole, White, Sue M, and Rauen, Katherine A
- Subjects
Face ,Heart ,Humans ,Heart Defects ,Congenital ,Abnormalities ,Multiple ,Developmental Disabilities ,Gene Expression Regulation ,Genotype ,Phenotype ,Germ-Line Mutation ,Disease Management ,Proto-Oncogene Proteins p21(ras) ,Guidelines as Topic ,Costello Syndrome ,Costello syndrome ,HRAS mutation ,RAS/MAPK ,RASopathy ,management guidelines ,Genetics ,Pediatric ,Congenital Structural Anomalies ,Health Services ,Clinical Research ,Management of diseases and conditions ,7.3 Management and decision making ,Good Health and Well Being ,RAS ,MAPK ,Clinical Sciences - Abstract
Costello syndrome (CS) is a RASopathy caused by activating germline mutations in HRAS. Due to ubiquitous HRAS gene expression, CS affects multiple organ systems and individuals are predisposed to cancer. Individuals with CS may have distinctive craniofacial features, cardiac anomalies, growth and developmental delays, as well as dermatological, orthopedic, ocular, and neurological issues; however, considerable overlap with other RASopathies exists. Medical evaluation requires an understanding of the multifaceted phenotype. Subspecialists may have limited experience in caring for these individuals because of the rarity of CS. Furthermore, the phenotypic presentation may vary with the underlying genotype. These guidelines were developed by an interdisciplinary team of experts in order to encourage timely health care practices and provide medical management guidelines for the primary and specialty care provider, as well as for the families and affected individuals across their lifespan. These guidelines are based on expert opinion and do not represent evidence-based guidelines due to the lack of data for this rare condition.
- Published
- 2019
15. First International Conference on RASopathies and Neurofibromatoses in Asia: Identification and advances of new therapeutics
- Author
-
Rauen, Katherine A, Alsaegh, Abeer, Ben‐Shachar, Shay, Berman, Yemima, Blakeley, Jaishri, Cordeiro, Isabel, Elgersma, Ype, Evans, D Gareth, Fisher, Michael J, Frayling, Ian M, George, Joshi, Huson, Susan M, Kerr, Bronwyn, Khire, Uday, Korf, Bruce, Legius, Eric, Messiaen, Ludwine, van Minkelen, Rick, Nampoothiri, Sheela, Ngeow, Joanne, Parada, Luis F, Phadke, Shubha, Pillai, Ashok, Plotkin, Scott R, Puri, Ratna, Raji, Anup, Ramesh, Vijaya, Ratner, Nancy, Shankar, Suma P, Sharda, Sheetal, Tambe, Anant, Vikkula, Miikka, Widemann, Brigitte C, Wolkenstein, Pierre, and Upadhyaya, Meena
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Neurofibromatosis ,Neurosciences ,Brain Disorders ,Intellectual and Developmental Disabilities (IDD) ,Rare Diseases ,Pediatric ,Congenital ,Biomarkers ,Disease Management ,Genetic Association Studies ,Genetic Predisposition to Disease ,Humans ,Mitogen-Activated Protein Kinases ,Molecular Diagnostic Techniques ,Molecular Targeted Therapy ,Neurofibromatoses ,Signal Transduction ,Translational Research ,Biomedical ,ras Proteins ,clinical trial ,neurofibromatoses ,RASopathy ,signal transduction pathway ,therapy ,Clinical Sciences ,Clinical sciences - Abstract
The neurofibromatoses, which include neurofibromatosis type I (NF1), neurofibromatosis type II (NF2), and schwannomatosis, are a group of syndromes characterized by tumor growth in the nervous system. The RASopathies are a group of syndromes caused by germline mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. The RASopathies include NF1, Noonan syndrome, Noonan syndrome with multiple lentigines, Costello syndrome, cardio-facio-cutaneous syndrome, Legius syndrome, capillary malformation arterio-venous malformation syndrome, and SYNGAP1 autism. Due to their common underlying pathogenetic etiology, all these syndromes have significant phenotypic overlap of which one common feature include a predisposition to tumors, which may be benign or malignant. Together as a group, they represent one of the most common multiple congenital anomaly syndromes estimating to affect approximately one in 1000 individuals worldwide. The subcontinent of India represents one of the largest populations in the world, yet remains underserved from an aspect of clinical genetics services. In an effort to bridge this gap, the First International Conference on RASopathies and Neurofibromatoses in Asia: Identification and Advances of New Therapeutics was held in Kochi, Kerala, India. These proceedings chronicle this timely and topical international symposium directed at discussing the best practices and therapies for individuals with neurofibromatoses and RASopathies.
- Published
- 2019
16. Proceedings of the fifth international RASopathies symposium: When development and cancer intersect
- Author
-
Rauen, Katherine A, Schoyer, Lisa, Schill, Lisa, Stronach, Beth, Albeck, John, Andresen, Brage S, Cavé, Hélène, Ellis, Michelle, Fruchtman, Steven M, Gelb, Bruce D, Gibson, Christopher C, Gripp, Karen, Hefner, Erin, Huang, William YC, Itkin, Maxim, Kerr, Bronwyn, Linardic, Corinne M, McMahon, Martin, Oberlander, Beverly, Perlstein, Ethan, Ratner, Nancy, Rogers, Leslie, Schenck, Annette, Shankar, Suma, Shvartsman, Stanislav, Stevenson, David A, Stites, Edward C, Stork, Philip JS, Sun, Cheng, Therrien, Marc, Ullian, Erik M, Widemann, Brigitte C, Yeh, Erika, Zampino, Giuseppe, Zenker, Martin, Timmer, William, and McCormick, Frank
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Rare Diseases ,Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Gene Expression Regulation ,Genetic Association Studies ,Genetic Predisposition to Disease ,Human Development ,Humans ,Models ,Biological ,Molecular Targeted Therapy ,Neoplasms ,Organogenesis ,Signal Transduction ,Syndrome ,ras Proteins ,cardio-facio-cutaneous syndrome ,clinical trial ,Costello syndrome ,Legius syndrome ,neurofibromatosis type 1 ,Noonan syndrome ,RAS/MAPK ,RASopathies ,signal transduction pathway ,therapy ,Genetics ,Clinical Sciences ,Clinical sciences - Abstract
This report summarizes and highlights the fifth International RASopathies Symposium: When Development and Cancer Intersect, held in Orlando, Florida in July 2017. The RASopathies comprise a recognizable pattern of malformation syndromes that are caused by germ line mutations in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) pathway. Because of their common underlying pathogenetic etiology, there is significant overlap in their phenotypic features, which includes craniofacial dysmorphology, cardiac, cutaneous, musculoskeletal, gastrointestinal and ocular abnormalities, neurological and neurocognitive issues, and a predisposition to cancer. The RAS pathway is a well-known oncogenic pathway that is commonly found to be activated in somatic malignancies. As in somatic cancers, the RASopathies can be caused by various pathogenetic mechanisms that ultimately impact or alter the normal function and regulation of the MAPK pathway. As such, the RASopathies represent an excellent model of study to explore the intersection of the effects of dysregulation and its consequence in both development and oncogenesis.
- Published
- 2018
17. Migalastat improves diarrhea in patients with Fabry disease: clinical-biomarker correlations from the phase 3 FACETS trial
- Author
-
Schiffmann, Raphael, Bichet, Daniel G, Jovanovic, Ana, Hughes, Derralynn A, Giugliani, Roberto, Feldt-Rasmussen, Ulla, Shankar, Suma P, Barisoni, Laura, Colvin, Robert B, Jennette, J Charles, Holdbrook, Fred, Mulberg, Andrew, Castelli, Jeffrey P, Skuban, Nina, Barth, Jay A, and Nicholls, Kathleen
- Subjects
Rare Diseases ,Neurodegenerative ,Clinical Research ,Brain Disorders ,Digestive Diseases ,Clinical Trials and Supportive Activities ,Emerging Infectious Diseases ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,1-Deoxynojirimycin ,Adolescent ,Adult ,Aged ,Biomarkers ,Diarrhea ,Fabry Disease ,Female ,Humans ,Kidney ,Male ,Middle Aged ,Mutation ,Trihexosylceramides ,Young Adult ,Amenable mutation ,Fabry disease ,GSRS ,Gastrointestinal ,Globotriaosylceramide ,Lyso-Gb3 ,Migalastat ,Pharmacological chaperone ,Other Medical and Health Sciences ,Genetics & Heredity - Abstract
BackgroundFabry disease is frequently characterized by gastrointestinal symptoms, including diarrhea. Migalastat is an orally-administered small molecule approved to treat the symptoms of Fabry disease in patients with amenable mutations.MethodsWe evaluated minimal clinically important differences (MCID) in diarrhea based on the corresponding domain of the patient-reported Gastrointestinal Symptom Rating Scale (GSRS) in patients with Fabry disease and amenable mutations (N = 50) treated with migalastat 150 mg every other day or placebo during the phase 3 FACETS trial (NCT00925301).ResultsAfter 6 months, significantly more patients receiving migalastat versus placebo experienced improvement in diarrhea based on a MCID of 0.33 (43% vs 11%; p = .02), including the subset with baseline diarrhea (71% vs 20%; p = .02). A decline in kidney peritubular capillary globotriaosylceramide inclusions correlated with diarrhea improvement; patients with a reduction > 0.1 were 5.6 times more likely to have an improvement in diarrhea than those without (p = .031).ConclusionsMigalastat was associated with a clinically meaningful improvement in diarrhea in patients with Fabry disease and amenable mutations. Reductions in kidney globotriaosylceramide may be a useful surrogate endpoint to predict clinical benefit with migalastat in patients with Fabry disease.Trial registrationNCT00925301 ; June 19, 2009.
- Published
- 2018
18. Phenotypic characteristics of the p.Asn215Ser (p.N215S) GLA mutation in male and female patients with Fabry disease: A multicenter Fabry Registry study
- Author
-
Germain, Dominique P, Brand, Eva, Burlina, Alessandro, Cecchi, Franco, Garman, Scott C, Kempf, Judy, Laney, Dawn A, Linhart, Aleš, Maródi, László, Nicholls, Kathy, Ortiz, Alberto, Pieruzzi, Federico, Shankar, Suma P, Waldek, Stephen, Wanner, Christoph, and Jovanovic, Ana
- Subjects
Clinical Research ,Neurodegenerative ,Cardiovascular ,Pediatric ,Aging ,Rare Diseases ,Heart Disease ,GLA ,Fabry disease ,cardiac variant ,p.Asn215Ser ,p.N215S ,phenotype ,Medicinal and Biomolecular Chemistry ,Genetics ,Clinical Sciences - Abstract
The p.Asn215Ser or p.N215S GLA variant has been associated with late-onset cardiac variant of Fabry disease. To expand on the scarce phenotype data, we analyzed natural history data from 125 p.N215S patients (66 females, 59 males) enrolled in the Fabry Registry (NCT00196742) and compared it with data from 401 patients (237 females, 164 males) harboring mutations associated with classic Fabry disease. We evaluated interventricular septum thickness (IVST), left ventricular posterior wall thickness (LVPWT), estimated glomerular filtration rate and severe clinical events. In p.N215S males, mildly abnormal mean IVST and LVPWT values were observed in patients aged 25-34 years, and values gradually increased with advancing age. Mean values were similar to those of classic males. In p.N215S females, these abnormalities occurred primarily in patients aged 55-64 years. Severe clinical events in p.N215S patients were mainly cardiac (males 31%, females 8%) while renal and cerebrovascular events were rare. Renal impairment occurred in 17% of p.N215S males (mostly in patients aged 65-74 years), and rarely in females (3%). p.N215S is a disease-causing mutation with severe clinical manifestations found primarily in the heart. Cardiac involvement may become as severe as in classic Fabry patients, especially in males.
- Published
- 2018
19. Retinopathy and optic atrophy: Expanding the phenotypic spectrum of pathogenic variants in the AARS2 gene
- Author
-
Peragallo, Jason H, Keller, Stephanie, van der Knaap, Marjo S, Soares, Bruno P, and Shankar, Suma P
- Subjects
Clinical Research ,Eye Disease and Disorders of Vision ,Brain Disorders ,Genetics ,Neurosciences ,Rare Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Eye ,Neurological ,Alanine-tRNA Ligase ,Child ,Preschool ,Genetic Testing ,Humans ,Leukoencephalopathies ,Magnetic Resonance Imaging ,Male ,Mutation ,Missense ,Optic Atrophy ,Phenotype ,Retinal Diseases ,Visual Acuity ,Whole Exome Sequencing ,Exome Sequencing ,AARS2 ,leukodystrophy ,optic atrophy ,retinopathy ,Opthalmology and Optometry ,Ophthalmology & Optometry - Abstract
BackgroundOptic atrophy may be the sequela of optic nerve injury due to any insult, including isolated and syndromic genetic diseases. Alanyl-tRNA synthetase 2 (AARS2) pathogenic variants have been reported to cause leukodystrophy with ovarian failure, and cardiomyopathy (#615889) as well as combined oxidative phosphorylation deficiency-8 (#614096). We report a young child who presented with decreased vision due to optic atrophy and was found to harbor missense variants in the AARS2 gene expanding the phenotypic expression of the AARS2 gene.Materials and methodsSingle observational case report with genetic testing, laboratory testing, neurologic and ophthalmic clinical examinations, and neuroimaging performed at a tertiary academic medical center.ResultsAn 18-month old Korean boy was noted to have a progressive decline in visual function. The physical exam revealed bilateral optic atrophy, peripheral retinal bone spicule pigmentation, and absent patellar reflexes. Electromyography was consistent with demyelinating polyneuropathy. Magnetic resonance imaging (MRI) of the brain and spine showed cerebellar and supratentorial white matter multifocal changes with areas of restricted diffusion, and dorsal column signal abnormalities. Whole exome sequencing revealed two missense variants in the AARS2 gene [c.1519G>C (p.V507L) and c.2165G>A (p.R722Q)], found to be in trans on parental testing.ConclusionsMissense variants in the AARS2 gene are the likely cause of the retinopathy and optic atrophy in this patient. This finding expands the phenotypic spectrum of the AARS2 gene.
- Published
- 2018
20. Genetic testing for inherited colorectal cancer and polyposis, 2021 revision: a technical standard of the American College of Medical Genetics and Genomics (ACMG)
- Author
-
Mao, Rong, Krautscheid, Patti, Graham, Rondell P., Ganguly, Arupa, Shankar, Suma, Ferber, Matthew, and Hegde, Madhuri
- Published
- 2021
- Full Text
- View/download PDF
21. Outcomes after 18 months of eliglustat therapy in treatment‐naïve adults with Gaucher disease type 1: The phase 3 ENGAGE trial
- Author
-
Mistry, Pramod K, Lukina, Elena, Turkia, Hadhami Ben, Shankar, Suma P, Baris, Hagit, Ghosn, Marwan, Mehta, Atul, Packman, Seymour, Pastores, Gregory, Petakov, Milan, Assouline, Sarit, Balwani, Manisha, Danda, Sumita, Hadjiev, Evgueniy, Ortega, Andres, Gaemers, Sebastiaan JM, Tayag, Regina, and Peterschmitt, M Judith
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Clinical Research ,Clinical Trials and Supportive Activities ,Hematology ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Enzyme Inhibitors ,Enzyme Replacement Therapy ,Follow-Up Studies ,Gaucher Disease ,Glucosylceramidase ,Humans ,Liver ,Organ Size ,Pyrrolidines ,Spleen ,Treatment Outcome ,Cardiorespiratory Medicine and Haematology ,Immunology ,Cardiovascular medicine and haematology - Abstract
Eliglustat, an oral substrate reduction therapy, is a first-line treatment for adults with Gaucher disease type 1 (GD1) who are poor, intermediate, or extensive CYP2D6 metabolizers (>90% of patients). In the primary analysis of the Phase 3 ENGAGE trial (NCT00891202), eliglustat treatment for 9 months resulted in significant reductions in spleen and liver volumes and increases in hemoglobin concentration and platelet count compared with placebo. We report 18-month outcomes of patients who entered the trial extension period, in which all patients received eliglustat. Of 40 trial patients, 39 entered the extension period, and 38 completed 18 months. Absolute values and percent change over time were determined for spleen and liver volume, hemoglobin concentration, platelet count, bone mineral density, bone marrow burden, and Gaucher disease biomarkers. For patients randomized to eliglustat in the double-blind period, continuing treatment with eliglustat for 9 more months resulted in incremental improvement of all disease parameters. For patients randomized to placebo in the double-blind period, eliglustat treatment during the 9-month, open-label period resulted in significant decrease of spleen and liver volumes and significant increase of hemoglobin and platelets, with a similar rate of change to patients who had received eliglustat in the double-blind period. Eliglustat treatment was also associated with improvement in bone marrow burden score, bone mineral density, and established biomarkers of Gaucher disease, including reduction of the bioactive lipid, glucosylsphingosine. These findings underscore the efficacy of eliglustat in treatment-naïve patients. Eliglustat was well-tolerated, and there were no new safety concerns with longer-term exposure.
- Published
- 2017
22. Oral pharmacological chaperone migalastat compared with enzyme replacement therapy in Fabry disease: 18-month results from the randomised phase III ATTRACT study
- Author
-
Hughes, Derralynn A, Nicholls, Kathleen, Shankar, Suma P, Sunder-Plassmann, Gere, Koeller, David, Nedd, Khan, Vockley, Gerard, Hamazaki, Takashi, Lachmann, Robin, Ohashi, Toya, Olivotto, Iacopo, Sakai, Norio, Deegan, Patrick, Dimmock, David, Eyskens, François, Germain, Dominique P, Goker-Alpan, Ozlem, Hachulla, Eric, Jovanovic, Ana, Lourenco, Charles M, Narita, Ichiei, Thomas, Mark, Wilcox, William R, Bichet, Daniel G, Schiffmann, Raphael, Ludington, Elizabeth, Viereck, Christopher, Kirk, John, Yu, Julie, Johnson, Franklin, Boudes, Pol, Benjamin, Elfrida R, Lockhart, David J, Barlow, Carrolee, Skuban, Nina, Castelli, Jeffrey P, Barth, Jay, and Feldt-Rasmussen, Ulla
- Subjects
Pediatric ,Neurodegenerative ,Clinical Trials and Supportive Activities ,Rare Diseases ,Clinical Research ,Orphan Drug ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,1-Deoxynojirimycin ,Administration ,Oral ,Adolescent ,Adult ,Aged ,Enzyme Replacement Therapy ,Fabry Disease ,Female ,Humans ,Lysosomes ,Male ,Middle Aged ,Molecular Chaperones ,Treatment Outcome ,alpha-Galactosidase ,Fabry disease ,Pharmacological chaperone ,enzyme replacement therapy ,lyso-Gb3 ,lysosomal storage disorder ,Biological Sciences ,Medical and Health Sciences ,Genetics & Heredity - Abstract
BackgroundFabry disease is an X-linked lysosomal storage disorder caused by GLA mutations, resulting in α-galactosidase (α-Gal) deficiency and accumulation of lysosomal substrates. Migalastat, an oral pharmacological chaperone being developed as an alternative to intravenous enzyme replacement therapy (ERT), stabilises specific mutant (amenable) forms of α-Gal to facilitate normal lysosomal trafficking.MethodsThe main objective of the 18-month, randomised, active-controlled ATTRACT study was to assess the effects of migalastat on renal function in patients with Fabry disease previously treated with ERT. Effects on heart, disease substrate, patient-reported outcomes (PROs) and safety were also assessed.ResultsFifty-seven adults (56% female) receiving ERT (88% had multiorgan disease) were randomised (1.5:1), based on a preliminary cell-based assay of responsiveness to migalastat, to receive 18 months open-label migalastat or remain on ERT. Four patients had non-amenable mutant forms of α-Gal based on the validated cell-based assay conducted after treatment initiation and were excluded from primary efficacy analyses only. Migalastat and ERT had similar effects on renal function. Left ventricular mass index decreased significantly with migalastat treatment (-6.6 g/m2 (-11.0 to -2.2)); there was no significant change with ERT. Predefined renal, cardiac or cerebrovascular events occurred in 29% and 44% of patients in the migalastat and ERT groups, respectively. Plasma globotriaosylsphingosine remained low and stable following the switch from ERT to migalastat. PROs were comparable between groups. Migalastat was generally safe and well tolerated.ConclusionsMigalastat offers promise as a first-in-class oral monotherapy alternative treatment to intravenous ERT for patients with Fabry disease and amenable mutations.Trial registration numberNCT00925301; Pre-results.
- Published
- 2017
23. Atypical presentation of neuronal ceroid lipofuscinosis type 8 in a sibling pair and review of the eye findings and neurological features.
- Author
-
Sanchez, Rossana L, Yan, Jiong, Richards, Sarah, Mierau, Gary, Wartchow, Eric P, Collins, Christin D, and Shankar, Suma P
- Subjects
Chromosome microarray ,Epilepsy ,Lysosomal storage disorder ,Neuronal ceroid lipofuscinosis ,Next generation sequencing panel ,Vision loss - Abstract
Purpose:To report atypical presentation of neuronal ceroid lipofuscinoses type 8 (CLN8) to the eye clinic and review clinical features of CLN8. Observations:Detailed eye exam by slit lamp exam, indirect ophthalmoscopy, fundus photography, optical coherence tomography, visual fields and electroretinogram (ERG). Molecular genetic testing using Next Generation Sequencing panel (NGS) and array Comparative Genomic Hybridization (aCGH).The siblings in this study presented to the eye clinic with retinitis pigmentosa and cystoid macular edema, and a history of seizures but no severe neurocognitive deficits or regression. Genetic testing identified a c.200C > T (p.A67V) variant in the CLN8 gene and a deletion encompassing the entire gene. Electron microscopy of lymphocytes revealed fingerprint inclusions in both siblings. Conclusions:and Importance: Pathogenic variants in CLN8 account for the retinitis pigmentosa and seizures in our patients however, currently, they do not have regression or neurocognitive decline. The presentation of NCL can be very diverse and it is important for ophthalmologists to consider this in the differential diagnosis of retinal disorders with seizures or other neurological features. Molecular genetic testing of multiple genes causing isolated and syndromic eye disorders using NGS panels and aCGH along with additional complementary testing may often be required to arrive at a definitive diagnosis.
- Published
- 2016
24. Enzyme replacement therapy with taliglucerase alfa: 36‐month safety and efficacy results in adult patients with Gaucher disease previously treated with imiglucerase
- Author
-
Pastores, Gregory M, Shankar, Suma P, Petakov, Milan, Giraldo, Pilar, Rosenbaum, Hanna, Amato, Dominick J, Szer, Jeffrey, Chertkoff, Raul, Brill‐Almon, Einat, and Zimran, Ari
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Clinical Research ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Adult ,Aged ,Chemokines ,CC ,Drug Substitution ,Enzyme Replacement Therapy ,Female ,Gaucher Disease ,Glucosylceramidase ,Hemoglobins ,Hexosaminidases ,Humans ,Liver ,Male ,Middle Aged ,Organ Size ,Platelet Count ,Spleen ,Treatment Outcome ,Young Adult ,Cardiorespiratory Medicine and Haematology ,Immunology ,Cardiovascular medicine and haematology - Abstract
Taliglucerase alfa is the first available plant cell-expressed human recombinant therapeutic protein. It is indicated for treatment of patients with type 1 Gaucher disease (GD) in adult and pediatric patients in several countries. Study PB-06-002 examined the safety and efficacy of taliglucerase alfa for 9 months in patients who previously received imiglucerase. The results of adult patients from Study PB-06-002 who continued receiving taliglucerase alfa in extension Study PB-06-003 for up to 36 months are reported here. Eighteen patients received at least one dose of taliglucerase alfa in Study PB-06-003; 10 patients completed 36 total months of therapy, and four patients who transitioned to commercial drug completed 30-33 months of treatment. In patients who completed 36 total months of treatment, mean percent (±standard error) changes from baseline/time of switch to taliglucerase alfa to 36 months were as follows: hemoglobin concentration, -1.0% (±1.9%; n = 10); platelet count, +9.3% (±9.8%; n = 10); spleen volume measured in multiples of normal (MN), -19.8% (±9.9%; n = 7); liver volume measured in MN, +0.9% (±5.4%; n = 8); chitotriosidase activity, -51.5% (±8.1%; n = 10); and CCL18 concentration, -36.5 (±8.0%; n = 10). Four patients developed antidrug antibodies, including one with evidence of neutralizing activity in vitro. All treatment-related adverse events were mild or moderate and transient. The 36-month results of switching from imiglucerase to taliglucerase alfa treatment in adults with GD provide further data on the clinical safety and efficacy of taliglucerase alfa beyond the initial 9 months of the original study. www.clinicaltrials.gov identifier NCT00705939. Am. J. Hematol. 91:661-665, 2016. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
25. Autosomal Dominant Retinal Dystrophies Caused by a Founder Splice Site Mutation, c.828+3A>T, in PRPH2 and Protein Haplotypes in trans as ModifiersPRPH2 Retinal Dystrophies
- Author
-
Shankar, Suma P, Hughbanks-Wheaton, Dianna K, Birch, David G, Sullivan, Lori S, Conneely, Karen N, Bowne, Sara J, Stone, Edwin M, and Daiger, Stephen P
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Genetics ,Neurodegenerative ,Eye Disease and Disorders of Vision ,Brain Disorders ,Clinical Research ,Rare Diseases ,Neurosciences ,Aetiology ,2.1 Biological and endogenous factors ,Eye ,Adolescent ,Adult ,Aged ,Aged ,80 and over ,DNA Mutational Analysis ,Dark Adaptation ,Disease Progression ,Electroretinography ,Female ,Founder Effect ,Haplotypes ,Humans ,Male ,Middle Aged ,Mutation ,Pedigree ,Peripherins ,Phenotype ,Polymorphism ,Single-Stranded Conformational ,RNA Splice Sites ,Retina ,Retinal Dystrophies ,Visual Acuity ,Visual Fields ,Young Adult ,Biological Sciences ,Medical and Health Sciences ,Ophthalmology & Optometry ,Ophthalmology and optometry - Abstract
PurposeWe determined the phenotypic variation, disease progression, and potential modifiers of autosomal dominant retinal dystrophies caused by a splice site founder mutation, c.828+3A>T, in the PRPH2 gene.MethodsA total of 62 individuals (19 families) harboring the PRPH2 c.828+3A>T mutation, had phenotype analysis by fundus appearance, electrophysiology, and visual fields. The PRPH2 haplotypes in trans were sequenced for potential modifying variants and generalized estimating equations (GEE) used for statistical analysis.ResultsSeveral distinct phenotypes caused by the PRPH2 c.828+3A>T mutation were observed and fell into two clinical categories: Group I (N = 44) with mild pattern dystrophies (PD) and Group II (N = 18) with more severe cone-rod dystrophy (CRD), retinitis pigmentosa (RP), and central areolar chorioretinal dystrophy (CACD). The PRPH2 Gln304-Lys310-Asp338 protein haplotype in trans was found in Group I only (29.6% vs. 0%), whereas the Glu304-Lys310-Gly338 haplotype was predominant in Group II (94.4% vs. 70.4%). Generalized estimating equations analysis for PD versus the CRD/CACD/RP phenotypes in individuals over 43 years alone with the PRPH2 haplotypes in trans and age as predictors, adjusted for correlation within families, confirmed a significant effect of haplotype on severity (P = 0.03) with an estimated odds ratio of 7.16 (95% confidence interval [CI] = [2.8, 18.4]).ConclusionsThe PRPH2 c.828+3A>T mutation results in multiple distinct phenotypes likely modified by protein haplotypes in trans; the odds of having the CACD/RP-like phenotype (versus the PD phenotype) are 7.16 times greater with a Glu304-Lys310-Gly338 haplotype in trans. Further functional studies of the modifying haplotypes in trans and PRPH2 splice variants may offer therapeutic targets.
- Published
- 2016
26. An open-label clinical trial of agalsidase alfa enzyme replacement therapy in children with Fabry disease who are naïve to enzyme replacement therapy
- Author
-
Goker-Alpan, Ozlem, Longo, Nicola, McDonald, Marie, Shankar, Suma P, Schiffmann, Raphael, Chang, Peter, Shen, Yinghua, and Pano, Arian
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Clinical Research ,Cardiovascular ,Pediatric ,Clinical Trials and Supportive Activities ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,Administration ,Intravenous ,Adolescent ,Child ,Enzyme Replacement Therapy ,Fabry Disease ,Female ,Humans ,Isoenzymes ,Male ,Recombinant Proteins ,alpha-Galactosidase ,Fabry disease ,agalsidase alfa ,efficacy ,enzyme replacement therapy ,pediatric study ,safety ,Pharmacology and Pharmaceutical Sciences ,Pharmacology and pharmaceutical sciences - Abstract
BackgroundFollowing a drug manufacturing process change, safety/efficacy of agalsidase alfa were evaluated in enzyme replacement therapy (ERT)-naïve children with Fabry disease.MethodsIn an open-label, multicenter, Phase II study (HGT-REP-084; Shire), 14 children aged ≥7 years received 0.2 mg/kg agalsidase alfa every other week for 55 weeks. Primary endpoints: safety, changes in autonomic function (2-hour Holter monitoring). Secondary endpoints: estimated glomerular filtration rate, left ventricular mass index (LVMI), midwall fractional shortening, pharmacodynamic parameters, and patient-reported quality-of-life.ResultsAmong five boys (median 10.2 [range 6.7, 14.4] years) and nine girls (14.8 [10.1, 15.9] years), eight patients experienced infusion-related adverse events (vomiting, n=4; nausea, n=3; dyspnea, n=3; chest discomfort, n=2; chills, n=2; dizziness, n=2; headache, n=2). One of these had several hypersensitivity episodes. However, no patient discontinued for safety reasons and no serious adverse events occurred. One boy developed immunoglobulin G (IgG) and neutralizing antidrug antibodies. Overall, no deterioration in cardiac function was observed in seven patients with low/abnormal SDNN (standard deviation of all filtered RR intervals;
- Published
- 2016
27. MULTIMODAL IMAGING OF A FAMILY WITH SPINOCEREBELLAR ATAXIA TYPE 7 DEMONSTRATING PHENOTYPIC VARIATION AND PROGRESSION OF RETINAL DEGENERATION
- Author
-
Levinson, Joshua D, Yan, Jiong, Lambert, Scott R, and Shankar, Suma P
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Neurodegenerative ,Neurosciences ,Clinical Research ,Rare Diseases ,Biomedical Imaging ,Brain Disorders ,Macular Degeneration ,Eye Disease and Disorders of Vision ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,Eye ,Neurological ,Adolescent ,Adult ,Child ,Disease Progression ,Female ,Humans ,Male ,Multimodal Imaging ,Retinal Degeneration ,Retrospective Studies ,Spinocerebellar Ataxias ,Opthalmology and Optometry ,Ophthalmology and optometry - Abstract
PurposeTo report the variability and progression of clinical presentation in three family members with spinocerebellar ataxia Type 7 including early recognizable features on retinal imaging and magnetic resonance imaging.MethodsRetrospective case series.ResultsThe proband, Patient 1 (mother) presented at age 26 with light perception vision. Initial examination was significant for optic disc pallor, vascular attenuation, and central macular atrophy. Two years later, her vision declined to no light perception, and fundus examination demonstrated marked progression of macular atrophy and peripheral bone spicule formation. Seven years after the onset of vision loss, neurologic examination demonstrated ataxia, dysarthria, and slowed saccades. Genetic testing of ATXN7 identified heterozygous 61-CAG trinucleotide repeat expansion confirming the diagnosis of spinocerebellar ataxia Type 7. Patient 2 (son) presented at age 11 with visual acuity of 20/300 bilaterally and decreased color vision. Funduscopic examination was notable for disc pallor, vascular attenuation, and peripheral pigmentary changes. Electroretinography demonstrated diminished rod and cone function, and Goldmann visual field testing revealed paracentral scotoma. Patient 3 (daughter) presented at age 14 with visual acuity of 20/50 bilaterally and minimal funduscopic changes. The only significant ophthalmic finding was retinal thinning with atrophy of the outer nuclear layer and subfoveal ellipsoid zone on optical coherence tomography. Early cerebellar volume loss was also noted on magnetic resonance imaging.ConclusionThe clinical presentation of spinocerebellar ataxia Type 7 can vary widely even within the same family. In individuals with vision loss and normal fundus examination, careful evaluation of optical coherence tomography and brain magnetic resonance imaging facilitates early diagnosis and genetic testing.
- Published
- 2016
28. Evaluation of disease burden and response to treatment in adults with type 1 gaucher disease using a validated disease severity scoring system (DS3)
- Author
-
Weinreb, Neal J, Finegold, David N, Feingold, Eleanor, Zeng, Zhen, Rosenbloom, Barry E, Shankar, Suma P, and Amato, Dominick
- Subjects
Clinical Research ,Neurodegenerative ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,Adolescent ,Adult ,Aged ,Alleles ,Child ,Child ,Preschool ,Female ,Gaucher Disease ,Genotype ,Homozygote ,Humans ,Male ,Middle Aged ,Retrospective Studies ,Young Adult ,Other Medical and Health Sciences ,Genetics & Heredity - Abstract
BackgroundGD1-DS3 is an integrated assessment of type 1 Gaucher disease (GD1) burden based on bone, hematologic and visceral domains. We investigated this disease severity scoring system (DS3) methodology for initial assessment, long-term follow-up and evaluation of treatment responses.MethodsWe enrolled 133 treated adult GD1 patients. Baseline DS3 scores were calculated near the initial treatment date and patients stratified by severity as marked (DS3 6.00-19.00), moderate (DS3 3.00-5.99), mild (DS3 < 3.00). Follow-up scores were calculated annually. Minimal clinically important improvement (MCII), is defined as ΔDS3 of -3.1.ResultsPatient characteristicsN370S was the most common allele (118 patients had at least one), 52 were N370S/N370S (48/52 were Ashkenazi Jews), N370S/L444P was the most common genotype among non-Jews. Median age of treatment: 45 years; median follow-up: 14 years. Baseline DS3 scores: Patients with marked disease (N = 58; median 7.84) were least likely to be N370S homozygous (19 %) and most likely to have had splenectomy (53 %), early age at diagnosis (median 18 years) and major pre-treatment bone pathology (76 %). Among patients with moderate disease (N = 53; median 4.33), 49 % were N370S/N370S, 15.1 % had splenectomy and 17 % had major bone disease. Median age at diagnosis: 32 years. No patient with mild disease (N = 22; median 2.4) had splenectomy or major skeletal disease. Median age at diagnosis: 40 years. 68 % were N370S homozygous. Response to treatment: Health-state transitions occurred primarily during the early treatment years. At Year 5, among 48 evaluable patients with marked baseline disease, eight were unchanged in severity status whereas 40 had MCII of varying degrees with 11 scored as mild. Among 42 evaluable moderate patients, none worsened, 16 remained moderate and 26 improved to mild. Among 16 evaluable mild patients, 14 remained so and 2 had DS3 scores in the low moderate range.ConclusionsDS3 is effective for assessing disease burden in GD1 and for monitoring response. ERT was associated with MCII in DS3 scores in patients with high severity. Nevertheless, despite better DS3 scores with treatment, GD1 patients especially those with splenectomy and pre-treatment bone pathology, continued to have bone complications.
- Published
- 2015
29. Neurocognition across the spectrum of mucopolysaccharidosis type I: Age, severity, and treatment
- Author
-
Shapiro, Elsa G, Nestrasil, Igor, Rudser, Kyle, Delaney, Kathleen, Kovac, Victor, Ahmed, Alia, Yund, Brianna, Orchard, Paul J, Eisengart, Julie, Niklason, Gregory R, Raiman, Julian, Mamak, Eva, Cowan, Morton J, Bailey-Olson, Mara, Harmatz, Paul, Shankar, Suma P, Cagle, Stephanie, Ali, Nadia, Steiner, Robert D, Wozniak, Jeffrey, Lim, Kelvin O, and Whitley, Chester B
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Mucopolysaccharidoses (MPS) ,Genetics ,Transplantation ,Rare Diseases ,Clinical Research ,Clinical Trials and Supportive Activities ,Adolescent ,Adult ,Age Factors ,Child ,Child ,Preschool ,Cognition ,Cognition Disorders ,Enzyme Replacement Therapy ,Female ,Gray Matter ,Hematopoietic Stem Cell Transplantation ,Humans ,Infant ,Male ,Mucopolysaccharidosis I ,Patient Outcome Assessment ,White Matter ,Young Adult ,Mucopolysaccharidosis Type I ,Neurocognition ,Genotypes ,Neuroimaging ,Genetics & Heredity ,Clinical sciences - Abstract
ObjectivesPrecise characterization of cognitive outcomes and factors that contribute to cognitive variability will enable better understanding of disease progression and treatment effects in mucopolysaccharidosis type I (MPS I). We examined the effects on cognition of phenotype, genotype, age at evaluation and first treatment, and somatic disease burden.MethodsSixty patients with severe MPS IH (Hurler syndrome treated with hematopoietic cell transplant and 29 with attenuated MPS I treated with enzyme replacement therapy), were studied with IQ measures, medical history, genotypes. Sixty-seven patients had volumetric MRI. Subjects were grouped by age and phenotype and MRI and compared to 96 normal controls.ResultsPrior to hematopoietic cell transplant, MPS IH patients were all cognitively average, but post-transplant, 59% were below average, but stable. Genotype and age at HCT were associated with cognitive ability. In attenuated MPS I, 40% were below average with genotype and somatic disease burden predicting their cognitive ability. White matter volumes were associated with IQ for controls, but not for MPS I. Gray matter volumes were positively associated with IQ in controls and attenuated MPS I patients, but negatively associated in MPS IH.ConclusionsCognitive impairment, a major difficulty for many MPS I patients, is associated with genotype, age at treatment and somatic disease burden. IQ association with white matter differed from controls. Many attenuated MPS patients have significant physical and/or cognitive problems and receive insufficient support services. Results provide direction for future clinical trials and better disease management.
- Published
- 2015
30. Founder Effect of a c.828+3A>T Splice Site Mutation in Peripherin 2 (PRPH2) Causing Autosomal Dominant Retinal Dystrophies
- Author
-
Shankar, Suma P, Birch, David G, Ruiz, Richard S, Hughbanks-Wheaton, Dianna K, Sullivan, Lori S, Bowne, Sara J, Stone, Edwin M, and Daiger, Stephen P
- Subjects
Biomedical and Clinical Sciences ,Ophthalmology and Optometry ,Neurosciences ,Neurodegenerative ,Eye Disease and Disorders of Vision ,Genetics ,Prevention ,Rare Diseases ,Eye ,Base Sequence ,Case-Control Studies ,Founder Effect ,Genetic Linkage ,Humans ,Molecular Sequence Data ,Mutation ,Peripherins ,Polymorphism ,Single-Stranded Conformational ,RNA Splice Sites ,Retinal Dystrophies ,Reverse Transcriptase Polymerase Chain Reaction ,Sequence Analysis ,DNA ,Ophthalmology and optometry - Abstract
ImportanceScreening for splice site mutation c.828+3A>T in the peripherin 2 (PRPH2) gene should be a high priority in families with highly variable retinal dystrophies. The correction of missplicing is a potential therapeutic target.ObjectiveTo determine the prevalence, genetic origin, and molecular mechanism of a donor c.828+3A>T mutation in the PRPH2 (peripherin 2, retinal degeneration slow) gene in individuals with retinal dystrophies.Design, setting, and participantsCase-control study that took place at the University of Texas Health Science Center, the University of Iowa, and the Retina Foundation of the Southwest, from January 1, 1987, to August 1, 2014, including affected individuals from 200 families with a diagnosis of autosomal dominant retinitis pigmentosa, 35 families with unspecified macular dystrophies, and 116 families with pattern dystrophy. Participants were screened for the c.828+3A>T mutation by restriction-enzyme digest, single-strand conformational polymorphism screening, or bidirectional sequencing. Haplotypes of polymorphic markers flanking the PRPH2 locus and sequence variants within the gene were determined by denaturing gel electrophoresis or automated capillary-based cycle sequencing. The effect of the splice site mutation on the PRPH2 transcript was analyzed using NetGene2, a splice prediction program and by the reverse transcription polymerase chain reaction of illegitimate transcripts from peripheral white blood cells.Main outcomes and measuresResults of testing for splice site mutation, haplotypes, and alternate transcripts.ResultsThe PRPH2 mutation was found in 97 individuals of 19 independently ascertained families with a clinical diagnosis of retinitis pigmentosa, macular dystrophy, and/or pattern dystrophy. All affected individuals also shared a rare haplotype of approximately 644 kilobase pairs containing the c.828+3A>T mutation, which extends from the short tandem repeat polymorphism D6S282 to c.1013G>A (rs434102, a single-nucleotide polymorphism) in exon 3 of PRPH2, suggesting this mutation is from a common ancestor and is a founder mutation. It has a prevalence of 2% in families diagnosed as having autosomal dominant retinitis pigmentosa and 10% in families with variable clinical diagnosis of pattern, macular, and retinal dystrophies. Individuals with the c.828+3A>T mutation expressed a PRPH2 transcript not found in control participants and that was consistent with abnormal splicing.Conclusions and relevanceThe PRPH2 c.828+3A>T splice site mutation is a frequent cause of inherited retinal dystrophies and is owing to the founder effect. The likely cause of disease is the missplicing of the PRPH2 message that results in a truncated protein product. Identifying the genetic etiology assists in more accurate management and possible future therapeutic options.
- Published
- 2015
31. Effect of Oral Eliglustat on Splenomegaly in Patients With Gaucher Disease Type 1: The ENGAGE Randomized Clinical Trial
- Author
-
Mistry, Pramod K, Lukina, Elena, Turkia, Hadhami Ben, Amato, Dominick, Baris, Hagit, Dasouki, Majed, Ghosn, Marwan, Mehta, Atul, Packman, Seymour, Pastores, Gregory, Petakov, Milan, Assouline, Sarit, Balwani, Manisha, Danda, Sumita, Hadjiev, Evgueniy, Ortega, Andres, Shankar, Suma, Solano, Maria Helena, Ross, Leorah, Angell, Jennifer, and Peterschmitt, M Judith
- Subjects
Digestive Diseases ,Clinical Trials and Supportive Activities ,Hematology ,Clinical Research ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Administration ,Oral ,Adult ,Double-Blind Method ,Enzyme Inhibitors ,Female ,Gaucher Disease ,Glucosyltransferases ,Humans ,Male ,Organ Size ,Pyrrolidines ,Spleen ,Splenomegaly ,Young Adult ,Medical and Health Sciences ,General & Internal Medicine - Abstract
ImportanceGaucher disease type 1 is characterized by hepatosplenomegaly, anemia, thrombocytopenia, and skeletal disease. A safe, effective oral therapy is needed.ObjectiveTo determine whether eliglustat, a novel oral substrate reduction therapy, safely reverses clinical manifestations in untreated adults with Gaucher disease type 1.Design, setting, and participantsPhase 3, randomized, double-blind, placebo-controlled trial conducted at 18 sites in 12 countries from November 2009 to July 2012 among eligible patients with splenomegaly plus thrombocytopenia and/or anemia. Of 72 patients screened, 40 were enrolled.InterventionsPatients were stratified by spleen volume and randomized 1:1 to receive eliglustat (50 or 100 mg twice daily; n = 20) or placebo (n = 20) for 9 months.Main outcomes and measuresThe primary efficacy end point was percentage change in spleen volume in multiples of normal from baseline to 9 months; secondary efficacy end points were change in hemoglobin level and percentage changes in liver volume and platelet count.ResultsAll patients had baseline splenomegaly and thrombocytopenia (mostly moderate or severe), most had mild or moderate hepatomegaly, and 20% had mild anemia. Least-square mean spleen volume decreased by 27.77% (95% CI, -32.57% to -22.97%) in the eliglustat group (from 13.89 to 10.17 multiples of normal) vs an increase of 2.26% (95% CI, -2.54% to 7.06%) in the placebo group (from 12.50 to 12.84 multiples of normal) for an absolute treatment difference of -30.03% (95% CI, -36.82% to -23.24%; P
- Published
- 2015
32. Oral Migalastat HCl Leads to Greater Systemic Exposure and Tissue Levels of Active α-Galactosidase A in Fabry Patients when Co-Administered with Infused Agalsidase
- Author
-
Warnock, David G, Bichet, Daniel G, Holida, Myrl, Goker-Alpan, Ozlem, Nicholls, Kathy, Thomas, Mark, Eyskens, Francois, Shankar, Suma, Adera, Mathews, Sitaraman, Sheela, Khanna, Richie, Flanagan, John J, Wustman, Brandon A, Barth, Jay, Barlow, Carrolee, Valenzano, Kenneth J, Lockhart, David J, Boudes, Pol, and Johnson, Franklin K
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Clinical Trials and Supportive Activities ,Clinical Research ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,6.1 Pharmaceuticals ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,1-Deoxynojirimycin ,Administration ,Oral ,Adult ,Area Under Curve ,Demography ,Fabry Disease ,Humans ,Infusion Pumps ,Isoenzymes ,Male ,Middle Aged ,Recombinant Proteins ,Skin ,alpha-Galactosidase ,General Science & Technology - Abstract
UnlabelledMigalastat HCl (AT1001, 1-Deoxygalactonojirimycin) is an investigational pharmacological chaperone for the treatment of α-galactosidase A (α-Gal A) deficiency, which leads to Fabry disease, an X-linked, lysosomal storage disorder. The currently approved, biologics-based therapy for Fabry disease is enzyme replacement therapy (ERT) with either agalsidase alfa (Replagal) or agalsidase beta (Fabrazyme). Based on preclinical data, migalastat HCl in combination with agalsidase is expected to result in the pharmacokinetic (PK) enhancement of agalsidase in plasma by increasing the systemic exposure of active agalsidase, thereby leading to increased cellular levels in disease-relevant tissues. This Phase 2a study design consisted of an open-label, fixed-treatment sequence that evaluated the effects of single oral doses of 150 mg or 450 mg migalastat HCl on the PK and tissue levels of intravenously infused agalsidase (0.2, 0.5, or 1.0 mg/kg) in male Fabry patients. As expected, intravenous administration of agalsidase alone resulted in increased α-Gal A activity in plasma, skin, and peripheral blood mononuclear cells (PBMCs) compared to baseline. Following co-administration of migalastat HCl and agalsidase, α-Gal A activity in plasma was further significantly increased 1.2- to 5.1-fold compared to agalsidase administration alone, in 22 of 23 patients (95.6%). Importantly, similar increases in skin and PBMC α-Gal A activity were seen following co-administration of migalastat HCl and agalsidase. The effects were not related to the administered migalastat HCl dose, as the 150 mg dose of migalastat HCl increased α-Gal A activity to the same extent as the 450 mg dose. Conversely, agalsidase had no effect on the plasma PK of migalastat. No migalastat HCl-related adverse events or drug-related tolerability issues were identified.Trial registrationClinicalTrials.gov NCT01196871.
- Published
- 2015
33. Characterization of Early Disease Status in Treatment-Naive Male Paediatric Patients with Fabry Disease Enrolled in a Randomized Clinical Trial
- Author
-
Wijburg, Frits A, Bénichou, Bernard, Bichet, Daniel G, Clarke, Lorne A, Dostalova, Gabriela, Fainboim, Alejandro, Fellgiebel, Andreas, Forcelini, Cassiano, Haack, Kristina An, Hopkin, Robert J, Mauer, Michael, Najafian, Behzad, Scott, C Ronald, Shankar, Suma P, Thurberg, Beth L, Tøndel, Camilla, Tylki-Szymańska, Anna, and Ramaswami, Uma
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Kidney Disease ,Rare Diseases ,Clinical Research ,Prevention ,Pediatric ,Clinical Trials and Supportive Activities ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Renal and urogenital ,Adolescent ,Biopsy ,Brain ,Child ,Child ,Preschool ,Demography ,Endothelium ,Vascular ,Fabry Disease ,Genotype ,Glomerular Filtration Rate ,Glycolipids ,Humans ,Iohexol ,Kidney ,Male ,Mutation ,Quality of Life ,Skin ,Sphingolipids ,Trihexosylceramides ,General Science & Technology - Abstract
Trial designThis analysis characterizes the degree of early organ involvement in a cohort of oligo-symptomatic untreated young patients with Fabry disease enrolled in an ongoing randomized, open-label, parallel-group, phase 3B clinical trial.MethodsMales aged 5-18 years with complete α-galactosidase A deficiency, without symptoms of major organ damage, were enrolled in a phase 3B trial evaluating two doses of agalsidase beta. Baseline disease characteristics of 31 eligible patients (median age 12 years) were studied, including cellular globotriaosylceramide (GL-3) accumulation in skin (n = 31) and kidney biopsy (n = 6; median age 15 years; range 13-17 years), renal function, and glycolipid levels (plasma, urine).ResultsPlasma and urinary GL-3 levels were abnormal in 25 of 30 and 31 of 31 patients, respectively. Plasma lyso-GL-3 was elevated in all patients. GL-3 accumulation was documented in superficial skin capillary endothelial cells (23/31 patients) and deep vessel endothelial cells (23/29 patients). The mean glomerular filtration rate (GFR), measured by plasma disappearance of iohexol, was 118.1 mL/min/1.73 m(2) (range 90.4-161.0 mL/min/1.73 m(2)) and the median urinary albumin/creatinine ratio was 10 mg/g (range 4.0-27.0 mg/g). On electron microscopy, renal biopsy revealed GL-3 accumulation in all glomerular cell types (podocytes and parietal, endothelial, and mesangial cells), as well as in peritubular capillary and non-capillary endothelial, interstitial, vascular smooth muscle, and distal tubules/collecting duct cells. Lesions indicative of early Fabry arteriopathy and segmental effacement of podocyte foot processes were found in all 6 patients.ConclusionsThese data reveal that in this small cohort of children with Fabry disease, histological evidence of GL-3 accumulation, and cellular and vascular injury are present in renal tissues at very early stages of the disease, and are noted before onset of microalbuminuria and development of clinically significant renal events (e.g. reduced GFR). These data give additional support to the consideration of early initiation of enzyme replacement therapy, potentially improving long-term outcome.Trial registrationClinicalTrials.gov NCT00701415.
- Published
- 2015
34. Cardio-Facio-Cutaneous Syndrome: Clinical Features, Diagnosis, and Management Guidelines
- Author
-
Pierpont, Mary Ella M, Magoulas, Pilar L, Adi, Saleh, Kavamura, Maria Ines, Neri, Giovanni, Noonan, Jacqueline, Pierpont, Elizabeth I, Reinker, Kent, Roberts, Amy E, Shankar, Suma, Sullivan, Joseph, Wolford, Melinda, Conger, Brenda, Santa Cruz, Molly, and Rauen, Katherine A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Brain Disorders ,Pediatric ,Rare Diseases ,Intellectual and Developmental Disabilities (IDD) ,Clinical Research ,Congenital Structural Anomalies ,7.3 Management and decision making ,Management of diseases and conditions ,Congenital ,Diagnosis ,Differential ,Disease Management ,Ectodermal Dysplasia ,Facies ,Failure to Thrive ,Genetic Testing ,Heart Defects ,Congenital ,Humans ,Practice Guidelines as Topic ,cardio-facio-cutaneous syndrome ,BRAF mutation ,management guidelines ,MEK1 mutation ,MEK2 mutation ,RASopathy ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Pediatrics ,Biomedical and clinical sciences ,Health sciences ,Psychology - Abstract
Cardio-facio-cutaneous syndrome (CFC) is one of the RASopathies that bears many clinical features in common with the other syndromes in this group, most notably Noonan syndrome and Costello syndrome. CFC is genetically heterogeneous and caused by gene mutations in the Ras/mitogen-activated protein kinase pathway. The major features of CFC include characteristic craniofacial dysmorphology, congenital heart disease, dermatologic abnormalities, growth retardation, and intellectual disability. It is essential that this condition be differentiated from other RASopathies, as a correct diagnosis is important for appropriate medical management and determining recurrence risk. Children and adults with CFC require multidisciplinary care from specialists, and the need for comprehensive management has been apparent to families and health care professionals caring for affected individuals. To address this need, CFC International, a nonprofit family support organization that provides a forum for information, support, and facilitation of research in basic medical and social issues affecting individuals with CFC, organized a consensus conference. Experts in multiple medical specialties provided clinical management guidelines for pediatricians and other care providers. These guidelines will assist in an accurate diagnosis of individuals with CFC, provide best practice recommendations, and facilitate long-term medical care.
- Published
- 2014
35. Reported outcomes of 453 pregnancies in patients with Gaucher disease: An analysis from the Gaucher outcome survey
- Author
-
Lau, Heather, Belmatoug, Nadia, Deegan, Patrick, Goker-Alpan, Ozlem, Schwartz, Ida Vanessa D., Shankar, Suma P, Panahloo, Zoya, and Zimran, Ari
- Published
- 2018
- Full Text
- View/download PDF
36. Consensus Statement of the Indian Academy of Pediatrics on Diagnosis and Management of Fragile X Syndrome in India
- Author
-
Sachdeva, Anupam, Jain, Prachi, Gunasekaran, Vinod, Mahay, Sunita Bijarnia, Mukherjee, Sharmila, Hagerman, Randi, Shankar, Suma, Kapoor, Seema, and Kedia, Shalini N
- Published
- 2019
- Full Text
- View/download PDF
37. P384: A novel nonsense variant in the DMBX1 gene associated with neurodevelopmental delay: A case report of three siblings
- Author
-
Sarkisian, Miriam, Shankar, Suma, and Herman, Kristin
- Published
- 2024
- Full Text
- View/download PDF
38. P327: Congenital disorder of glycosylation type IIa with a novel phenotype of bilateral congenital glaucoma
- Author
-
Egense, Alena, Do, Timothy, Herman, Kristin, Brandt, James, and Shankar, Suma
- Published
- 2024
- Full Text
- View/download PDF
39. Case 11.12.4 - Neurodevelopmental Cases: Autism Spectrum Disorder
- Author
-
Triano, Vivien Narcisa and Shankar, Suma
- Published
- 2024
- Full Text
- View/download PDF
40. List of Contributors
- Author
-
Al Saif, Hind, primary, Almeida, Tawany, additional, Babu, Kevin, additional, Bajaj, Gurjas S., additional, Bitar, Maya, additional, Brar, Vikram S., additional, Bregman, Jana, additional, Couser, Natario L., additional, Fard, Sara, additional, Hara, Yurika, additional, Humberson, Jennifer, additional, Hunter, Thomas, additional, Hutchinson, Amy K., additional, Kaleem, Mona, additional, Knight, O'Rese J., additional, Lambert, Scott R., additional, Love, Eleanor, additional, Masood, Maheer, additional, Mathews, Michaela, additional, Melvani, Rakhi, additional, Miller, Payton M., additional, Nasir, Mariam, additional, Ngo, Harrison, additional, Pande, Chetna, additional, Pandya, Arti, additional, Pflugrath, Adam E., additional, Powell, Cynthia M., additional, Randolph, Jessica, additional, Rhodes, Jennifer, additional, Richards, Nikisha Q., additional, Sahni, Dev R., additional, Sethi, Dhruv, additional, Shankar, Suma P., additional, Silverstein, Evan, additional, Smith, Laurie D., additional, Smith-Marshall, Janine, additional, and Toffoli, Daniela, additional
- Published
- 2019
- Full Text
- View/download PDF
41. Neurocutaneous Syndromes (Phakomatoses)
- Author
-
Shankar, Suma P., primary, Humberson, Jennifer, additional, Melvani, Rakhi, additional, and Couser, Natario L., additional
- Published
- 2019
- Full Text
- View/download PDF
42. Effect and Tolerability of Agalsidase Alfa in Patients with Fabry Disease Who Were Treatment Naïve or Formerly Treated with Agalsidase Beta or Agalsidase Alfa
- Author
-
Goker-Alpan, Ozlem, Nedd, Khan, Shankar, Suma P., Lien, Yeong-Hau H., Weinreb, Neal, Wijatyk, Anna, Chang, Peter, Martin, Rick, Zschocke, Johannes, Editor-in-chief, Baumgartner, Matthias, editor, Morava, Eva, editor, Patterson, Marc, editor, Rahman, Shamima, editor, and Peters, Verena, editor
- Published
- 2015
- Full Text
- View/download PDF
43. Prader-Willi and Angelman Syndromes: Mechanisms and Management
- Author
-
Ma,Van K, Mao,Rong, Toth,Jessica N, Fulmer,Makenzie L, Egense,Alena S, Shankar,Suma P, Ma,Van K, Mao,Rong, Toth,Jessica N, Fulmer,Makenzie L, Egense,Alena S, and Shankar,Suma P
- Abstract
Van K Ma,1,2 Rong Mao,3,4 Jessica N Toth,3 Makenzie L Fulmer,3,4 Alena S Egense,1,2 Suma P Shankar1,2,5 1Department of Pediatrics, University of California Davis, Sacramento, CA, USA; 2MIND Institute, University of California Davis, Sacramento, CA, USA; 3Molecular Genetics and Genomics, ARUP Laboratories, Salt Lake City, UT, USA; 4Department of Pathology, University of Utah, Salt Lake City, UT, USA; 5Department of Ophthalmology, University of California Davis, Sacramento, CA, USACorrespondence: Suma P Shankar, MIND Institute, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA, Tel +1 916 703 0235, Fax + 1 916 703 0203, Email Spshankar@ucdavis.eduAbstract: Prader-Willi syndrome (PWS) and Angelman syndrome (AS) are genetic imprinting disorders resulting from absent or reduced expression of paternal or maternal genes in chromosome 15q11q13 region, respectively. The most common etiology is deletion of the maternal or paternal 15q11q13 region. Methylation is the first line for molecular diagnostic testing; MS-MLPA is the most sensitive test. The molecular subtype of PWS/AS provides more accurate recurrence risk information for parents and for the individual affected with the condition. Management should include a multidisciplinary team by various medical subspecialists and therapists. Developmental and behavioral management of PWS and AS in infancy and early childhood includes early intervention services and individualized education programs for school-aged children. Here, we compare and discuss the mechanisms, pathophysiology, clinical features, and management of the two imprinting disorders, PWS and AS.Keywords: imprinting disorders, uniparental disomy, chromosome 15, developmental delay, hyperphagia, obesity
- Published
- 2023
44. Insights into the perinatal phenotype of Kabuki syndrome in infants identified by genome‐wide sequencing
- Author
-
Wigby, Kristen, primary, Hammer, Monia, additional, Tokita, Mari, additional, Patel, Priyanka, additional, Jones, Marilyn C., additional, Larson, Austin, additional, Bartolomei, Frances Velez, additional, Dykzeul, Natalie, additional, Slavotinek, Anne, additional, Yip, Tiffany, additional, Bandres‐Ciga, Sara, additional, Simpson, Brittany N., additional, Suhrie, Kristen, additional, Shankar, Suma, additional, Veith, Regan, additional, Bragg, Jennifer, additional, Powell, Cynthia, additional, Kingsmore, Stephen F., additional, Dimmock, David, additional, Maron, Jill, additional, Davis, Jonathan, additional, and Del Campo, Miguel, additional
- Published
- 2023
- Full Text
- View/download PDF
45. P302: Clinical genome sequencing and targeted mouse modeling in nonverbal or minimally verbal individuals with autism spectrum disorders and neurodevelopmental delays
- Author
-
Shankar, Suma, primary, Grimsrud, Kristin, additional, Ma, Van, additional, Shankar, Prabhu, additional, Osborne, Ellen, additional, Higa, Leigh Ann, additional, Iyer, Ishaan, additional, Lloyd, Kent, additional, and Rauen, Katherine, additional
- Published
- 2023
- Full Text
- View/download PDF
46. P668: Automated identification and indexing of genes and variants using bidirectional encoder representation from transformers
- Author
-
Vora, Jayneel, primary, Shankar, Suma, additional, Abbeduto, Leonard, additional, and Higareda, Abigail, additional
- Published
- 2023
- Full Text
- View/download PDF
47. Genetic Factors Modifying Clinical Expression of Autosomal Dominant RP
- Author
-
Daiger, Stephen P., Shankar, Suma P., Schindler, Alice B., Sullivan, Lori S., Bowne, Sara J., King, Terri M., Daw, E. Warick, Stone, Edwin M., Heckenlively, John R., Back, Nathan, editor, Cohen, Irun R., editor, Kritchevsky, David, editor, Lajtha, Abel, editor, Paoletti, Rodolfo, editor, Hollyfield, Joe G., editor, Anderson, Robert E., editor, and LaVail, Matthew M., editor
- Published
- 2006
- Full Text
- View/download PDF
48. Retinal dystrophies: A look beyond the eyes
- Author
-
Tang, Vincent Duong, primary, Egense, Alena, additional, Yiu, Glenn, additional, Meyers, Elijah, additional, Moshiri, Ala, additional, and Shankar, Suma P., additional
- Published
- 2022
- Full Text
- View/download PDF
49. Atypical presentation of Angelman syndrome with intact expressive language due to low‐level mosaicism
- Author
-
Punatar, Ruchi, primary, Egense, Alena, additional, Mao, Rong, additional, Procter, Melinda, additional, Bosworth, Michelle, additional, Quigley, Denise I., additional, Angkustsiri, Kathleen, additional, and Shankar, Suma P., additional
- Published
- 2022
- Full Text
- View/download PDF
50. Atypical presentation of Angelman syndrome with intact expressive language due to low-level mosaicism.
- Author
-
Punatar, Ruchi, Punatar, Ruchi, Egense, Alena, Mao, Rong, Procter, Melinda, Bosworth, Michelle, Quigley, Denise I, Angkustsiri, Kathleen, Shankar, Suma P, Punatar, Ruchi, Punatar, Ruchi, Egense, Alena, Mao, Rong, Procter, Melinda, Bosworth, Michelle, Quigley, Denise I, Angkustsiri, Kathleen, and Shankar, Suma P
- Abstract
BackgroundAngelman syndrome (AS) occurs due to a lack of expression or function of the maternally inherited UBE3A gene. Individuals with AS typically have significant developmental delay, severe speech impairment with absent to minimal verbal language, gait abnormalities including ataxia, and an incongruous happy demeanor. The majority of individuals with AS also have seizures and microcephaly. Some individuals with mosaic AS have been reported to have expressive language and milder levels of developmental delay.Case reportWe report a male patient presenting with mild to moderate intellectual disability, hyperphagia, obesity, and the ability to communicate verbally. His phenotype was suggestive of Prader-Willi syndrome. However, methylation testing was positive for Angelman syndrome and additional methylation specific multiplex ligation-dependent amplification (MS-MLPA) study revealed low-level mosaicism for AS.ConclusionA broader phenotypic spectrum should be considered for AS as patients with atypical presentations may otherwise elude diagnosis.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.