1
|
Crossnohere NL, Campoamor NB, Camino E, Dresnick E, Martschenko DO, Rodrigues V, Apkon S, Hazlett A, Mittur D, Rodriguez PE, Bridges JFP, Armstrong N. Barriers to diverse clinical trial participation in Duchenne muscular dystrophy: Engaging Hispanic/Latina caregivers and health professionals. Orphanet J Rare Dis 2024; 19:207. [PMID: 38773664 PMCID: PMC11110421 DOI: 10.1186/s13023-024-03209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/05/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Despite the increasing availability of clinical trials in Duchenne muscular dystrophy, racial/ethnic minorities and other populations facing health disparities remain underrepresented in clinical trials evaluating products for Duchenne. We sought to understand the barriers faced by Hispanic/Latino families specifically and underrepresented groups more generally to clinical trial participation in Duchenne. METHODS We engaged two participant groups: Hispanic/Latino caregivers of children with Duchenne in the US, including Puerto Rico, and health professionals within the broader US Duchenne community. Caregiver interviews explored attitudes towards and experiences with clinical trials, while professional interviews explored barriers to clinical trial participation among socio-demographically underrepresented families (e.g., low income, rural, racial/ethnic minority, etc.). Interviews were analyzed aggregately and using a thematic analysis approach. An advisory group was engaged throughout the course of the study to inform design, conduct, and interpretation of findings generated from interviews. RESULTS Thirty interviews were conducted, including with 12 Hispanic/Latina caregivers and 18 professionals. We identified barriers to clinical trial participation at various stages of the enrollment process. In the initial identification of patients, barriers included lack of awareness about trials and clinical trial locations at clinics that were less likely to serve diverse patients. In the prescreening process, barriers included ineligibility, anticipated non-compliance in clinical trial protocols, and language discrimination. In screening, barriers included concerns about characteristics of the trial, as well as mistrust/lack of trust. In consent and recruitment, barriers included lack of timely decision support, logistical factors (distance, time, money), and lack of translated study materials. CONCLUSIONS Numerous barriers hinder participation in Duchenne clinical trials for Hispanic/Latino families and other populations experiencing health disparities. Addressing these barriers necessitates interventions across multiple stages of the clinical trial enrollment process. Recommendations to enhance participation opportunities include developing clinical trial decision support tools, translating prominent clinical trials educational resources such as ClinicalTrials.gov, fostering trusting family-provider relationships, engaging families in clinical trial design, and establishing ethical guidelines for pre-screening potentially non-compliant patients.
Collapse
Affiliation(s)
- Norah L Crossnohere
- Division of General Internal Medicine, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Nicola B Campoamor
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Erin Dresnick
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | | | - Viana Rodrigues
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Susan Apkon
- Department of Physical Medicine and Rehabilitation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Dhruv Mittur
- Patient partner, Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Priscilla E Rodriguez
- Diversity Inclusion Advocacy Manager, EveryLife Foundation for Rare Diseases, Washington, DC, USA
| | - John F P Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
| | | |
Collapse
|
2
|
Bhattacharyya O, Campoamor NB, Armstrong N, Freed M, Schrader R, Crossnohere NL, Bridges JFP. Assessing the Benefits and Harms Associated with Early Diagnosis from the Perspective of Parents with Multiple Children Diagnosed with Duchenne Muscular Dystrophy. Int J Neonatal Screen 2024; 10:32. [PMID: 38651397 PMCID: PMC11036293 DOI: 10.3390/ijns10020032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/26/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare neuromuscular disorder diagnosed in childhood. Limited newborn screening in the US often delays diagnosis. With multiple FDA-approved therapies, early diagnosis is crucial for timely treatment but may entail other benefits and harms. Using a community-based survey, we explored how parents of siblings with DMD perceived early diagnosis of one child due to a prior child's diagnosis. We assessed parents' viewpoints across domains including diagnostic journey, treatment initiatives, service access, preparedness, parenting, emotional impact, and caregiving experience. We analyzed closed-ended responses on a -1.0 to +1.0 scale to measure the degree of harm or benefit parents perceived and analyzed open-ended responses thematically. A total of 45 parents completed the survey, with an average age of 43.5 years and 20.0% identifying as non-white. Younger siblings were diagnosed 2 years earlier on average (p < 0.001). Overall, parents viewed early diagnosis positively (mean: 0.39), particularly regarding school preparedness (+0.79), support services (+0.78), treatment evaluation (+0.68), and avoiding diagnostic odyssey (+0.67). Increased worry was a common downside (-0.40). Open-ended responses highlighted improved outlook and health management alongside heightened emotional distress and treatment burdens. These findings address gaps in the evidence by documenting the effectiveness of early screening and diagnosis of DMD using sibling data.
Collapse
Affiliation(s)
- Oindrila Bhattacharyya
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (O.B.); (N.B.C.); (J.F.P.B.)
| | - Nicola B. Campoamor
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (O.B.); (N.B.C.); (J.F.P.B.)
| | - Niki Armstrong
- Foundation for Angelman Syndrome Therapeutics, Austin, TX 78704, USA;
| | - Megan Freed
- Parent Project Muscular Dystrophy, Washington, DC 20005, USA; (M.F.); (R.S.)
| | - Rachel Schrader
- Parent Project Muscular Dystrophy, Washington, DC 20005, USA; (M.F.); (R.S.)
| | - Norah L. Crossnohere
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 43202, USA
| | - John F. P. Bridges
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA; (O.B.); (N.B.C.); (J.F.P.B.)
| |
Collapse
|
3
|
Armstrong N, Apkon S, Berggren KN, Braun C, Ciafaloni E, Connolly A, Kennedy A, Kuntz N, Mathews K, McGuire M, Parad R, Scavina M, Scharf RJ, Waldrop M. The Early Care (0-3 Years) In Duchenne Muscular Dystrophy Meeting Report. J Neuromuscul Dis 2024; 11:525-533. [PMID: 38189762 DOI: 10.3233/jnd-230180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Objective This report summarizes the key discussions from the "Early Care (0-3 years) in Duchenne Muscular Dystrophy" meeting, which aimed to address the challenges and opportunities in the diagnosis and care of Duchenne muscular dystrophy (DMD) and female carriers within the 0-3-year age group. Methods The meeting brought together experts and healthcare providers who shared insights, discussed advancements in DMD care, and identified research needs. Presentations covered diagnostic challenges, approved therapies, clinical trials, identification of young female carriers, and the importance of clinical care and support for families. Results The meeting highlighted the importance of timely diagnosis and the lack of evidence-based guidelines for the care of children with DMD aged 0-3 years. Diagnostic challenges were discussed, including delays in receiving a DMD diagnosis and disparities based on ethnicity. The potential benefits and process of newborn screening were addressed.Approved therapeutic interventions, such as corticosteroids and exon-skipping drugs, were explored, with studies indicating the potential benefits of early initiation of corticosteroid therapy and the safety of exon-skipping drugs in DMD. Clinical trials involving infants and young boys were discussed, focusing on drugs like ataluren, vamorolone, and gene therapies.The meeting emphasized the importance of clinical care and support for families, including comprehensive information provision, early intervention services, and individualized support. The identification and care of young female carriers were also addressed. Conclusion The meeting provided a platform for experts and healthcare providers to discuss and identify key aspects of early care for children with DMD aged 0-3 years. The meeting emphasized the need for early diagnosis, evidence-based guidelines, and comprehensive care and support for affected children and their families. Further research, collaboration, and the development of consensus guidelines are needed to improve early diagnosis, treatment, and outcomes in this population.
Collapse
Affiliation(s)
| | - Susan Apkon
- Children's Hospital Colorado/University of Colorado, Aurora, CO, USA
| | - Kiera N Berggren
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
| | - Catherine Braun
- University of Virginia Children's Hospital, Charlottesville, VA, USA
| | | | | | - Annie Kennedy
- EveryLife Foundation for Rare Diseases, Washington, DC, USA
| | - Nancy Kuntz
- Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Katherine Mathews
- Stead Family Department of Pediatrics, University of Iowa, , Iowa City, IA, USA
| | | | - Richard Parad
- Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Mena Scavina
- Nemours Children's Hospital-Delaware, Wilmington, DE, USA
| | - Rebecca J Scharf
- University of Virginia Children's Hospital, Charlottesville, VA, USA
| | | |
Collapse
|
4
|
Mercuri E, Seferian A, Servais L, Deconinck N, Stevenson H, Ni X, Zhang W, East L, Yonren S, Muntoni F, Deconinck N, Van Coster R, Vanlander A, Seferian A, De Lucia S, Gidaro T, Brande LV, Servais L, Kirschner J, Borell S, Mercuri E, Brogna C, Pane M, Fanelli L, Norcia G, Muntoni F, Brusa C, Chesshyre M, Maresh K, Pitchforth J, Schottlaender L, Scoto M, Silwal A, Trucco F. Safety, tolerability and pharmacokinetics of eteplirsen in young boys aged 6–48 months with Duchenne muscular dystrophy amenable to exon 51 skipping. Neuromuscul Disord 2023; 33:476-483. [PMID: 37207382 DOI: 10.1016/j.nmd.2023.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
Eteplirsen is FDA-approved for the treatment of Duchenne muscular dystrophy (DMD) in exon 51 skip-amenable patients. Previous studies in boys > 4 years of age indicate eteplirsen is well tolerated and attenuates pulmonary and ambulatory decline compared with matched natural history cohorts. Here the safety, tolerability and pharmacokinetics of eteplirsen in boys aged 6-48 months is evaluated. In this open-label, multicenter, dose-escalation study (NCT03218995), boys with a confirmed mutation of the DMD gene amenable to exon 51 skipping (Cohort 1: aged 24-48 months, n = 9; Cohort 2: aged 6 to < 24 months, n = 6) received ascending doses (2, 4, 10, 20, 30 mg/kg) of once-weekly eteplirsen intravenously over 10 weeks, continuing at 30 mg/kg up to 96 weeks. Endpoints included safety (primary) and pharmacokinetics (secondary). All 15 participants completed the study. Eteplirsen was well tolerated with no treatment-related discontinuations, deaths or evidence of kidney toxicity. Most treatment-emergent adverse events were mild; most common were pyrexia, cough, nasopharyngitis, vomiting, and diarrhea. Eteplirsen pharmacokinetics were consistent between both cohorts and with previous clinical experience in boys with DMD > 4 years of age. These data support the safety and tolerability of eteplirsen at the approved 30-mg/kg dose in boys as young as 6 months old.
Collapse
|
5
|
Den Hartog L, Asakura A. Implications of notch signaling in duchenne muscular dystrophy. Front Physiol 2022; 13:984373. [PMID: 36237531 PMCID: PMC9553129 DOI: 10.3389/fphys.2022.984373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
This review focuses upon the implications of the Notch signaling pathway in muscular dystrophies, particularly Duchenne muscular dystrophy (DMD): a pervasive and catastrophic condition concerned with skeletal muscle degeneration. Prior work has defined the pathogenesis of DMD, and several therapeutic approaches have been undertaken in order to regenerate skeletal muscle tissue and ameliorate the phenotype. There is presently no cure for DMD, but a promising avenue for novel therapies is inducing muscle regeneration via satellite cells (muscle stem cells). One specific target using this approach is the Notch signaling pathway. The canonical Notch signaling pathway has been well-characterized and it ultimately governs cell fate decision, cell proliferation, and induction of differentiation. Additionally, inhibition of the Notch signaling pathway has been directly implicated in the deficits seen with muscular dystrophies. Here, we explore the connection between the Notch signaling pathway and DMD, as well as how Notch signaling may be targeted to improve the muscle degeneration seen in muscular dystrophies.
Collapse
|