1
|
Jarnes JR, Pillai NR, Ahmed A, Shrestha S, Stark M, Whitley CB. Dose-intensive therapy (DIT) for infantile Pompe disease: A pilot study. Mol Genet Metab Rep 2025; 42:101179. [PMID: 39802096 PMCID: PMC11720876 DOI: 10.1016/j.ymgmr.2024.101179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Background The current standard of care for infantile-onset Pompe disease (IOPD), a severe form of acid α-glucosidase enzyme activity deficiency is: (1) detection by newborn screening, (2) early initiation of intravenous enzyme replacement therapy (ERT) using recombinant human acid alpha-glucosidase (rhGAA), with higher doses of rhGAA increasingly used to improve clinical outcomes, and (3) immune tolerization induction (ITI) using to prevent anti-rhGAA antibody formation, with methotrexate (MTX), rituximab, and IVIG used for patients who are cross-reactive immunologic material negative (CRIM-) and monotherapy with MTX used in patients who are cross-reactive immunologic material positive (CRIM+). Objectives/methods A pilot study evaluates a dose-intensive therapy (DIT) using high-dose ERT (40 mg/kg/week) and more frequent exposure to ERT (i.e., 3 times weekly administration) to mitigate anti-rhGAA antibody formation, as an alternative to the standard therapeutic approach for IOPD. Results In the first patient, DIT resulted in rapid normalization of the following: (1) bi-ventricular hypertrophy, (2) urine HEX-4, (3) CK, (4) liver transaminases. At 7 years of age, the patient continues the DIT regimen. To date, all pediatric developmental milestones have been met on time, anti-rhGAA antibodies have been negative and the patient is able to attend school and maintain normal activities of daily living. Conclusions Over a 7-year period, DIT for CRIM-positive IOPD was well tolerated in the first patient treated. Excellent clinical outcomes were achieved, and anti-rhGAA antibodies levels were consistently undetectable. Assessments of more patients, that includes patients with CRIM-, as well as CRIM+ IOPD, will determine if this approach consistently achieves improved clinical outcomes and immune tolerization.
Collapse
Affiliation(s)
- Jeanine R. Jarnes
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Nishitha R. Pillai
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
| | - Alia Ahmed
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
| | - Sofia Shrestha
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- M Health Fairview Masonic Children's Hospital, Minneapolis, MN, USA
| | - Molly Stark
- M Health Fairview Masonic Children's Hospital, Minneapolis, MN, USA
| | - Chester B. Whitley
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Advanced Therapies Program, University of Minnesota, Fairview, Minneapolis, MN, USA
- Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Fares AH, Desai AK, Case LE, Sharon C, Klinepeter A, Kirby A, Lisi MT, Koch RL, Kishnani PS. Optimizing clinical outcomes: The journey of twins with CRIM-negative infantile-onset Pompe disease on high-dose enzyme replacement therapy and immunomodulation. Mol Genet Metab Rep 2024; 41:101141. [PMID: 39314994 PMCID: PMC11419802 DOI: 10.1016/j.ymgmr.2024.101141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Infantile-onset Pompe disease (IOPD) is caused by a deficiency in the enzyme acid alpha-glucosidase (GAA). It is characterized by severe and progressive hypertrophic cardiomyopathy and muscle weakness with death in the first 2 years of life if left untreated. Enzyme replacement therapy (ERT) with alglucosidase-alfa is lifesaving, but its effectiveness is influenced by the patient's cross-reactive immunologic material (CRIM) status, dose of ERT, and the development of high antibody titers, which can reduce the therapy's efficacy. The inability of CRIM-negative IOPD patients to produce native GAA exposes them to a high risk of development of anti-rhGAA IgG antibody titers, leading to treatment failure. We present the case of CRIM-negative dizygotic twins treated with high-dose alglucosidase-alfa (40 mg/kg/week), initiated at 28 days (Twin A) and 44 days (Twin B). Both twins received immune tolerance induction (ITI) with rituximab, methotrexate, and IVIG to mitigate antibody response. Initial evaluations revealed elevated left ventricular mass index (LVMI) and elevated biomarkers (urine glucose tetrasaccharide (Glc4), creatine kinase (CK), and aspartate aminotransferase (AST)) in both twins. Following treatment, cardiac function and biomarkers normalized within several months, with a slight delay in Twin B compared to Twin A, likely attributed to the later initiation of ERT. Both twins safely tolerated ITI, achieving immune tolerance with low antibody titers. At 28 months, the twins transitioned to avalglucosidase-alfa (40 mg/kg every other week (EOW)), which was well tolerated without an increase in antibody titers. At 39 months, both twins exhibited normal cardiac function, LVMI, and biomarkers. Motor skills continued to improve, though some kinematic concerns persisted. These cases underscore the importance of early, high-dose ERT combined with ITI in managing CRIM-negative IOPD. While transitioning to avalglucosidase-alfa at 40 mg/kg/EOW was beneficial and well-tolerated in our patients, further studies are needed to confirm its long-term efficacy compared to the high-dose weekly 40 mg/kg alglucosidase-alfa.
Collapse
Affiliation(s)
- Angie H. Fares
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Ankit K. Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Laura E. Case
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
- Doctor of Physical Therapy Division, Department of Orthopaedics, Duke University Medical Center, Durham, North Carolina, USA
| | - Cassie Sharon
- Department of Rehabilitation Services, Pediatric Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy Klinepeter
- Department of Rehabilitation Services, Pediatric Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Amelia Kirby
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Matthew T. Lisi
- Department of Pediatrics, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Rebecca L. Koch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
3
|
Moschetti M, Lo Curto A, Giacomarra M, Francofonte D, Zizzo C, Messina E, Duro G, Colomba P. Mutation Spectrum of GAA Gene in Pompe Disease: Current Knowledge and Results of an Italian Study. Int J Mol Sci 2024; 25:9139. [PMID: 39273088 PMCID: PMC11394944 DOI: 10.3390/ijms25179139] [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] [Received: 06/12/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 09/15/2024] Open
Abstract
Studying a patient with Pompe disease (PD) is like opening Pandora's box. The specialist is faced with numerous clinical features similar to those of several diseases, and very often the symptoms are well hidden and none is associated with this rare disease. In recent years, scientific interest in this disease has been growing more and more, but still no symptom is recognized as key to a correct diagnosis of it, nor is there any specific disease marker to date. New diagnostic/therapeutic proposals on disease allow for the diffusion of knowledge of this pathology for timely diagnosis of the patient. Due to unawareness and difficulty in diagnosis, many adults with PD are diagnosed with great delay. In this article, we report and discuss current knowledge of PD and provide new data from work conducted on a cohort of 2934 Italian subjects recruited in recent years. A genetic analysis of the GAA gene was performed on patients with significant clinical signs and pathological enzyme activity to define the genetic profile of subjects. This identified 39 symptomatic PD subjects with low acid alpha-glucosidase enzyme activity and the presence of two causative mutations in GAA gene regions. Furthermore, 22 subjects with genetic variants of uncertain significance (GVUS) were identified.
Collapse
Affiliation(s)
- Marta Moschetti
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Alessia Lo Curto
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Miriam Giacomarra
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Daniele Francofonte
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Carmela Zizzo
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Elisa Messina
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Giovanni Duro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Paolo Colomba
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| |
Collapse
|
4
|
İnci A, Ezgü FS, Tümer L. Advances in Immune Tolerance Induction in Enzyme Replacement Therapy. Paediatr Drugs 2024; 26:287-308. [PMID: 38664313 PMCID: PMC11074017 DOI: 10.1007/s40272-024-00627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 05/07/2024]
Abstract
Inborn errors of metabolism (IEMs) are a group of genetic diseases that occur due to the either deficiency of an enzyme involved in a metabolic/biochemical pathway or other disturbances in the metabolic pathway including transport protein or activator protein deficiencies, cofactor deficiencies, organelle biogenesis, maturation or trafficking problems. These disorders are collectively significant due to their substantial impact on both the well-being and survival of affected individuals. In the quest for effective treatments, enzyme replacement therapy (ERT) has emerged as a viable strategy for patients with many of the lysosomal storage disorders (LSD) and enzyme substitution therapy in the rare form of the other inborn errors of metabolism including phenylketonuria and hypophosphatasia. However, a major challenge associated with enzyme infusion in patients with these disorders, mainly LSD, is the development of high antibody titres. Strategies focusing on immunomodulation have shown promise in inducing immune tolerance to ERT, leading to improved overall survival rates. The implementation of immunomodulation concurrent with ERT administration has also resulted in a decreased occurrence of IgG antibody development compared with cases treated solely with ERT. By incorporating the knowledge gained from current approaches and analysing the outcomes of immune tolerance induction (ITI) modalities from clinical and preclinical trials have demonstrated significant improvement in the efficacy of ERT. In this comprehensive review, the progress in ITI modalities is assessed, drawing insights from both clinical and preclinical trials. The focus is on evaluating the advancements in ITI within the context of IEM, specifically addressing LSDs managed through ERT.
Collapse
Affiliation(s)
- Aslı İnci
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey.
| | - Fatih Süheyl Ezgü
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
- Department of Paediatric Genetic, Gazi University School of Medicine, Ankara, Turkey
| | - Leyla Tümer
- Department of Paediatric Metabolism and Nutrition, Gazi University School of Medicine, Emniyet Street, Yenimahalle, Ankara, Turkey
| |
Collapse
|
5
|
Kumar V, Barwal A, Sharma N, Mir DS, Kumar P, Kumar V. Therapeutic proteins: developments, progress, challenges, and future perspectives. 3 Biotech 2024; 14:112. [PMID: 38510462 PMCID: PMC10948735 DOI: 10.1007/s13205-024-03958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
Proteins are considered magic molecules due to their enormous applications in the health sector. Over the past few decades, therapeutic proteins have emerged as a promising treatment option for various diseases, particularly cancer, cardiovascular disease, diabetes, and others. The formulation of protein-based therapies is a major area of research, however, a few factors still hinder the large-scale production of these therapeutic products, such as stability, heterogenicity, immunogenicity, high cost of production, etc. This review provides comprehensive information on various sources and production of therapeutic proteins. The review also summarizes the challenges currently faced by scientists while developing protein-based therapeutics, along with possible solutions. It can be concluded that these proteins can be used in combination with small molecular drugs to give synergistic benefits in the future.
Collapse
Affiliation(s)
- Vimal Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Arti Barwal
- Department of Microbial Biotechnology, Panjab University, South Campus, Sector-25, Chandigarh, 160014 India
| | - Nitin Sharma
- Department of Biotechnology, Chandigarh Group of Colleges, Mohali, Punjab 140307 India
| | - Danish Shafi Mir
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| | - Pradeep Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, 173229 India
| | - Vikas Kumar
- University Institute of Biotechnology, Chandigarh University, Gharuan, Mohali, Punjab 140413 India
| |
Collapse
|
6
|
Desai AK, Shrivastava G, Grant CL, Wang RY, Burt TD, Kishnani PS. An updated management approach of Pompe disease patients with high-sustained anti-rhGAA IgG antibody titers: experience with bortezomib-based immunomodulation. Front Immunol 2024; 15:1360369. [PMID: 38524130 PMCID: PMC10959098 DOI: 10.3389/fimmu.2024.1360369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction High sustained anti-rhGAA antibody titers (HSAT; ≥12,800) are directly linked to reduced efficacy of enzyme replacement therapy (ERT) and subsequent clinical deterioration in infantile-onset Pompe disease (IOPD). We have previously demonstrated the safety and effectiveness of a bortezomib-based immune-tolerance induction (ITI) regimen (bortezomib, rituximab, methotrexate, and IVIG) in eliminating HSAT. Methods Here, we describe two IOPD cases (patients 6 and 8) who developed HSAT at 8 and 10 weeks on ERT despite transient low-dose methotrexate ITI administration in the ERT-naïve setting and were treated with a bortezomib-based ITI regimen, and we compare their courses to a series of six historical patients (patients 1-5, and 7) with a similar presentation who exemplify our evolving approach to treatment. Results In total, patients 6 and 8 received 16 and 8 doses of bortezomib (4 doses=1 cycle) respectively reducing titers from 25,600 to seronegative, but differences in the course of their therapy were instructive regarding the optimal approach to initial treatment of HSAT; specifically, patient 6 was treated initially with only a single course of bortezomib rescue therapy, while patient 8 received two back-to-back courses. Patient 8 received IVIG therapy throughout the immunosuppression whereas patient 6 received IVIG therapy and was switched to subcutaneous IgG replacement. Patient 6 had a transient reduction in anti-rhGAA antibodies, after receiving a single initial cycle of bortezomib, but had a recurrence of high anti-rhGAA antibody titer after 160 weeks that required 3 additional cycles of bortezomib to ultimately achieve tolerance. In contrast, patient 8 achieved tolerance after being given two consecutive cycles of bortezomib during their initial treatment and had B cell recovery by week 54. Since the reduction in anti-rhGAA antibodies, both patients are doing well clinically, and have decreasing ALT, AST, and CK. No major infections leading to interruption of treatment were observed in either patient. The bortezomib-based ITI was safe and well-tolerated, and patients continue to receive ERT at 40 mg/kg/week. Discussion These case studies and our previous experience suggest that to achieve an effective reduction of anti-rhGAA antibodies in the setting of HSAT, bortezomib should be initiated at the earliest sign of high anti-rhGAA antibodies with a minimum of two consecutive cycles as shown in the case of patient 8. It is important to note that, despite initiation of ERT at age 2.3 weeks, patient 8 quickly developed HSAT. We recommend close monitoring of anti-rhGAA antibodies and early intervention with ITI as soon as significantly elevated anti-rhGAA antibody titers are noted.
Collapse
Affiliation(s)
- Ankit K. Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Garima Shrivastava
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Christina L. Grant
- Division of Genetics and Metabolism, Children’s National Hospital, Washington, DC, United States
| | - Raymond Y. Wang
- Division of Metabolic Disorders, Children’s Hospital of Orange County, Orange, CA, United States
- Department of Pediatrics, University of California-Irvine School of Medicine, Orange, CA, United States
| | - Trevor D. Burt
- Division of Neonatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Children’s Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, United States
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
7
|
Desai AK, Smith PB, Yi JS, Rosenberg AS, Burt TD, Kishnani PS. Immunophenotype associated with high sustained antibody titers against enzyme replacement therapy in infantile-onset Pompe disease. Front Immunol 2024; 14:1301912. [PMID: 38250073 PMCID: PMC10798041 DOI: 10.3389/fimmu.2023.1301912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction The efficacy of enzyme replacement therapy (ERT) with alglucosidase alfa for infantile-onset Pompe disease (IOPD) is limited in some patients due to the development of high and sustained antibody titers (HSAT; ≥12,800). Methods We carried out detailed immunophenotyping of IOPD patients (n=40), including analysis of circulating cell populations by flow cytometry and plasma cytokines by multiplex array, to determine whether patients with HSAT have unique immunological characteristics compared to those with low titers (LT; <12,800). Results Compared to patients with LT, patients who develop HSAT were skewed toward a type 2 immune profile, with an increased frequency of Th2 cells that was positively correlated with levels of Th2 (IL-4, IL-5, IL-13) and pro-inflammatory (IL-6, TNF-α, MIP-1α, MIP-1β) cytokines. B cells were increased in HSAT patients with a decreased fraction of unswitched memory B cells. Plasma GM-CSF concentrations were lower on average in HSAT patients, while CXCL11 was elevated. Finally, using principal components analysis, we derived an HSAT Signature Score that successfully stratified patients according to their antibody titers. Discussion The immune profiles revealed in this study not only identify potential biomarkers of patients that developed HSAT but also provide insights into the pathophysiology of HSAT that will ultimately lead to improved immunotherapy strategies.
Collapse
Affiliation(s)
- Ankit K. Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - P. Brian Smith
- Division of Neonatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Duke Clinical Research Institute, Durham, NC, United States
| | - John S. Yi
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | | | - Trevor D. Burt
- Division of Neonatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
- Children’s Health and Discovery Initiative, Duke University School of Medicine, Durham, NC, United States
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| |
Collapse
|
8
|
Noel JC, Lagassé D, Golding B, Sauna ZE. Emerging approaches to induce immune tolerance to therapeutic proteins. Trends Pharmacol Sci 2023; 44:1028-1042. [PMID: 37903706 DOI: 10.1016/j.tips.2023.10.002] [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] [Received: 09/25/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 11/01/2023]
Abstract
Immunogenicity affects the safety and efficacy of therapeutic proteins. This review is focused on approaches for inducing immunological tolerance to circumvent the immunogenicity of therapeutic proteins in the clinic. The few immune tolerance strategies that are used in the clinic tend to be inefficient and expensive and typically involve global immunosuppression, putting patients at risk of infections. The hallmark of a desirable immune tolerance regimen is the specific alleviation of immune responses to the therapeutic protein. In the past decade, proof-of-principle studies have demonstrated that emerging technologies, including nanoparticle-based delivery of immunomodulators, cellular targeting and depletion, cellular engineering, gene therapy, and gene editing, can be leveraged to promote tolerance to therapeutic proteins. We discuss the potential of these novel approaches and the barriers that need to be overcome for translation into the clinic.
Collapse
Affiliation(s)
- Justine C Noel
- Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Daniel Lagassé
- Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Basil Golding
- Division of Plasma Derivatives, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Zuben E Sauna
- Division of Hemostasis, Office of Plasma Protein Therapeutics, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
9
|
Herzeg A, Borges B, Lianoglou BR, Gonzalez-Velez J, Canepa E, Munar D, Young SP, Bali D, Gelb MH, Chakraborty P, Kishnani PS, Harmatz P, Cohen JL, MacKenzie TC. Intrauterine enzyme replacement therapies for lysosomal storage disorders: Current developments and promising future prospects. Prenat Diagn 2023; 43:1638-1649. [PMID: 37955580 PMCID: PMC11155627 DOI: 10.1002/pd.6460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023]
Abstract
Lysosomal storage disorders (LSDs) are a group of monogenic condition, with many characterized by an enzyme deficiency leading to the accumulation of an undegraded substrate within the lysosomes. For those LSDs, postnatal enzyme replacement therapy (ERT) represents the standard of care, but this treatment has limitations when administered only postnatally because, at that point, prenatal disease sequelae may be irreversible. Furthermore, most forms of ERT, specifically those administered systemically, are currently unable to access certain tissues, such as the central nervous system (CNS), and furthermore, may initiate an immune response. In utero enzyme replacement therapy (IUERT) is a novel approach to address these challenges evaluated in a first-in-human clinical trial for IUERT in LSDs (NCT04532047). IUERT has numerous advantages: in-utero intervention may prevent early pathology; the CNS can be accessed before the blood-brain barrier forms; and the unique fetal immune system enables exposure to new proteins with the potential to prevent an immune response and may induce sustained tolerance. However, there are challenges and limitations for any fetal procedure that involves two patients. This article reviews the current state of IUERT for LSDs, including its advantages, limitations, and potential future directions for definitive therapies.
Collapse
Affiliation(s)
- Akos Herzeg
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
| | - Beltran Borges
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
| | - Billie R. Lianoglou
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
| | - Juan Gonzalez-Velez
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, California, USA
| | - Emma Canepa
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
| | - Dane Munar
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
| | - Sarah P. Young
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, North Carolina, USA
| | - Deeksha Bali
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, North Carolina, USA
| | - Michel H. Gelb
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Pranesh Chakraborty
- Department of Pediatrics, Children’s Hospital of Eastern Ontario and University of Ottawa, Ottawa, Ontario, Canada
| | - Priya S. Kishnani
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, North Carolina, USA
| | - Paul Harmatz
- Benioff Children’s Hospital, University of California, San Francisco, California, USA
| | - Jennifer L. Cohen
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, North Carolina, USA
| | - Tippi C. MacKenzie
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, California, USA
- Department of Surgery, University of California, San Francisco, California, USA
- Benioff Children’s Hospital, University of California, San Francisco, California, USA
| |
Collapse
|
10
|
Hannah WB, Derks TGJ, Drumm ML, Grünert SC, Kishnani PS, Vissing J. Glycogen storage diseases. Nat Rev Dis Primers 2023; 9:46. [PMID: 37679331 DOI: 10.1038/s41572-023-00456-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 09/09/2023]
Abstract
Glycogen storage diseases (GSDs) are a group of rare, monogenic disorders that share a defect in the synthesis or breakdown of glycogen. This Primer describes the multi-organ clinical features of hepatic GSDs and muscle GSDs, in addition to their epidemiology, biochemistry and mechanisms of disease, diagnosis, management, quality of life and future research directions. Some GSDs have available guidelines for diagnosis and management. Diagnostic considerations include phenotypic characterization, biomarkers, imaging, genetic testing, enzyme activity analysis and histology. Management includes surveillance for development of characteristic disease sequelae, avoidance of fasting in several hepatic GSDs, medically prescribed diets, appropriate exercise regimens and emergency letters. Specific therapeutic interventions are available for some diseases, such as enzyme replacement therapy to correct enzyme deficiency in Pompe disease and SGLT2 inhibitors for neutropenia and neutrophil dysfunction in GSD Ib. Progress in diagnosis, management and definitive therapies affects the natural course and hence morbidity and mortality. The natural history of GSDs is still being described. The quality of life of patients with these conditions varies, and standard sets of patient-centred outcomes have not yet been developed. The landscape of novel therapeutics and GSD clinical trials is vast, and emerging research is discussed herein.
Collapse
Affiliation(s)
- William B Hannah
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| | - Terry G J Derks
- Division of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Sarah C Grünert
- Department of General Paediatrics, Adolescent Medicine and Neonatology, Medical Centre-University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Paediatrics, Duke University Medical Center, Durham, NC, USA
| | - John Vissing
- Copenhagen Neuromuscular Center, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
11
|
Gómez-Cebrián N, Gras-Colomer E, Poveda Andrés JL, Pineda-Lucena A, Puchades-Carrasco L. Omics-Based Approaches for the Characterization of Pompe Disease Metabolic Phenotypes. BIOLOGY 2023; 12:1159. [PMID: 37759559 PMCID: PMC10525434 DOI: 10.3390/biology12091159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Lysosomal storage disorders (LSDs) constitute a large group of rare, multisystemic, inherited disorders of metabolism, characterized by defects in lysosomal enzymes, accessory proteins, membrane transporters or trafficking proteins. Pompe disease (PD) is produced by mutations in the acid alpha-glucosidase (GAA) lysosomal enzyme. This enzymatic deficiency leads to the aberrant accumulation of glycogen in the lysosome. The onset of symptoms, including a variety of neurological and multiple-organ pathologies, can range from birth to adulthood, and disease severity can vary between individuals. Although very significant advances related to the development of new treatments, and also to the improvement of newborn screening programs and tools for a more accurate diagnosis and follow-up of patients, have occurred over recent years, there exists an unmet need for further understanding the molecular mechanisms underlying the progression of the disease. Also, the reason why currently available treatments lose effectiveness over time in some patients is not completely understood. In this scenario, characterization of the metabolic phenotype is a valuable approach to gain insights into the global impact of lysosomal dysfunction, and its potential correlation with clinical progression and response to therapies. These approaches represent a discovery tool for investigating disease-induced modifications in the complete metabolic profile, including large numbers of metabolites that are simultaneously analyzed, enabling the identification of novel potential biomarkers associated with these conditions. This review aims to highlight the most relevant findings of recently published omics-based studies with a particular focus on describing the clinical potential of the specific metabolic phenotypes associated to different subgroups of PD patients.
Collapse
Affiliation(s)
- Nuria Gómez-Cebrián
- Drug Discovery Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Elena Gras-Colomer
- Pharmacy Department, Hospital Manises of Valencia, 46940 Valencia, Spain
| | | | - Antonio Pineda-Lucena
- Molecular Therapeutics Program, Centro de Investigación Médica Aplicada, 31008 Pamplona, Spain
| | | |
Collapse
|
12
|
Cohen JL, Chakraborty P, Fung-Kee-Fung K, Schwab ME, Bali D, Young SP, Gelb MH, Khaledi H, DiBattista A, Smallshaw S, Moretti F, Wong D, Lacroix C, El Demellawy D, Strickland KC, Lougheed J, Moon-Grady A, Lianoglou BR, Harmatz P, Kishnani PS, MacKenzie TC. In Utero Enzyme-Replacement Therapy for Infantile-Onset Pompe's Disease. N Engl J Med 2022; 387:2150-2158. [PMID: 36351280 PMCID: PMC10794051 DOI: 10.1056/nejmoa2200587] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Patients with early-onset lysosomal storage diseases are ideal candidates for prenatal therapy because organ damage starts in utero. We report the safety and efficacy results of in utero enzyme-replacement therapy (ERT) in a fetus with CRIM (cross-reactive immunologic material)-negative infantile-onset Pompe's disease. The family history was positive for infantile-onset Pompe's disease with cardiomyopathy in two previously affected deceased siblings. After receiving in utero ERT and standard postnatal therapy, the current patient had normal cardiac and age-appropriate motor function postnatally, was meeting developmental milestones, had normal biomarker levels, and was feeding and growing well at 13 months of age.
Collapse
Affiliation(s)
- Jennifer L Cohen
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Pranesh Chakraborty
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Karen Fung-Kee-Fung
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Marisa E Schwab
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Deeksha Bali
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Sarah P Young
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Michael H Gelb
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Hamid Khaledi
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Alicia DiBattista
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Stacey Smallshaw
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Felipe Moretti
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Derek Wong
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Catherine Lacroix
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Dina El Demellawy
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Kyle C Strickland
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Jane Lougheed
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Anita Moon-Grady
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Billie R Lianoglou
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Paul Harmatz
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Priya S Kishnani
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| | - Tippi C MacKenzie
- From the Department of Pediatrics, Division of Medical Genetics (J.L.C., D.B., S.P.Y., P.S.K.), and the Department of Pathology (K.C.S.), Duke University, Durham, NC; the Department of Pediatrics, Children's Hospital of Eastern Ontario and University of Ottawa (P.C., S.S., D.W., C.L., D.E.D., J.L.), the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Ottawa Hospital, University of Ottawa (K.F.-K.-F., F.M.), and Children's Hospital of Eastern Ontario Research Institute (P.C., A.D.) - all in Ottawa; the University of California, San Francisco (UCSF) Benioff Children's Hospital and the UCSF Center for Maternal-Fetal Precision Medicine, San Francisco (M.E.S., A.M.-G., B.R.L., P.H., T.C.M.); and the Department of Chemistry, University of Washington, Seattle (M.H.G., H.K.)
| |
Collapse
|
13
|
Rana J, Muñoz MM, Biswas M. Oral tolerance to prevent anti-drug antibody formation in protein replacement therapies. Cell Immunol 2022; 382:104641. [PMID: 36402002 PMCID: PMC9730862 DOI: 10.1016/j.cellimm.2022.104641] [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: 09/21/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022]
Abstract
Protein based therapeutics have successfully improved the quality of life for patients of monogenic disorders like hemophilia, Pompe and Fabry disease. However, a significant proportion of patients develop immune responses towards intravenously infused therapeutic protein, which can complicate or neutralize treatment and compromise patient safety. Strategies aimed at circumventing immune responses following therapeutic protein infusion can greatly improve therapeutic efficacy. In recent years, antigen-based oral tolerance induction has shown promising results in the prevention and treatment of autoimmune diseases, food allergies and can prevent anti-drug antibody formation to protein replacement therapies. Oral tolerance exploits regulatory mechanisms that are initiated in the gut associated lymphoid tissue (GALT) to promote active suppression of orally ingested antigen. In this review, we outline general perceptions and current knowledge about the mechanisms of oral tolerance, including tissue specific sites of tolerance induction and the cells involved, with emphasis on antigen presenting cells and regulatory T cells. We define several factors, such as cytokines and metabolites that impact the stability and expansion potential of these immune modulatory cells. We highlight preclinical studies that have been performed to induce oral tolerance to therapeutic proteins or enzymes for single gene disorders, such as hemophilia or Pompe disease. These studies mainly utilize a transgenic plant-based system for oral delivery of antigen in conjugation with fusion protein technology that favors the prevention of antigen degradation in the stomach while enhancing uptake in the small intestine by antigen presenting cells and regulatory T cell induction, thereby promoting antigen specific systemic tolerance.
Collapse
Affiliation(s)
- Jyoti Rana
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maite Melero Muñoz
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
14
|
Yang CF, Liao TWE, Chu YL, Chen LZ, Huang LY, Yang TF, Ho HC, Kao SM, Niu DM. Long-term outcomes of very early treated infantile-onset Pompe disease with short-term steroid premedication: experiences from a nationwide newborn screening programme. J Med Genet 2022; 60:430-439. [PMID: 36137614 DOI: 10.1136/jmg-2022-108675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Starting enzyme replacement therapy (ERT) before severe irreversible muscular damage occurs is important in infantile-onset Pompe disease (IOPD). This long-term follow-up study demonstrates our diagnostic and treatment strategies for IOPD and compares our clinical outcomes with those of other medical centres. METHODS In this long-term follow-up study, we analysed the outcomes of very early ERT with premedication hydrocortisone in patients with IOPD. Out of 1 228 539 infants screened between 1 January 2010 and 28 February 2021, 33 newborns had confirmed IOPD in Taipei Veterans General Hospital. Twenty-six were regularly treated and monitored at Taipei Veterans General Hospital. Echocardiographic parameters, biomarkers, IgG antibodies against alglucosidase alpha, pulmonary function variables and developmental status were all assessed regularly over an average follow-up duration of 6.18±3.14 years. We compared the long-term treatment outcomes of our patients with those of other research groups. RESULTS The average age at ERT initiation was 9.75±3.17 days for patients with classic IOPD. The average of the latest antialglucosidase alpha IgG titre was 669.23±1159.23. All enrolled patients had normal heart sizes, motor milestones, cognitive function and pulmonary function that were near-normal to normal. Compared with patients in other studies, our patients had better outcomes in all aspects. CONCLUSION Very early ERT using our rapid diagnostic and treatment strategy enabled our patients with IOPD to have better outcomes than patients in other medical centres.
Collapse
Affiliation(s)
- Chia-Feng Yang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ting-Wei Ernie Liao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Ling Chu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Li-Zhen Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ling-Yi Huang
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Internal Medicine, Division of Nephrology, Taipei City Hospital Heping Fuyou Branch, Taipei, Taiwan
| | - Tsui-Feng Yang
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hui-Chen Ho
- Taipei Institute of Pathology, Neonatal Screening Center, Taipei, Taiwan
| | - Shu-Min Kao
- Chinese Foundation of Health, Neonatal Screening Center, Taipei, Taiwan
| | - Dau-Ming Niu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan .,Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
15
|
Curelaru S, Desai AK, Fink D, Zehavi Y, Kishnani PS, Spiegel R. A favorable outcome in an infantile-onset Pompe patient with cross reactive immunological material (CRIM) negative disease with high dose enzyme replacement therapy and adjusted immunomodulation. Mol Genet Metab Rep 2022; 32:100893. [PMID: 35813979 PMCID: PMC9263520 DOI: 10.1016/j.ymgmr.2022.100893] [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: 05/07/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 11/30/2022] Open
Abstract
Infantile onset Pompe disease (IOPD) is a rare devastating disease that presents in early infancy with rapidly progressive hypertrophic cardiomyopathy, severe generalized myopathy and death within the first year of life. The emergence of enzyme replacement therapy (ERT) with recombinant human acid alpha glucosidase (rhGAA) has improved the natural course of IOPD with a significant impact on cardiomyopathy but has a more limited effect on the progression of myopathy and consequently the later deterioration of the disease. Possible reasons for reduced ERT efficacy include insufficient enzyme, partial targeting of skeletal muscle and the development of IgG rhGAA antibodies especially in patients who are cross-reactive immunological material (CRIM) negative. We report a CRIM-negative IOPD female patient who started treatment upon diagnosis at 4.5 months with ERT at 20 mg/kg every other week and a course of combined immunomodulation with rituximab, methotrexate and IVIG according to the published Duke protocol and increased ERT within a month to 40 mg/kg/week. Despite initial good clinical response to ERT and immunomodulation, monthly monitoring identified a gradual increase of serum antibody titers to rhGAA necessitating a second course of immunomodulation with bortezomib and maintenance rituximab and methotrexate. A gradual reduction in frequency of immunotherapy was instituted and over a period of 14 months was discontinued. Serum anti-rhGAA antibody titers remained negative for 5 months since cessation of immunomodulation and the patient is now immune tolerant with recovery of CD19. At the age of 30 months the patient is walking independently and has normal cardiac function and anatomy. We recommend initiating ERT at 40 mg/kg/week in CRIM-negative IOPD patients, concomitant with immunomodulation and monthly monitoring of serum anti-rhGAA IgG titers upon confirmation of the diagnosis.
Collapse
Affiliation(s)
- Shiri Curelaru
- Department of Pediatrics B, Emek Medical Center, Afula, Israel
| | - Ankit K. Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - Daniel Fink
- Pediatric Cardiology Unit, Emek Medical Center, Afula, Israel
| | - Yoav Zehavi
- Department of Pediatrics B, Emek Medical Center, Afula, Israel
- Rappaport School of Medicine, Technion, Haifa, Israel
| | - Priya S. Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Health System, Durham, NC, United States
| | - Ronen Spiegel
- Department of Pediatrics B, Emek Medical Center, Afula, Israel
- Rappaport School of Medicine, Technion, Haifa, Israel
- Corresponding author at: Department of Pediatrics B, Emek Medical Center, Afula 1834111, Israel.
| |
Collapse
|
16
|
Gragnaniello V, Deodato F, Gasperini S, Donati MA, Canessa C, Fecarotta S, Pascarella A, Spadaro G, Concolino D, Burlina A, Parenti G, Strisciuglio P, Fiumara A, Casa RD. Immune responses to alglucosidase in infantile Pompe disease: recommendations from an Italian pediatric expert panel. Ital J Pediatr 2022; 48:41. [PMID: 35248118 PMCID: PMC8898438 DOI: 10.1186/s13052-022-01219-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background Classic infantile onset of Pompe disease (c-IOPD) leads to hypotonia and hypertrophic cardiomyopathy within the first days to weeks of life and, without treatment, patients die of cardiorespiratory failure in their first 1–2 years of life. Enzymatic replacement therapy (ERT) with alglucosidase alfa is the only available treatment, but adverse immune reactions can reduce ERT’s effectiveness and safety. It is therefore very important to identify strategies to prevent and manage these complications. Several articles have been written on this disease over the last 10 years, but no univocal indications have been established. Methods Our study presents a review of the current literature on management of immune responses to ERT in c-IOPD as considered by an Italian study group of pediatric metabolists and immunologists in light of our shared patient experience. Results We summarize the protocols for the management of adverse reactions to ERT, analyzing their advantages and disadvantages, and provide expert recommendations for their optimal management, to the best of current knowledge. However, further studies are needed to improve actual management protocols, which still have several limitations.
Collapse
|
17
|
van Kooten HA, Ditters IAM, Hoogeveen-Westerveld M, Jacobs EH, van den Hout JMP, van Doorn PA, Pijnappel WWMP, van der Ploeg AT, van der Beek NAME. Antibodies against recombinant human alpha-glucosidase do not seem to affect clinical outcome in childhood onset Pompe disease. Orphanet J Rare Dis 2022; 17:31. [PMID: 35109913 PMCID: PMC8812154 DOI: 10.1186/s13023-022-02175-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/16/2022] [Indexed: 01/16/2023] Open
Abstract
Background Enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA, alglucosidase alfa) has improved survival, motor outcomes, daily life activity and quality of life in Pompe patients. However, ERT in Pompe disease often induces formation of antibodies, which may reduce the efficacy of treatment and can lead to adverse events. In this study antibody formation and their effect on clinical outcome in patients with childhood onset Pompe disease treated with enzyme replacement therapy (ERT) with recombinant human alpha-glucosidase (rhGAA) are analyzed. Methods Enzyme-linked immunosorbent assay (ELISA) was used to determine anti-rhGAA antibody titers at predefined time points. The effect of antibodies on rhGAA activity (neutralizing effects) was measured in vitro. Clinical effects were evaluated by assessing muscle strength (MRC score) and function (QMFT-score), pulmonary function and infusion associated reactions (IARs). Results Twenty-two patients were included (age at start ERT 1.1–16.4 years, median treatment duration 12.4 years). Peak antibody titers were low (< 1:1250) in 9%, intermediate (1:1250–1:31,250) in 68% and high (≥ 1:31250) in 23% of patients; three patients (14%) had more than one titer of ≥ 1:31,250. Four patients (18%) experienced IARs; two patients from the high titer group had 86% of all IARs. Inhibition of intracellular GAA activity (58%) in vitro was found in one sample. The clinical course did not appear to be influenced by antibody titers. Conclusions Ninety-one percent of childhood onset Pompe patients developed anti-rhGAA antibodies (above background level), a minority of whom had high antibody titers at repeated time points, which do not seem to interfere with clinical outcome. High antibody titers may be associated with the occurrence of IARs. Although the majority of patients does not develop high titers; antibody titers should be determined in case of clinical deterioration. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02175-2.
Collapse
Affiliation(s)
- Harmke A van Kooten
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Imke A M Ditters
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marianne Hoogeveen-Westerveld
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Edwin H Jacobs
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Johanna M P van den Hout
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Pieter A van Doorn
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Department of Clinical Genetics, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nadine A M E van der Beek
- Department of Neurology, Center for Lysosomal and Metabolic Diseases, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands. .,Department of Neurology, Erasmus University Medical Center, Mailbox 2040, 3000 CA, Rotterdam, the Netherlands.
| |
Collapse
|
18
|
Starosta RT, Hou YCC, Leestma K, Singh P, Viehl L, Manwaring L, Granadillo JL, Schroeder MC, Colombo JN, Whitehead H, Dickson PI, Hulbert ML, Nguyen HT. Infantile-onset Pompe disease complicated by sickle cell anemia: Case report and management considerations. Front Pediatr 2022; 10:944178. [PMID: 36245745 PMCID: PMC9555291 DOI: 10.3389/fped.2022.944178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Infantile-onset Pompe disease (IOPD) is a rare, severe disorder of lysosomal storage of glycogen that leads to progressive cardiac and skeletal myopathy. IOPD is a fatal disease in childhood unless treated with enzyme replacement therapy (ERT) from an early age. Sickle cell anemia (SCA) is a relatively common hemoglobinopathy caused by a specific variant in the hemoglobin beta-chain. Here we report a case of a male newborn of African ancestry diagnosed and treated for IOPD and SCA. Molecular testing confirmed two GAA variants, NM_000152.5: c.842G>C, p.(Arg281Pro) and NM_000152.5: c.2560C>T, p.(Arg854*) in trans, and homozygosity for the HBB variant causative of SCA, consistent with his diagnosis. An acute neonatal presentation of hypotonia and cardiomyopathy required ERT with alglucosidase alfa infusions preceded by immune tolerance induction (ITI), as well as chronic red blood cell transfusions and penicillin V potassium prophylaxis for treatment of IOPD and SCA. Clinical course was further complicated by multiple respiratory infections. We review the current guidelines and interventions taken to optimize his care and the pitfalls of those guidelines when treating patients with concomitant conditions. To the best of our knowledge, no other case reports of the concomitance of these two disorders was found. This report emphasizes the importance of newborn screening, early intervention, and treatment considerations for this complex patient presentation of IOPD and SCA.
Collapse
Affiliation(s)
- Rodrigo Tzovenos Starosta
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Ying-Chen Claire Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Katelyn Leestma
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Prapti Singh
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Luke Viehl
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Linda Manwaring
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Jorge Luis Granadillo
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Molly C Schroeder
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jamie N Colombo
- Division of Pediatric Cardiology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Halana Whitehead
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Patricia Irene Dickson
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Monica L Hulbert
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Hoanh Thi Nguyen
- Division of Clinical Genetics and Genomics, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Lagassé HAD, McCormick Q, Sauna ZE. Secondary failure: immune responses to approved protein therapeutics. Trends Mol Med 2021; 27:1074-1083. [PMID: 34493437 DOI: 10.1016/j.molmed.2021.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/23/2022]
Abstract
Recombinant therapeutic proteins are a broad class of biological products used to replace dysfunctional human proteins in individuals with genetic defects (e.g., factor VIII for hemophilia) or, in the case of monoclonal antibodies, bind to disease targets involved in cancers, autoimmune disorders, or other conditions. Unfortunately, immunogenicity (immune response to the drug) remains a key impediment, potentially affecting the safety and efficacy of these therapeutics. Immunogenicity risk is routinely evaluated during the licensure of therapeutic proteins. However, despite eliciting anti-drug immune responses in at least some patients, most protein drugs are nevertheless licensed as they address unmet medical needs. The pre-licensure immunogenicity assessments of therapeutic proteins are the subject of numerous reviews and white papers. However, observation and clinical management of the immunogenicity of approved therapeutic proteins face additional challenges. We survey the immunogenicity of approved therapeutic proteins, discuss the clinical management of immunogenicity, and identify the challenges to establishing clinically relevant immunogenicity assays for use in routine clinical practice.
Collapse
Affiliation(s)
- H A Daniel Lagassé
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Quinn McCormick
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Zuben E Sauna
- Hemostasis Branch, Division of Plasma Protein Therapeutics, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US Food and Drug Administration (FDA), Silver Spring, MD, USA.
| |
Collapse
|
20
|
De Groot AS, Desai AK, Lelias S, Miah SMS, Terry FE, Khan S, Li C, Yi JS, Ardito M, Martin WD, Kishnani PS. Immune Tolerance-Adjusted Personalized Immunogenicity Prediction for Pompe Disease. Front Immunol 2021; 12:636731. [PMID: 34220802 PMCID: PMC8242953 DOI: 10.3389/fimmu.2021.636731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 05/20/2021] [Indexed: 11/13/2022] Open
Abstract
Infantile-onset Pompe disease (IOPD) is a glycogen storage disease caused by a deficiency of acid alpha-glucosidase (GAA). Treatment with recombinant human GAA (rhGAA, alglucosidase alfa) enzyme replacement therapy (ERT) significantly improves clinical outcomes; however, many IOPD children treated with rhGAA develop anti-drug antibodies (ADA) that render the therapy ineffective. Antibodies to rhGAA are driven by T cell responses to sequences in rhGAA that differ from the individuals' native GAA (nGAA). The goal of this study was to develop a tool for personalized immunogenicity risk assessment (PIMA) that quantifies T cell epitopes that differ between nGAA and rhGAA using information about an individual's native GAA gene and their HLA DR haplotype, and to use this information to predict the risk of developing ADA. Four versions of PIMA have been developed. They use EpiMatrix, a computational tool for T cell epitope identification, combined with an HLA-restricted epitope-specific scoring feature (iTEM), to assess ADA risk. One version of PIMA also integrates JanusMatrix, a Treg epitope prediction tool to identify putative immunomodulatory (regulatory) T cell epitopes in self-proteins. Using the JanusMatrix-adjusted version of PIMA in a logistic regression model with data from 48 cross-reactive immunological material (CRIM)-positive IOPD subjects, those with scores greater than 10 were 4-fold more likely to develop ADA (p<0.03) than those that had scores less than 10. We also confirmed the hypothesis that some GAA epitopes are immunomodulatory. Twenty-one epitopes were tested, of which four were determined to have an immunomodulatory effect on T effector response in vitro. The implementation of PIMA V3J on a secure-access website would allow clinicians to input the individual HLA DR haplotype of their IOPD patient and the GAA pathogenic variants associated with each GAA allele to calculate the patient's relative risk of developing ADA, enhancing clinical decision-making prior to initiating treatment with ERT. A better understanding of immunogenicity risk will allow the implementation of targeted immunomodulatory approaches in ERT-naïve settings, especially in CRIM-positive patients, which may in turn improve the overall clinical outcomes by minimizing the development of ADA. The PIMA approach may also be useful for other types of enzyme or factor replacement therapies.
Collapse
Affiliation(s)
- Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ankit K Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | | | | | | | | | - Cindy Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - John S Yi
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | | | | | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
21
|
Li C, Desai AK, Gupta P, Dempsey K, Bhambhani V, Hopkin RJ, Ficicioglu C, Tanpaiboon P, Craigen WJ, Rosenberg AS, Kishnani PS. Transforming the clinical outcome in CRIM-negative infantile Pompe disease identified via newborn screening: the benefits of early treatment with enzyme replacement therapy and immune tolerance induction. Genet Med 2021; 23:845-855. [PMID: 33495531 PMCID: PMC8107133 DOI: 10.1038/s41436-020-01080-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/30/2022] Open
Abstract
Purpose: To assess the magnitude of benefit to early treatment initiation, enabled by newborn screening or prenatal diagnosis, in patients with cross-reactive immunological material (CRIM)-negative infantile Pompe disease (IPD), treated with enzyme replacement therapy (ERT) and prophylactic immune tolerance induction (ITI) with rituximab, methotrexate, and IVIG. Methods: A total of 41 CRIM-negative IPD patients were evaluated. Amongst patients who were treated with ERT+ITI (n=30), those who were invasive ventilator-free at baseline and had ≥6 months of follow-up were stratified based on age at treatment initiation: 1) early (≤4 weeks), 2) intermediate (>4 and ≤15 weeks), and 3) late (>15 weeks). A historical cohort of 11 CRIM-negative patients with IPD treated with ERT monotherapy served as an additional comparator group. Results: Twenty patients were included; five, seven, and eight in early, intermediate, and late treatment groups, respectively. Genotypes were similar across the three groups. Early-treated patients showed significant improvements in left ventricular mass index, motor and pulmonary outcomes, as well as biomarkers creatine kinase and urinary glucose tetrasaccharide, compared to those treated later. Conclusion: Our preliminary data suggest that early treatment with ERT+ITI can transform the long-term CRIM-negative IPD phenotype, which represents the most severe end of the Pompe disease spectrum.
Collapse
Affiliation(s)
- Cindy Li
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Ankit K Desai
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Punita Gupta
- St. Joseph's University Hospital, Paterson, NJ, USA
| | - Katherine Dempsey
- Center for Human Genetics and Department of Genetics and Genome Sciences, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH, USA
| | - Vikas Bhambhani
- Children's Hospitals and Clinics of Minnesota, Minneapolis, MN, USA
| | - Robert J Hopkin
- Division of Medical Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Can Ficicioglu
- The Children's Hospital of Philadelphia, Division of Genetics and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Pranoot Tanpaiboon
- Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Amy S Rosenberg
- Division of Biologics Review and Research 3, Office of Biotechnology Products, Center for Drug Evaluation and Research, US FDA, Bethesda, MD, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|