1
|
Xie SQ, Ding XF, Zhang B, Shi FX, Zhong LL, Huang H. [A case of progressive ossifying myositis caused by ACVR1 gene mutation]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:961-966. [PMID: 39267512 PMCID: PMC11404458 DOI: 10.7499/j.issn.1008-8830.2401060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
A 2-year-and-10-month-old boy presented with multiple masses in the neck and chest for over 3 months. The child had a history of unstable walking, with hard lumps visible at the injury sites after falls, which would resolve on their own. Following a recent injury, a mass was discovered in the posterior neck, protruding above the skin surface and accompanied by limited joint movement. Gradually, new masses were found on the left side of the neck, back near the scapular lower angle, in the scapular fossa, and along the left axillary midline. Magnetic resonance imaging examination showed diffuse low signal on T1-weighted images and high signal on T2-weighted images in the bilateral posterior neck and back muscles two months ago. A CT scan revealed muscle swelling, with areas of patchy low density and multiple nodular high-density ossifications within some muscles. Genetic testing results indicated a mutation in the ACVR1 gene, leading to the final diagnosis of progressive ossifying myositis in this patient. This article summarizes the etiology, diagnosis, and treatment of one case of progressive ossifying myositis, providing a reference for clinicians.
Collapse
Affiliation(s)
- Si-Qin Xie
- Department of Pediatrics, Hunan Normal University First Affiliated Hospital/Hunan Provincial People's Hospital, Changsha 410005, China
| | - Xiao-Fang Ding
- Department of Pediatrics, Hunan Normal University First Affiliated Hospital/Hunan Provincial People's Hospital, Changsha 410005, China
| | - Bing Zhang
- Department of Pediatrics, Hunan Normal University First Affiliated Hospital/Hunan Provincial People's Hospital, Changsha 410005, China
| | | | - Li-Li Zhong
- Department of Pediatrics, Hunan Normal University First Affiliated Hospital/Hunan Provincial People's Hospital, Changsha 410005, China
| | - Han Huang
- Department of Pediatrics, Hunan Normal University First Affiliated Hospital/Hunan Provincial People's Hospital, Changsha 410005, China
| |
Collapse
|
2
|
Wang Y, Nguyen JH, de Ruiter RD, Mendell J, Srinivasan D, Davis JD, Eekhoff EMW. Garetosmab in Fibrodysplasia Ossificans Progressiva: Clinical Pharmacology Results from the Phase 2 LUMINA-1 Trial. J Clin Pharmacol 2024; 64:264-274. [PMID: 37694449 DOI: 10.1002/jcph.2344] [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: 05/23/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
Here, we report the clinical pharmacology data from LUMINA-1 (NCT03188666), a Phase 2 trial that evaluated garetosmab (a monoclonal antibody against activin A) in patients with fibrodysplasia ossificans progressiva. Forty-four patients were randomly assigned to intravenous 10 mg/kg of garetosmab or placebo every 4 weeks in a double-blind 28-week treatment period, followed by a 28-week open-label treatment period with garetosmab, and subsequent open-label extension. Serum samples were obtained to assess pharmacokinetics (PK), immunogenicity, and bone morphogenetic protein 9 (BMP9). Comparative exposure-response analyses for efficacy and safety were performed with trough concentrations (Ctrough ) of garetosmab prior to dosing. Steady-state PK was reached 12-16 weeks after the first dose of garetosmab, with mean (standard deviation) Ctrough of 105 ± 30.8 mg/L. Immunogenicity assessments showed anti-garetosmab antibody formation in 1 patient (1/43; 2.3%); titers were low, and did not affect PK or clinical efficacy. Median concentrations of BMP9 in serum were approximately 40 pg/mL at baseline. There were no meaningful differences in PK or BMP9 concentration-time profiles between patients who did and did not experience epistaxis or death. The comparative exposure-response analyses demonstrated no association between Ctrough and efficacy or safety. PK findings were consistent with prior data in healthy volunteers and were typical for a monoclonal antibody administered at doses sufficient to saturate target-mediated clearance. There were no trends that suggested patients with higher serum exposures to garetosmab were more likely to experience a reduction in heterotopic ossification or adverse events. Garetosmab is being further evaluated in the Phase 3 OPTIMA trial.
Collapse
Affiliation(s)
- Yuhuan Wang
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Ruben D de Ruiter
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Vrije Universiteit, Amsterdam UMC Expert Center in Rare Bone Disease, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | | | | | | | - E Marelise W Eekhoff
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers (UMC), Vrije Universiteit, Amsterdam UMC Expert Center in Rare Bone Disease, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Srinivasan D, Arostegui M, Goebel EJ, Hart KN, Aykul S, Lees-Shepard JB, Idone V, Hatsell SJ, Economides AN. How Activin A Became a Therapeutic Target in Fibrodysplasia Ossificans Progressiva. Biomolecules 2024; 14:101. [PMID: 38254701 PMCID: PMC10813747 DOI: 10.3390/biom14010101] [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: 12/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by episodic yet cumulative heterotopic ossification (HO) of skeletal muscles, tendons, ligaments, and fascia. FOP arises from missense mutations in Activin Receptor type I (ACVR1), a type I bone morphogenetic protein (BMP) receptor. Although initial findings implicated constitutive activity of FOP-variant ACVR1 (ACVR1FOP) and/or hyperactivation by BMPs, it was later shown that HO in FOP requires activation of ACVR1FOP by Activin A. Inhibition of Activin A completely prevents HO in FOP mice, indicating that Activin A is an obligate driver of HO in FOP, and excluding a key role for BMPs in this process. This discovery led to the clinical development of garetosmab, an investigational antibody that blocks Activin A. In a phase 2 trial, garetosmab inhibited new heterotopic bone lesion formation in FOP patients. In contrast, antibodies to ACVR1 activate ACVR1FOP and promote HO in FOP mice. Beyond their potential clinical relevance, these findings have enhanced our understanding of FOP's pathophysiology, leading to the identification of fibroadipogenic progenitors as the cells that form HO, and the discovery of non-signaling complexes between Activin A and wild type ACVR1 and their role in tempering HO, and are also starting to inform biological processes beyond FOP.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Aris N. Economides
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA; (D.S.); (M.A.); (E.J.G.); (K.N.H.); (S.A.); (J.B.L.-S.); (V.I.); (S.J.H.)
| |
Collapse
|
4
|
Riege D, Herschel S, Fenkl T, Schade D. Small-Molecule Probes as Pharmacological Tools for the Bone Morphogenetic Protein Signaling Pathway. ACS Pharmacol Transl Sci 2023; 6:1574-1599. [PMID: 37974621 PMCID: PMC10644459 DOI: 10.1021/acsptsci.3c00170] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023]
Abstract
The bone morphogenetic protein (BMP) pathway is highly conserved and plays central roles in health and disease. The quality and quantity of its signaling outputs are regulated at multiple levels, offering pharmacological options for targeted modulation. Both target-centric and phenotypic drug discovery (PDD) approaches were applied to identify small-molecule BMP inhibitors and stimulators. In this Review, we accumulated and systematically classified the different reported chemotypes based on their targets as well as modes-of-action, and herein we illustrate the discovery history of selected candidates. A comprehensive summary of available biochemical, cellular, and in vivo activities is provided for the most relevant BMP modulators, along with recommendations on their preferred use as chemical probes to study BMP-related (patho)physiological processes. There are a number of high-quality probes used as BMP inhibitors that potently and selectively interrogate the kinase activities of distinct type I (16 chemotypes available) and type II receptors (3 chemotypes available). In contrast, only a few high-quality BMP stimulator modalities have been introduced to the field due to a lack of profound target knowledge. FK506-derived macrolides such as calcineurin-sparing FKBP12 inhibitors currently represent the best-characterized chemical tools for direct activation of BMP-SMAD signaling at the receptor level. However, several PDD campaigns succeeded in expanding the druggable space of BMP stimulators. Albeit the majority of them do not entirely fulfill the strict chemical probe criteria, many chemotypes exhibit unique and unrecognized mechanisms as pathway potentiators or synergizers, serving as valuable pharmacological tools for BMP perturbation.
Collapse
Affiliation(s)
- Daniel Riege
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Sven Herschel
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Teresa Fenkl
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Dennis Schade
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
- Partner
Site Kiel, DZHK, German Center for Cardiovascular
Research, 24105 Kiel, Germany
| |
Collapse
|
5
|
Marino R, Dube L, Ogier J, Le Quan Sang KH. The Pharmacokinetic Profile of Palovarotene: An Open-Label Phase I Trial Investigating the Effect of Food and Potential for Drug-Drug Interaction in Healthy Participants. Eur J Drug Metab Pharmacokinet 2023; 48:691-707. [PMID: 37804430 PMCID: PMC10624720 DOI: 10.1007/s13318-023-00856-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND AND OBJECTIVES Palovarotene is under development for the treatment of fibrodysplasia ossificans progressiva (FOP). The objectives of this study were to evaluate palovarotene pharmacokinetics under fed versus fasted conditions and its induction potential towards cytochrome P450 3A4 (CYP3A4) substrate, midazolam. METHODS In this phase I, open-label trial (NCT04829773), palovarotene pharmacokinetics were characterized after repeated once-daily dosing. In one cohort, healthy participants received three doses of palovarotene 20 mg on Days 1, 6, and 11, as whole capsules under fasted or fed conditions, or sprinkled on food under fed conditions. In another cohort, individuals received midazolam 2 mg on Days 1 and 15 and a daily dose of palovarotene 20 mg on Days 2-15. Palovarotene and midazolam pharmacokinetics, including area under the concentration-time curve from time zero to infinity (AUC(0-∞)) and maximum observed plasma drug concentration (Cmax), were assessed. Adverse events (AEs) were recorded. RESULTS Overall, 23 participants completed each part. Palovarotene Cmax and AUC(0-∞) increased by 16.5% and 39.7% under fed versus fasted conditions. Pharmacokinetics were comparable between the whole capsule and sprinkled on food, under fed conditions. Midazolam AUC(0-∞) and Cmax decreased by 13.3% and 9.7% upon palovarotene co-administration over 14 days, less than that required to be considered a weak CYP3A4 inducer. Plasma palovarotene exposures were comparable after single and multiple doses. No serious AEs were reported. CONCLUSIONS These data support palovarotene administration after a meal, as a whole capsule or sprinkled on food. Palovarotene at 20 mg/day is a not a clinical inducer of CYP3A4. These results provide insights into palovarotene pharmacokinetics, aiding optimization of administration for patients with FOP. CLINICAL TRIALS REGISTRATION NUMBER NCT04829773.
Collapse
Affiliation(s)
| | - Louise Dube
- Pleiades Consultation Inc., Phoenix, AZ, USA
| | | | - Kim-Hanh Le Quan Sang
- Département de Génétique Clinique, Hôpital Universitaire Necker-Enfants Malades, Institut Imagine, Université Paris Cité, Paris, France.
| |
Collapse
|
6
|
Upadhyay J, Iwasaka-Neder J, Golden E, Bixby S. Synthetic CT Assessment of Lesions in Children With Rare Musculoskeletal Diseases. Pediatrics 2023; 152:e2022061027. [PMID: 37416976 PMCID: PMC10389770 DOI: 10.1542/peds.2022-061027] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2023] [Indexed: 07/08/2023] Open
Abstract
Imaging modalities such as computed tomography (CT) are critical for monitoring musculoskeletal abnormalities in children with rare diseases. However, CT exposes patients to radiation, which limits its utility in the clinical setting, particularly during longitudinal evaluation. Synthetic CT is a novel, noncontrast, and rapid MRI method that can provide CT-like images without any radiation exposure and is easily performed in conjunction with traditional MRI, which detects soft-tissue and bone marrow abnormalities. To date, an evaluation of synthetic CT in pediatric patients with rare musculoskeletal diseases has been lacking. In this case series, the capability of synthetic CT to identify musculoskeletal lesions accurately in 2 rare disease patients is revealed. In Case 1, synthetic CT, in agreement with routine CT, identified an intraosseous lesion in the right femoral neck in a 16-year-old female with fibrous dysplasia, whereas standard-of-care MRIs additionally revealed mild surrounding edema-like bone marrow signal. For Case 2, synthetic CT applied to a 12-year-old female with fibrodysplasia ossificans progressiva revealed heterotopic ossification present along the cervical spine that had caused the fusion of multiple vertebrae. Our evaluation of synthetic CT offers important insights into the feasibility and utility of this methodology in children with rare diseases affecting the musculoskeletal system.
Collapse
Affiliation(s)
- Jaymin Upadhyay
- Departments of Anesthesiology, Critical Care and Pain Medicine
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | - Jade Iwasaka-Neder
- Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emma Golden
- Departments of Anesthesiology, Critical Care and Pain Medicine
| | - Sarah Bixby
- Radiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Lalonde RL, Nicolas HA, Cutler RS, Pantekidis I, Zhang W, Yelick PC. Functional comparison of human ACVR1 and zebrafish Acvr1l FOP-associated variants in embryonic zebrafish. Dev Dyn 2023; 252:605-628. [PMID: 36606464 PMCID: PMC10311797 DOI: 10.1002/dvdy.566] [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: 07/13/2021] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Fibrodysplasia ossificans progressiva (FOP), a rare disease characterized by progressive heterotopic ossification of muscle and connective tissues, is caused by autosomal dominant activating mutations in the type I receptor, ACVR1/ALK2. The classic human FOP variant, ACVR1R206H , shows increased bone morphogenetic protein (BMP) signaling and activation by activins. RESULTS Here, we performed in vivo functional characterization of human ACVR1R206H and orthologous zebrafish Acvr1lR203H using early embryonic zebrafish dorsoventral patterning as a phenotypic readout for receptor activity. Our results showed that human ACVR1R206H and zebrafish Acvr1lR203H exhibit functional differences in early embryonic zebrafish, and that human ACVR1R206H retained its signaling activity in the absence of a ligand-binding domain (LBD). We also showed, for the first time, that zebrafish Acvr2ba/Acvr2bb receptors are required for human ACVR1R206H signaling in early embryonic zebrafish. CONCLUSIONS Together, these data provide new insight into ACVR1R206H signaling pathways that may facilitate the design of new and effective therapies for FOP patients.
Collapse
Affiliation(s)
- Robert L. Lalonde
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, 136 Harrison Avenue, Boston, MA, USA 02111
| | - Hannah A. Nicolas
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON, Canada K1N 6N5
| | - Rowan S. Cutler
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, 136 Harrison Avenue, Boston, MA, USA 02111
| | - Irene Pantekidis
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, 136 Harrison Avenue, Boston, MA, USA 02111
| | - Weibo Zhang
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, 136 Harrison Avenue, Boston, MA, USA 02111
| | - Pamela C. Yelick
- Tufts University School of Dental Medicine, Division of Craniofacial and Molecular Genetics, 136 Harrison Avenue, Boston, MA, USA 02111
| |
Collapse
|
8
|
Chan JCK, Kuong EE, Chan JPK, Luk HM, Fung JLF, Tung JYL, Chung BHY. Fibrodysplasia ossificans progressiva in Hong Kong-A case report series. Front Pediatr 2023; 11:1152731. [PMID: 37181433 PMCID: PMC10167838 DOI: 10.3389/fped.2023.1152731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare condition. The diagnosis could be challenging due to its rarity and non-specific presenting symptoms. However, early diagnosis and appropriate management help in preserving patients' function and quality of life. Herein, we report the diagnostic journeys and clinical courses of 8 patients with FOP in Hong Kong and illustrate the challenges involved.
Collapse
Affiliation(s)
- Joshua Chun Ki Chan
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Evelyn Eugenie Kuong
- Skeletal Dysplasia Joint Clinic, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Duchess of Kent Children's Hospital, Hong Kong, Hong Kong SAR, China
| | - Joyce Pui Kwan Chan
- Skeletal Dysplasia Joint Clinic, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
- Department of Radiology, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
| | - Ho Ming Luk
- Clinical Genetics Service Unit, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
| | - Jasmine Lee Fong Fung
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Joanna Yuet-ling Tung
- Skeletal Dysplasia Joint Clinic, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
| | - Brian Hon Yin Chung
- Skeletal Dysplasia Joint Clinic, Hong Kong Children's Hospital, Hong Kong, Hong Kong SAR, China
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Farid A, Golden E, Robicheau S, Hu A, Cheung K, Yu PB, Rutkove SB, Upadhyay J. Diminished muscle integrity in patients with fibrodysplasia ossificans progressiva assessed with at-home electrical impedance myography. Sci Rep 2022; 12:20908. [PMID: 36463382 PMCID: PMC9719538 DOI: 10.1038/s41598-022-25610-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 12/01/2022] [Indexed: 12/05/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare disorder involving skeletal dysplasia and heterotopic ossification (HO) of muscle and connective tissue. We aimed to define a novel biomarker in FOP that enables reliable assessment of musculoskeletal tissue integrity. Considering logistical difficulties that FOP patients often face, our goal was to identify an at-home biomarker technique. Electrical impedance myography (EIM) is a non-invasive, portable method that can inform on muscle health. 15 FOP patients (age 10-52) and 13 healthy controls were assessed. Using EIM, multiple muscle groups were characterized per participant in a 45-min period. The Cumulative Analogue Joint Involvement Scale (CAJIS) was implemented to determine mobility burden severity. We additionally evaluated physical activity levels via a Patient-Reported Outcomes Measurement Information System (PROMIS)-based questionnaire. Relative to controls, FOP patients demonstrated significantly lower regional and whole-body phase values at 50 kHz and 100 kHz, indicating more diseased muscle tissue. Lower whole-body phase and reactance values, and higher resistance values, were associated with greater FOP burden (CAJIS score range: 4-30) and lower physical activity levels at 50 kHz and 100 kHz. This study points to the potential utility of EIM as a clinical biomarker tool capable of characterizing muscle integrity in FOP.
Collapse
Affiliation(s)
- Alexander Farid
- grid.38142.3c000000041936754XDepartment of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Emma Golden
- grid.38142.3c000000041936754XDepartment of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | | | - Alice Hu
- grid.492584.6Myolex Inc, Boston, MA USA
| | - Kin Cheung
- BioSAS Consulting, Inc., Wellesley, MA USA
| | - Paul B. Yu
- grid.38142.3c000000041936754XDivision of Cardiology, Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Seward B. Rutkove
- grid.239395.70000 0000 9011 8547Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA USA
| | - Jaymin Upadhyay
- grid.38142.3c000000041936754XDepartment of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDepartment of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| |
Collapse
|
10
|
Eekhoff EM, de Ruiter RD, Smilde BJ, Schoenmaker T, de Vries TJ, Netelenbos C, Hsiao EC, Scott C, Haga N, Grunwald Z, De Cunto CL, di Rocco M, Delai PLR, Diecidue RJ, Madhuri V, Cho TJ, Morhart R, Friedman CS, Zasloff M, Pals G, Shim JH, Gao G, Kaplan F, Pignolo RJ, Micha D. Gene Therapy for Fibrodysplasia Ossificans Progressiva: Feasibility and Obstacles. Hum Gene Ther 2022; 33:782-788. [PMID: 35502479 PMCID: PMC9419966 DOI: 10.1089/hum.2022.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/06/2022] [Indexed: 02/02/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare and devastating genetic disease, in which soft connective tissue is converted into heterotopic bone through an endochondral ossification process. Patients succumb early as they gradually become trapped in a second skeleton of heterotopic bone. Although the underlying genetic defect is long known, the inherent complexity of the disease has hindered the discovery of effective preventions and treatments. New developments in the gene therapy field have motivated its consideration as an attractive therapeutic option for FOP. However, the immune system's role in FOP activation and the as-yet unknown primary causative cell, are crucial issues which must be taken into account in the therapy design. While gene therapy offers a potential therapeutic solution, more knowledge about FOP is needed to enable its optimal and safe application.
Collapse
Affiliation(s)
- Elisabeth M.W. Eekhoff
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Ruben D. de Ruiter
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Bernard J. Smilde
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam, Vrije Universiteit, Amsterdam, The Netherlands
| | - Coen Netelenbos
- Section Endocrinology, Department of Internal Medicine, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, Institute for Human Genetics, Program in Craniofacial Biology, the Institute for Regeneration Medicine, University of California, San Francisco, California, USA
| | - Christiaan Scott
- Division of Paediatric Rheumatology, Department of Paediatrics and Child Heath, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa
| | - Nobuhiko Haga
- Rehabilitation Services Bureau, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Saitama, Japan
| | - Zvi Grunwald
- Department of Anesthesiology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, USA
| | - Carmen L. De Cunto
- Department of Pediatrics, Section Pediatric Rheumatology, Hospital Italiano Buenos Aires, Argentina
| | - Maja di Rocco
- Department of Pediatrics, Unit of Rare Diseases, IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Patricia L. R. Delai
- Teaching and Research Institute of the Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Robert J. Diecidue
- Department of Oral and Maxillofacial Surgery, Sidney Kimmel Medical College, Philadelphia, USA
| | - Vrisha Madhuri
- Department of Paediatric Orthopaedics and Center for Stem Cell Research, Christian Medical College and Hospital, Vellore, India
| | - Tae-Joon Cho
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Rolf Morhart
- Department of Pediatrics, Garmisch-Partenkichen Medical Center, Garmisch-Partenkirchen, Germany
| | - Clive S. Friedman
- Schulich School of Medicine and Dentistry, Western University, Clinical Skills Building, London, Ontario, Canada
| | - Michael Zasloff
- Surgery and Pediatrics, MedStar Georgetown Transplant Institute, Washington, District of Columbia, USA
| | - Gerard Pals
- Department of Human Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jae-Hyuck Shim
- Department of Medicine/Rheumatology, Horae Gene Therapy Center, Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Department of Microbiology and Physiological Systems, Horae Gene Therapy Center, Viral Vector Core, Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Frederick Kaplan
- Department of Orthopaedic Surgery and Medicine, Center for Research in FOP and Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Dimitra Micha
- Department of Human Genetics, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam Rare Bone Disease Center, Amsterdam Bone Center, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Nguyen MH, Atasoylu O, Wu L, Kapilashrami K, Pusey M, Gallagher K, Lai CT, Zhao P, Barbosa J, Liu K, He C, Zhang C, Styduhar ED, Witten MR, Chen Y, Lin L, Yang YO, Covington M, Diamond S, Yeleswaram S, Yao W. Discovery of Novel Pyrazolopyrimidines as Potent, Selective, and Orally Bioavailable Inhibitors of ALK2. ACS Med Chem Lett 2022; 13:1159-1164. [DOI: 10.1021/acsmedchemlett.2c00206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Minh H. Nguyen
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Onur Atasoylu
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Liangxing Wu
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Kanishk Kapilashrami
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Michelle Pusey
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Karen Gallagher
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Cheng-Tsung Lai
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Peng Zhao
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Joseph Barbosa
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Kai Liu
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Chunhong He
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Colin Zhang
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Evan D. Styduhar
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Michael R. Witten
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Yaoyu Chen
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Luping Lin
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Yan-ou Yang
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Maryanne Covington
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Sharon Diamond
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Swamy Yeleswaram
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| | - Wenqing Yao
- Incyte Research Institute, Incyte Corporation, 1801 Augustine Cut-Off, Wilmington, Delaware 19803, United States
| |
Collapse
|
12
|
Aykul S, Huang L, Wang L, Das NM, Reisman S, Ray Y, Zhang Q, Rothman NJ, Nannuru KC, Kamat V, Brydges S, Troncone L, Johnsen L, Yu PB, Fazio S, Lees-Shepard J, Schutz K, Murphy AJ, Economides AN, Idone V, Hatsell SJ. ACVR1 antibodies exacerbate heterotopic ossification in fibrodysplasia ossificans progressiva (FOP) by activating FOP-mutant ACVR1. J Clin Invest 2022; 132:153792. [PMID: 35511419 PMCID: PMC9197526 DOI: 10.1172/jci153792] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 04/28/2022] [Indexed: 11/21/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder whose most debilitating pathology is progressive and cumulative heterotopic ossification (HO) of skeletal muscles, ligaments, tendons, and fascia. FOP is caused by mutations in the type I BMP receptor gene ACVR1, which enable ACVR1 to utilize its natural antagonist, activin A, as an agonistic ligand. The physiological relevance of this property is underscored by the fact that HO in FOP is exquisitely dependent on activation of FOP-mutant ACVR1 by activin A, an effect countered by inhibition of anti–activin A via monoclonal antibody treatment. Hence, we surmised that anti-ACVR1 antibodies that block activation of ACVR1 by ligands should also inhibit HO in FOP and provide an additional therapeutic option for this condition. Therefore, we generated anti-ACVR1 monoclonal antibodies that block ACVR1’s activation by its ligands. Surprisingly, in vivo, these anti-ACVR1 antibodies stimulated HO and activated signaling of FOP-mutant ACVR1. This property was restricted to FOP-mutant ACVR1 and resulted from anti-ACVR1 antibody–mediated dimerization of ACVR1. Conversely, wild-type ACVR1 was inhibited by anti-ACVR1 antibodies. These results uncover an additional property of FOP-mutant ACVR1 and indicate that anti-ACVR1 antibodies should not be considered as therapeutics for FOP.
Collapse
Affiliation(s)
- Senem Aykul
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Lily Huang
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Lili Wang
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Nanditha M Das
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Sandra Reisman
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Yonaton Ray
- Department of Therapeutic Proteins, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Qian Zhang
- Department of Aging/Age-Related Disorders, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Nyanza J Rothman
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Kalyan C Nannuru
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Vishal Kamat
- Department of Therapeutic Proteins, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Susannah Brydges
- Velocigene, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Luca Troncone
- Department of Medicine, Harvard Medical School, Boston, United States of America
| | - Laura Johnsen
- Research Program Management, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Paul B Yu
- Department of Medicine, Harvard Medical School, Boston, United States of America
| | - Sergio Fazio
- Research Council, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - John Lees-Shepard
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Kevin Schutz
- Antibody Engineering, Adimab, Lebanon, United States of America
| | - Andrew J Murphy
- Regeneron Pharmaceuticals, Inc., Tarryrtown, United States of America
| | - Aris N Economides
- Genome Engineering Technologies, and Skeletal Diseases, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Vincent Idone
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| | - Sarah J Hatsell
- Skeletal Diseases Therapeutic Focus Area, Regeneron Pharmaceuticals, Inc., Tarrytown, United States of America
| |
Collapse
|
13
|
Gupta A, Tripathi L, Pandey S, Dwivedi D. Biology of Bone Morphogenetic Proteins in Skeleton Disease: Osteonecrosis
in Sickle Cell Disease Patients. Curr Protein Pept Sci 2022; 23:264-270. [DOI: 10.2174/1389203723666220530104442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Sickle cell disease (SCD) is an autosomal recessive trait of genetic hemoglobin disorder whose prevalence is varied from 5 to 25 % of the world population. It is characterized by the presence of hemoglobin (HbS) instead of normal hemoglobin (HbA). An individual suffering from sickle cell disease is likely to be at risk of osteonecrosis which is a form of ischemic bone infarction which causes intolerable degenerative joint problems and can affect 30-50% of people with sickle cell disease. The femoral head is the most frequent epiphyseal location in osteonecrosis with sickle cell disease. In this review, the Bone morphogenetic protein (BMP)-a subfamily of transforming growth factor-β (TGF-β) characteristics, outlined the osteoblastogenesis potentiality via using combinatorial or advanced treatment approaches. In this review, we aim to describe the Bone morphogenetic proteins' role in Skeleton diseases and discuss the potent osteogenic BMPs (majorly BMP-2, BMP-6, and BMP-7) with therapeutic benefits.
Collapse
Affiliation(s)
- Ankita Gupta
- Shyam Shah Medical College, Multidisciplinary Research Unit, Rewa, M.P., India
| | - Lokesh Tripathi
- Department of Pathology, Shyam Shah Medical College, Rewa, M.P., India
| | - Sanjay Pandey
- Multidisciplinary Research Unit, Shyam Shah Medical College, Rewa, India
| | - Deepak Dwivedi
- Department of Pediatrics, Shyam Shah Medical College, Rewa, India
| |
Collapse
|
14
|
Tanaka H, Shimizu H, Yonemochi Y, Ozawa T, Toyoshima Y, Nakajima T, Kakita A. Fibrodysplasia ossificans progressiva: histopathological implications of aberrant BMP signalling for CNS dysgenesis. Neuropathol Appl Neurobiol 2022; 48:e12805. [PMID: 35238056 DOI: 10.1111/nan.12805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Hidetomo Tanaka
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Hiroshi Shimizu
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Yosuke Yonemochi
- Department of Neurology, National Hospital Organization Niigata National Hospital, Kashiwazaki, Niigata, Japan
| | - Tetsuo Ozawa
- Department of Internal Medicine, National Hospital Organization Niigata National Hospital, Kashiwazaki, Niigata, Japan
| | - Yasuko Toyoshima
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Takashi Nakajima
- Department of Neurology, National Hospital Organization Niigata National Hospital, Kashiwazaki, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| |
Collapse
|
15
|
Maruyama R, Nguyen Q, Roshmi RR, Touznik A, Yokota T. Allele-Selective Locked Nucleic Acid Gapmers for the Treatment of Fibrodysplasia Ossificans Progressiva Knock Down the Pathogenic ACVR1 R206H Transcript and Inhibit Osteogenic Differentiation. Nucleic Acid Ther 2022; 32:185-193. [PMID: 35085461 DOI: 10.1089/nat.2021.0009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder characterized by episodic heterotopic ossification. The median life span of people with this disorder is ∼40 years, and currently, there is no effective treatment available. More than 95% of cases are caused by a recurrent mutation (c.617G>A; R206H) of Activin A receptor, type I (ACVR1)/Activin receptor-like kinase-2 (ALK2), a bone morphogenetic protein type I receptor. The mutation renders ACVR1 responsive to activin A, which does not activate wild-type ACVR1. Ectopic activation of ACVR1R206H by activin A induces heterotopic ossification. Since ACVR1R206H is a hyperactive receptor, a promising therapeutic strategy is to decrease the activity of mutated ACVR1. To accomplish this goal, we developed locked nucleic acid (LNA) gapmers. These are short DNA oligonucleotides with LNA modification at both ends. They induce targeted mRNA degradation and specific knockdown of gene expression. We demonstrated that some of these gapmers efficiently knocked down ACVR1R206H expression at RNA levels, while ACVR1WT was mostly unaffected in human FOP fibroblasts. Also, the gapmers suppressed osteogenic differentiation induced by ACVR1R206H and activin A. These gapmers may be promising drug candidates for FOP. This novel strategy will also pave the way for antisense-mediated therapy of other autosomal dominant disorders.
Collapse
Affiliation(s)
- Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Rohini Roy Roshmi
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Aleksander Touznik
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.,The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, Edmonton, Canada
| |
Collapse
|
16
|
Kaplan FS, Groppe JC, Xu M, Towler OW, Grunvald E, Kalunian K, Kallish S, Al Mukaddam M, Pignolo RJ, Shore EM. An ACVR1 R375P pathogenic variant in two families with mild fibrodysplasia ossificans progressiva. Am J Med Genet A 2021; 188:806-817. [PMID: 34854557 DOI: 10.1002/ajmg.a.62585] [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: 06/30/2021] [Revised: 09/06/2021] [Accepted: 11/02/2021] [Indexed: 11/11/2022]
Abstract
Genetic variants are vital in informing clinical phenotypes, aiding physical diagnosis, guiding genetic counseling, understanding the molecular basis of disease, and potentially stimulating drug development. Here we describe two families with an ultrarare ACVR1 gain-of-function pathogenic variant (codon 375, Arginine > Proline; ACVR1R375P ) responsible for a mild nonclassic fibrodysplasia ossificans progressiva (FOP) phenotype. Both families include people with the ultrarare ACVR1R375P variant who exhibit features of FOP while other individuals currently do not express any clinical signs of FOP. Thus, the mild ACVR1R375P variant greatly expands the scope and understanding of this rare disorder.
Collapse
Affiliation(s)
- Frederick S Kaplan
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jay C Groppe
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, Texas, USA
| | - Meiqi Xu
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - O Will Towler
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eduardo Grunvald
- Division of General Internal Medicine, The Department of Medicine, The University of California San Diego, San Diego, California, USA
| | - Kenneth Kalunian
- Division of Rheumatology, Allergy and Immunology, The Department of Medicine, UC San Diego School of Medicine, La Jolla, California, USA
| | - Staci Kallish
- The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Translational Medicine and Human Genetics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mona Al Mukaddam
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert J Pignolo
- The Department of Medicine, The Mayo Clinic, Rochester, Minnesota, USA
| | - Eileen M Shore
- Department of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Genetics, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Rooney L, Jones C. Recent Advances in ALK2 Inhibitors. ACS OMEGA 2021; 6:20729-20734. [PMID: 34423181 PMCID: PMC8374899 DOI: 10.1021/acsomega.1c02983] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
Activin receptor-like kinase-2 (ALK2) is a type I bone morphogenetic protein (BMP) receptor which has a role in biological processes that control the development of bone, heart, brain, and other tissue. Gain of function mutations in ALK2 have been identified in fibrodysplasia ossificans progressiva (FOP) and the childhood brain tumor, diffuse intrinsic pontine glioma (DIPG), which has given focus to the development of ALK2 inhibitors as targeted treatments. This review covers the structural features of ALK2 inhibitors which contribute to their ALK2 potency and selectivity, and the pharmacokinetic or in vivo efficacy data available to demonstrate their suitability for treating a peripheral or CNS disease.
Collapse
|
18
|
Esposito P, Verzola D, Picciotto D, Cipriani L, Viazzi F, Garibotto G. Myostatin/Activin-A Signaling in the Vessel Wall and Vascular Calcification. Cells 2021; 10:2070. [PMID: 34440838 PMCID: PMC8393536 DOI: 10.3390/cells10082070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
A current hypothesis is that transforming growth factor-β signaling ligands, such as activin-A and myostatin, play a role in vascular damage in atherosclerosis and chronic kidney disease (CKD). Myostatin and activin-A bind with different affinity the activin receptors (type I or II), activating distinct intracellular signaling pathways and finally leading to modulation of gene expression. Myostatin and activin-A are expressed by different cell types and tissues, including muscle, kidney, reproductive system, immune cells, heart, and vessels, where they exert pleiotropic effects. In arterial vessels, experimental evidence indicates that myostatin may mostly promote vascular inflammation and premature aging, while activin-A is involved in the pathogenesis of vascular calcification and CKD-related mineral bone disorders. In this review, we discuss novel insights into the biology and physiology of the role played by myostatin and activin in the vascular wall, focusing on the experimental and clinical data, which suggest the involvement of these molecules in vascular remodeling and calcification processes. Moreover, we describe the strategies that have been used to modulate the activin downward signal. Understanding the role of myostatin/activin signaling in vascular disease and bone metabolism may provide novel therapeutic opportunities to improve the treatment of conditions still associated with high morbidity and mortality.
Collapse
Affiliation(s)
- Pasquale Esposito
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Daniela Verzola
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| | - Daniela Picciotto
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Leda Cipriani
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| | - Francesca Viazzi
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Giacomo Garibotto
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| |
Collapse
|
19
|
Hu Y, Hao X, Liu C, Ren C, Wang S, Yan G, Meng Y, Mishina Y, Shi C, Sun H. Acvr1 deletion in osteoblasts impaired mandibular bone mass through compromised osteoblast differentiation and enhanced sRANKL-induced osteoclastogenesis. J Cell Physiol 2021; 236:4580-4591. [PMID: 33251612 PMCID: PMC8048423 DOI: 10.1002/jcp.30183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/30/2022]
Abstract
Bone morphogenetic protein (BMP) signaling is well known in bone homeostasis. However, the physiological effects of BMP signaling on mandibles are largely unknown, as the mandible has distinct functions and characteristics from other bones. In this study, we investigated the roles of BMP signaling in bone homeostasis of the mandibles by deleting BMP type I receptor Acvr1 in osteoblast lineage cells with Osterix-Cre. We found mandibular bone loss in conditional knockout mice at the ages of postnatal day 21 and 42 in an age-dependent manner. The decreased bone mass was related to compromised osteoblast differentiation together with enhanced osteoclastogenesis, which was secondary to the changes in osteoblasts in vivo. In vitro study revealed that deletion of Acvr1 in the mandibular bone marrow stromal cells (BMSCs) significantly compromised osteoblast differentiation. When wild type bone marrow macrophages were cocultured with BMSCs lacking Acvr1 both directly and indirectly, both proliferation and differentiation of osteoclasts were induced as evidenced by an increase of multinucleated cells, compared with cocultured with control BMSCs. Furthermore, we demonstrated that the increased osteoclastogenesis in vitro was at least partially due to the secretion of soluble receptor activator of nuclear factor-κB ligand (sRANKL), which is probably the reason for the mandibular bone loss in vivo. Overall, our results proposed that ACVR1 played essential roles in maintaining mandibular bone homeostasis through osteoblast differentiation and osteoblast-osteoclast communication via sRANKL.
Collapse
Affiliation(s)
- Yue Hu
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Xinqing Hao
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Cangwei Liu
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Chunxia Ren
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Shuangshuang Wang
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Guangxing Yan
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Yuan Meng
- Department of Oral Pathology, School and Hospital of StomatologyChina Medical UniversityShenyangChina
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of DentistryUniversity of MichiganAnn ArborMichiganUSA
| | - Ce Shi
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| | - Hongchen Sun
- Department of Oral Pathology, Hospital of StomatologyJilin UniversityChangchunChina
- Key Laboratory of Tooth Development and Bone Remodeling of Jilin ProvinceChangchunChina
| |
Collapse
|
20
|
MyD88 Is Not Required for Muscle Injury-Induced Endochondral Heterotopic Ossification in a Mouse Model of Fibrodysplasia Ossificans Progressiva. Biomedicines 2021; 9:biomedicines9060630. [PMID: 34206078 PMCID: PMC8227787 DOI: 10.3390/biomedicines9060630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 01/09/2023] Open
Abstract
Excess inflammation and canonical BMP receptor (BMPR) signaling are coinciding hallmarks of the early stages of injury-induced endochondral heterotopic ossification (EHO), especially in the rare genetic disease fibrodysplasia ossificans progressiva (FOP). Multiple inflammatory signaling pathways can synergistically enhance BMP-induced Smad1/5/8 activity in multiple cell types, suggesting the importance of pathway crosstalk in EHO and FOP. Toll-like receptors (TLRs) and IL-1 receptors mediate many of the earliest injury-induced inflammatory signals largely via MyD88-dependent pathways. Thus, the hypothesis that MyD88-dependent signaling is required for EHO was tested in vitro and in vivo using global or Pdgfrα-conditional deletion of MyD88 in FOP mice. As expected, IL-1β or LPS synergistically increased Activin A (ActA)-induced phosphorylation of Smad 1/5 in fibroadipoprogenitors (FAPs) expressing Alk2R206H. However, conditional deletion of MyD88 in Pdgfrα-positive cells of FOP mice did not significantly alter the amount of muscle injury-induced EHO. Even more surprisingly, injury-induced EHO was not significantly affected by global deletion of MyD88. These studies demonstrate that MyD88-dependent signaling is dispensable for injury-induced EHO in FOP mice.
Collapse
|
21
|
Yamin G, Daghighi S, Mafee M. Fibrodysplasia ossificans progressiva (FOP) presenting as a rapidly growing non-calcified neck mass. J Radiol Case Rep 2021; 15:10-16. [PMID: 34276874 DOI: 10.3941/jrcr.v15i5.4103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an extremely rare autosomal dominant inherited disorder leading to mature ossification within soft tissues. We report a 62-year-old female with a 3-week history of a rapidly enlarging left neck mass with no associated symptoms. A neck CT showed a ~10 cm solid-appearing non-calcified left neck mass that markedly decreased in size on a one-month follow-up neck MRI, but with new extensive edema/intense enhancement in floor of the mouth. Prior radiographs documented hallux valgus and heterotopic ossification of the psoas/paraspinal muscles and shoulder girdle. In this case of FOP, no intervention was implemented and the symptoms improved over time and thus paralleled other such cases for flare-ups. Clinicians should be aware of this rare entity, as it is frequently misdiagnosed as cancer or other benign entities such as infection, resulting in biopsies that can often hasten disease progression.
Collapse
Affiliation(s)
- Ghiam Yamin
- Department of Radiology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Neuroimaging and Neurointervention, Stanford University School of Medicine, Stanford, CA, USA
| | - Shadi Daghighi
- Department of Radiology, University of California San Diego School of Medicine, San Diego, CA, USA
- Department of Internal Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA
| | - Mahmood Mafee
- Department of Radiology, University of California San Diego School of Medicine, San Diego, CA, USA
| |
Collapse
|
22
|
Martín-García D, Towler OW, Xu M, Alfonso-Hernández O, Oliveira PR, Alonso-Clavo M, Shore EM, Kaplan FS. Nonclassic fibrodysplasia ossificans progressiva: A child from Angola with an ACVR1 G328E variant. Am J Med Genet A 2021; 185:2572-2575. [PMID: 33973349 DOI: 10.1002/ajmg.a.62253] [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: 01/05/2021] [Revised: 03/03/2021] [Accepted: 04/24/2021] [Indexed: 11/09/2022]
Abstract
Little is known about FOP in Africa and few cases of nonclassic fibrodysplasia ossificans progressiva (FOP) have been reported on the continent. Here we report a three-year-old girl from Angola with a nonclassic FOP clinical presentation that is characterized by complex malformations of the toes and fingers, reduction defects of the digits, absence of nails, progressive heterotopic ossification, and a confirmed heterozygous ACVR1 variant at c.983G > A. Emerging knowledge of FOP can serve as a catalyst for increasing awareness of FOP in under-represented medical communities by achieving a correct FOP diagnosis, improving access of individuals with FOP to clinical trial recruitment, and enhancing the ability of affected individuals to be part of and interact with the international FOP community.
Collapse
Affiliation(s)
| | - O Will Towler
- Departments of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meiqi Xu
- Departments of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Osmany Alfonso-Hernández
- Faculty of Medicine, Katyavala Bwila University, Benguela, Angola.,Division of Orthopedics, Benguela's General Hospital, Benguela, Angola
| | - Paula R Oliveira
- Faculty of Medicine, Katyavala Bwila University, Benguela, Angola
| | - Marleny Alonso-Clavo
- Faculty of Medicine, Katyavala Bwila University, Benguela, Angola.,Division of Pediatrics, Benguela's General Hospital, Benguela, Angola
| | - Eileen M Shore
- Departments of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Departments of Genetics, The Perelman School of Medicine at the University of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Frederick S Kaplan
- Departments of Orthopaedic Surgery, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Departments of Medicine, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Tucker-Bartley A, Lemme J, Gomez-Morad A, Shah N, Veliu M, Birklein F, Storz C, Rutkove S, Kronn D, Boyce AM, Kraft E, Upadhyay J. Pain Phenotypes in Rare Musculoskeletal and Neuromuscular Diseases. Neurosci Biobehav Rev 2021; 124:267-290. [PMID: 33581222 PMCID: PMC9521731 DOI: 10.1016/j.neubiorev.2021.02.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
For patients diagnosed with a rare musculoskeletal or neuromuscular disease, pain may transition from acute to chronic; the latter yielding additional challenges for both patients and care providers. We assessed the present understanding of pain across a set of ten rare, noninfectious, noncancerous disorders; Osteogenesis Imperfecta, Ehlers-Danlos Syndrome, Achondroplasia, Fibrodysplasia Ossificans Progressiva, Fibrous Dysplasia/McCune-Albright Syndrome, Complex Regional Pain Syndrome, Duchenne Muscular Dystrophy, Infantile- and Late-Onset Pompe disease, Charcot-Marie-Tooth Disease, and Amyotrophic Lateral Sclerosis. Through the integration of natural history, cross-sectional, retrospective, clinical trials, & case studies we described pathologic and genetic factors, pain sources, phenotypes, and lastly, existing therapeutic approaches. We highlight that while rare diseases possess distinct core pathologic features, there are a number of shared pain phenotypes and mechanisms that may be prospectively examined and therapeutically targeted in a parallel manner. Finally, we describe clinical and research approaches that may facilitate more accurate diagnosis, monitoring, and treatment of pain as well as elucidation of the evolving nature of pain phenotypes in rare musculoskeletal or neuromuscular illnesses.
Collapse
Affiliation(s)
- Anthony Tucker-Bartley
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jordan Lemme
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrea Gomez-Morad
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Nehal Shah
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Miranda Veliu
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Frank Birklein
- Department of Neurology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Rhineland-Palatinate, 55131, Germany
| | - Claudia Storz
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital LMU Munich, Munich, Bavaria, 80539, Germany
| | - Seward Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - David Kronn
- Department of Pathology and Pediatrics, New York Medical College, Valhalla, NY, 10595, USA; Medical Genetics, Inherited Metabolic & Lysosomal Storage Disorders Center, Boston Children's Health Physicians, Westchester, NY, 10532, USA
| | - Alison M Boyce
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eduard Kraft
- Department of Orthopedics, Physical Medicine and Rehabilitation, University Hospital LMU Munich, Munich, Bavaria, 80539, Germany; Interdisciplinary Pain Unit, University Hospital LMU Munich, Munich, 80539, Germany
| | - Jaymin Upadhyay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
24
|
Bariker RH, Hugar SM. Chairside endodontic management of a child with fibrodysplasia ossificans progressiva. J Indian Soc Pedod Prev Dent 2021; 39:221-224. [PMID: 34341245 DOI: 10.4103/jisppd.jisppd_192_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an exceptionally atypical genetic disorder characterized by heterotopic bone formation within skeletal muscles, ligaments, tendons, and other connective tissues that affects one in 2 million people. A 4-year-old girl with FOP was referred to our practice with complaints of pain and discomfort in both sides of the lower jaw. Clinical examination revealed deep multiple caries and buccal gingival abscess in relation to multiple teeth. Endodontic care and conservative dental procedures were planned and performed chairside after a detailed discussion with FOP medical and dental experts. Very brief dental appointments were conducted with breaks to prevent muscle fatigue. No usage of regional anesthesia or dental dam clamps was done. The patient and her parents were counseled for oral hygiene maintenance, and periodic topical fluoride treatments were performed during successive follow-up appointments. The child is followed for 34 months post-treatment. The dental treatment modifications implemented for the present case were enough to institute good oral health and to prevent the creation of heterotopic ossifications in the maxillofacial region.
Collapse
Affiliation(s)
| | - Shivayogi M Hugar
- Department of Pedodontics and Preventive Dentistry, KAHER's KLE VK Institute of Dental Sciences, Belagavi, Karnataka, India
| |
Collapse
|
25
|
Cappato S, Gamberale R, Bocciardi R, Brunelli S. Genetic and Acquired Heterotopic Ossification: A Translational Tale of Mice and Men. Biomedicines 2020; 8:biomedicines8120611. [PMID: 33327623 PMCID: PMC7765130 DOI: 10.3390/biomedicines8120611] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/10/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
Heterotopic ossification is defined as an aberrant formation of bone in extraskeletal soft tissue, for which both genetic and acquired conditions are known. This pathologic process may occur in many different sites such as the skin, subcutaneous tissue, skeletal muscle and fibrous tissue adjacent to joints, ligaments, walls of blood vessels, mesentery and other. The clinical spectrum of this disorder is wide: lesions may range from small foci of ossification to massive deposits of bone throughout the body, typical of the progressive genetically determined conditions such as fibrodysplasia ossificans progressiva, to mention one of the most severe and disabling forms. The ectopic bone formation may be regarded as a failed tissue repair process in response to a variety of triggers and evolving towards bone formation through a multistage differentiation program, with several steps common to different clinical presentations and distinctive features. In this review, we aim at providing a comprehensive view of the genetic and acquired heterotopic ossification disorders by detailing the clinical and molecular features underlying the different human conditions in comparison with the corresponding, currently available mouse models.
Collapse
Affiliation(s)
- Serena Cappato
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Sciences (DINOGMI), University of Genova, 16132 Genova, Italy;
| | - Riccardo Gamberale
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Milano, Italy; (R.G.); (S.B.)
| | - Renata Bocciardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Sciences (DINOGMI), University of Genova, 16132 Genova, Italy;
- UOC Genetica Medica, IRCCS Giannina Gaslini, 16147 Genova, Italy
- Correspondence:
| | - Silvia Brunelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Milano, Italy; (R.G.); (S.B.)
| |
Collapse
|
26
|
Kaliya-Perumal AK, Carney TJ, Ingham PW. Fibrodysplasia ossificans progressiva: current concepts from bench to bedside. Dis Model Mech 2020; 13:13/9/dmm046441. [PMID: 32988985 PMCID: PMC7522019 DOI: 10.1242/dmm.046441] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heterotopic ossification (HO) is a disorder characterised by the formation of ectopic bone in soft tissue. Acquired HO typically occurs in response to trauma and is relatively common, yet its aetiology remains poorly understood. Genetic forms, by contrast, are very rare, but provide insights into the mechanisms of HO pathobiology. Fibrodysplasia ossificans progressiva (FOP) is the most debilitating form of HO. All patients reported to date carry heterozygous gain-of-function mutations in the gene encoding activin A receptor type I (ACVR1). These mutations cause dysregulated bone morphogenetic protein (BMP) signalling, leading to HO at extraskeletal sites including, but not limited to, muscles, ligaments, tendons and fascia. Ever since the identification of the causative gene, developing a cure for FOP has been a focus of investigation, and studies have decoded the pathophysiology at the molecular and cellular levels, and explored novel management strategies. Based on the established role of BMP signalling throughout HO in FOP, therapeutic modalities that target multiple levels of the signalling cascade have been designed, and some drugs have entered clinical trials, holding out hope of a cure. A potential role of other signalling pathways that could influence the dysregulated BMP signalling and present alternative therapeutic targets remains a matter of debate. Here, we review the recent FOP literature, including pathophysiology, clinical aspects, animal models and current management strategies. We also consider how this research can inform our understanding of other types of HO and highlight some of the remaining knowledge gaps. Summary: Fibrodysplasia ossificans progressiva is a rare disease characterised by progressive heterotopic bone formation. Here, we present a comprehensive summary of the recent literature on this debilitating condition and discuss approaches to solving this clinical puzzle.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore
| | - Tom J Carney
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore.,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos 138673, Singapore
| | - Philip W Ingham
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, 636921, Singapore .,Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos 138673, Singapore
| |
Collapse
|
27
|
Hwang C, Pagani CA, Das N, Marini S, Huber AK, Xie L, Jimenez J, Brydges S, Lim WK, Nannuru KC, Murphy AJ, Economides AN, Hatsell SJ, Levi B. Activin A does not drive post-traumatic heterotopic ossification. Bone 2020; 138:115473. [PMID: 32553795 DOI: 10.1016/j.bone.2020.115473] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/15/2022]
Abstract
Heterotopic ossification (HO), the formation of ectopic bone in soft tissues, has been extensively studied in its two primary forms: post-traumatic HO (tHO) typically found in patients who have experienced musculoskeletal or neurogenic injury and in fibrodysplasia ossificans progressiva (FOP), where it is genetically driven. Given that in both diseases HO arises via endochondral ossification, the molecular mechanisms behind both diseases have been postulated to be manifestations of similar pathways including those activated by BMP/TGFβ superfamily ligands. A significant step towards understanding the molecular mechanism by which HO arises in FOP was the discovery that FOP causing ACVR1 variants trigger HO in response to activin A, a ligand that does not activate signaling from wild type ACVR1, and that is not inherently osteogenic in wild type settings. The physiological significance of this finding was demonstrated by showing that activin A neutralizing antibodies stop HO in two different genetically accurate mouse models of FOP. In order to explore the role of activin A in tHO, we performed single cell RNA sequencing and compared the expression of activin A as well as other BMP pathway genes in tHO and FOP HO. We show that activin A is expressed in response to injury in both settings, but by different types of cells. Given that wild type ACVR1 does not transduce signal when engaged by activin A, we hypothesized that inhibition of activin A will not block tHO. Nonetheless, as activin A was expressed in tHO lesions, we tested its inhibition and compared it with inhibition of BMPs. We show here that anti-activin A does not block tHO, whereas agents such as antibodies that neutralize ACVR1 or ALK3-Fc (which blocks osteogenic BMPs) are beneficial, though not completely curative. These results demonstrate that inhibition of activin A should not be considered as a therapeutic strategy for ameliorating tHO.
Collapse
Affiliation(s)
- Charles Hwang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States of America
| | - Chase A Pagani
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States of America
| | | | - Simone Marini
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States of America
| | - Amanda K Huber
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States of America
| | - LiQin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | | | | | | | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States of America; Division of Plastic Surgery, Department of Surgery, University of Michigan Health System, 1500 E Medical Center Drive, SPC 5340, Ann Arbor, MI 48109-5340, United States of America.
| |
Collapse
|
28
|
Vanhoutte F, Liang S, Ruddy M, Zhao A, Drewery T, Wang Y, DelGizzi R, Forleo-Neto E, Rajadhyaksha M, Herman G, Davis JD. Pharmacokinetics and Pharmacodynamics of Garetosmab (Anti-Activin A): Results From a First-in-Human Phase 1 Study. J Clin Pharmacol 2020; 60:1424-1431. [PMID: 32557665 PMCID: PMC7586962 DOI: 10.1002/jcph.1638] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/15/2020] [Indexed: 12/30/2022]
Abstract
We describe outcomes from the first‐in‐human study of garetosmab (a fully human monoclonal antibody that inhibits activin A) under development for the treatment of fibrodysplasia ossificans progressiva (FOP). In a double‐blind, placebo‐controlled phase 1 study, 40 healthy women of nonchildbearing potential were randomized to receive a single dose of intravenous garetosmab 0.3, 1, 3, or 10 mg/kg; subcutaneous garetosmab 300 mg; or placebo. Serum concentrations of functional garetosmab (with ≥1 arm free to bind to target), total activin A, and antidrug antibodies were measured predose and up to 113 days post–first dose. Garetosmab demonstrated an acceptable safety profile with no dose‐limiting toxicities. Garetosmab displayed nonlinear pharmacokinetics with target‐mediated elimination. With increasing doses of intravenous garetosmab, mean peak concentration increased in a dose‐proportional manner; mean steady‐state estimates ranged from 41.4 to 47.8 mL/kg. A greater than dose‐proportional increase in mean area under the concentration‐time curve from time zero extrapolated to infinity (range, 72.2‐7520 mg*day/L) was observed, consistent with decreasing mean clearance (range, 4.35‐1.34 mL/day/kg). Following administration of intravenous garetosmab, mean concentrations of total activin A increased in a dose‐dependent manner. At 10 mg/kg, total activin A levels reached a state of little or no change between weeks 4 and 12, suggesting saturation of the target‐mediated pathway. No safety signals were seen in this study to preclude investigation in patients. Following intravenous administration, garetosmab concentrations decreased quickly, then decreased over time (reflecting linear elimination), and finally decreased in a nonlinear phase, reflecting target‐mediated elimination. Results here support further investigation. Garetosmab 10 mg/kg every 4 weeks intravenously is being evaluated in patients with FOP (NCT03188666).
Collapse
Affiliation(s)
| | - Su Liang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Marcella Ruddy
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - An Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Tiera Drewery
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - Yuhuan Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | | | | | | | - Gary Herman
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| | - John D Davis
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, USA
| |
Collapse
|
29
|
Aykul S, Corpina RA, Goebel EJ, Cunanan CJ, Dimitriou A, Kim HJ, Zhang Q, Rafique A, Leidich R, Wang X, McClain J, Jimenez J, Nannuru KC, Rothman NJ, Lees-Shepard JB, Martinez-Hackert E, Murphy AJ, Thompson TB, Economides AN, Idone V. Activin A forms a non-signaling complex with ACVR1 and type II Activin/BMP receptors via its finger 2 tip loop. eLife 2020; 9:54582. [PMID: 32515349 PMCID: PMC7326492 DOI: 10.7554/elife.54582] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/08/2020] [Indexed: 01/18/2023] Open
Abstract
Activin A functions in BMP signaling in two ways: it either engages ACVR1B to activate Smad2/3 signaling or binds ACVR1 to form a non-signaling complex (NSC). Although the former property has been studied extensively, the roles of the NSC remain unexplored. The genetic disorder fibrodysplasia ossificans progressiva (FOP) provides a unique window into ACVR1/Activin A signaling because in that disease Activin can either signal through FOP-mutant ACVR1 or form NSCs with wild-type ACVR1. To explore the role of the NSC, we generated 'agonist-only' Activin A muteins that activate ACVR1B but cannot form the NSC with ACVR1. Using one of these muteins, we demonstrate that failure to form the NSC in FOP results in more severe disease pathology. These results provide the first evidence for a biological role for the NSC in vivo and pave the way for further exploration of the NSC's physiological role in corresponding knock-in mice.
Collapse
Affiliation(s)
- Senem Aykul
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | | | | | | | - Qian Zhang
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | - Xin Wang
- Regeneron Pharmaceuticals, Tarrytown, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Rauner M, Seefried L, Shore E. Genetics and future therapy prospects of fibrodysplasia ossificans progressiva. MED GENET-BERLIN 2020. [DOI: 10.1007/s11825-019-00279-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant genetic condition characterised by progressive extra-skeletal bone formation in connective tissues. Over time, heterotopic ossification entombs patients within a second skeleton, drastically impairing their mobility and autonomy. Mutations in the ACVR1 gene have been identified as the cause of FOP. The single nucleotide missense mutation in ACVR1, c.617G > A, causes a single amino acid substitution, p.R206H, and is found in >90% of all patients. Heterotopic bone formation in FOP mimics embryonic skeletal endochondral ossification, with cartilage forming after fibroproliferative tissue condensation as an intermediate stage prior to osteogenesis and tissue ossification. In contrast to normal embryonic endochondral ossification, heterotopic ossification in FOP involves an inflammatory phase that precedes cartilage and bone formation. New insights into the mechanisms of action of heterotopic bone formation in FOP have led to the discovery of new potential treatment targets including inhibitors of BMP signalling, activin A inhibitors, and mTOR inhibitors. This review summarises the current knowledge on mutations causing FOP, as well as the molecular basis of heterotopic ossification and the therapeutic options that result from these discoveries.
Collapse
Affiliation(s)
- Martina Rauner
- 1 grid.4488.0 0000 0001 2111 7257 Department of Medicine III & Center for Healthy Aging Technische Universität Dresden 01307 Dresden Germany
| | - Lothar Seefried
- 2 grid.8379.5 0000 0001 1958 8658 Department of Orthopedics University of Würzburg Würzburg Germany
| | - Eileen Shore
- 3 grid.25879.31 0000 0004 1936 8972 Departments of Orthopedics and Genetics, Perelman School of Medicine University of Pennsylvania Philadelphia USA
| |
Collapse
|
31
|
Schoenmaker T, Botman E, Sariyildiz M, Micha D, Netelenbos C, Bravenboer N, Kelder A, Eekhoff EMW, De Vries TJ. Activin-A Induces Fewer, but Larger Osteoclasts From Monocytes in Both Healthy Controls and Fibrodysplasia Ossificans Progressiva Patients. Front Endocrinol (Lausanne) 2020; 11:501. [PMID: 32760351 PMCID: PMC7371852 DOI: 10.3389/fendo.2020.00501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is a rare genetic disease characterized by heterotopic ossification (HO) that occurs in muscle tissue, tendons, and ligaments. The disease is caused by mutations in the Activin receptor type I (ACVR1) gene resulting in enhanced responsiveness to Activin-A. Binding of this molecule to the mutated receptor induces HO. Bone metabolism normally requires the coupled action of osteoblasts and osteoclasts, which seems to be disturbed during HO. We hypothesize that Activin-A may also counteract the formation of osteoclasts in FOP patients. In this study we investigated the effect of Activin-A on osteoclast differentiation of CD14+ monocytes from FOP patients and healthy controls. The lymphocytic and monocytic cell populations were determined by FACS analysis. Expression of the mutated R206H receptor was assessed and confirmed by allele specific PCR. The effect of Activin-A on osteoclastogenesis was assessed by counting the number and size of multinucleated cells. Osteoclast activity was determined by culturing the cells on Osteo Assay plates. The influence of Activin-A on expression of various osteoclast related genes was studied with QPCR. Blood from FOP patients contained similar percentages of classical, intermediate, or non-classical monocytes as healthy controls. Addition of Activin-A to the osteoclastogenesis cultures resulted in fewer osteoclasts in both control and FOP cultures. The osteoclasts formed in the presence of Activin-A were, however, much larger and more active compared to the cultures without Activin-A. This effect was tempered when the Activin-A inhibitor follistatin was added to the Activin-A containing cultures. Expression of osteoclast specific genes Cathepsin K and TRAcP was upregulated, gene expression of osteoclastogenesis related genes M-CSF and DC-STAMP was downregulated by Activin-A. Since Activin-A is a promising target for inhibiting the formation of HO in FOP, it is important to know its effects on both osteoblasts and osteoclasts. Our study shows that Activin-A induces fewer, but larger and more active osteoclasts independent of the presence of the mutated ACVR1 receptor. When considering FOP as an Activin-A driven disease that acts locally, our findings suggest that Activin-A could cause a more pronounced local resorption by larger osteoclasts. Thus, when targeting Activin-A in patients with neutralizing antibodies, HO formation could potentially be inhibited, and osteoclastic activity could be slightly reduced, but then performed dispersedly by more and smaller osteoclasts.
Collapse
Affiliation(s)
- Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
- *Correspondence: Ton Schoenmaker
| | - Esmée Botman
- Department of Internal Medicine Section Endocrinology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Merve Sariyildiz
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Coen Netelenbos
- Department of Internal Medicine Section Endocrinology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Angele Kelder
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - E. Marelise W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teun J. De Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| |
Collapse
|
32
|
Towler OW, Shore EM, Kaplan FS. Skeletal malformations and developmental arthropathy in individuals who have fibrodysplasia ossificans progressiva. Bone 2020; 130:115116. [PMID: 31655222 DOI: 10.1016/j.bone.2019.115116] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 11/29/2022]
Abstract
RATIONALE Fibrodysplasia ossificans progressiva (FOP) is primarily a disease of progressive heterotopic ossification (HO) leading to impaired mobility throughout life. An additional diagnostic feature is a characteristic malformation of the great toes. The culpable gene for FOP,ACVR1 (activin A receptor type 1) has a clear effect on the induction of extra-skeletal bone formation. However, this bone morphogenetic protein (BMP) pathway receptor is expressed widely throughout skeletal development and has a seminal role in axial and appendicular chondrogenesis, prompting suspicion of widespread bone and joint defects in those with ACVR1 mutations. MATERIALS AND METHODS We analyzed baseline whole body (minus skull) computed tomographic (CT) scans of 113 individuals with classic clinical features of FOP and the ACVR1 (R206H) mutation who were enrolled in a non-interventional natural history study ((NCT02322255)) for skeletal malformations, atypical morphology, intra-articular synovial osteochondromatosis, developmental arthropathy, and associated degenerative joint phenotypes. Individuals were evaluated in three age groups: 4-13; 14-25; and 25-56 years old, based on historical models of FOP disease progression. RESULTS We found widespread evidence of developmental arthropathy throughout the axial and appendicular skeleton in all age groups (61M, 52F; ages: 4-56 years). Asymmetric narrowing and subchondral sclerosis were present throughout the joints of the normotopic skeleton and osteophytes were common in the hips and knees of individuals who have FOP in all age groups. The costovertebral joints, intervertebral facet joints, and proximal tibio-fibular joints frequently showed partial or total intra-articular ankylosis, particularly after age 13. The hips of FOP subjects are frequently malformed and dysplastic. We also found evidence of degenerative joint phenotypes after age 13, particularly in the spine, sacro-iliac joints, and lower limbs. CONCLUSIONS The effects of ACVR1 mutation on the normotopic skeletons of individuals who have FOP extend beyond malformation of the great toes and include both morphological defects and developmental arthropathy. Associated degenerative joint disease occurring at multiple sites starts in adolescence and progresses throughout life. These phenotypes appear to be uncoupled from heterotopic bone formation, indicating a potential role for ACVR1 in the development and progression of degenerative joint disease. SIGNIFICANCE FOP is a disease of not only progressive heterotopic ossification, but also widespread and extensive developmental arthropathy and associated degenerative joint disease. These findings have relevance for understanding the natural history of FOP and for designing and evaluating clinical trials with emerging therapeutics.
Collapse
Affiliation(s)
- O Will Towler
- Departments of Orthopaedic Surgery, The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Departments of The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States
| | - Eileen M Shore
- Departments of Orthopaedic Surgery, The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Departments of Genetics, The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Departments of The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States
| | - Frederick S Kaplan
- Departments of Orthopaedic Surgery, The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Departments of Medicine, The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States; Departments of The Center for Research in FOP & Related Disorders, The Perelman School of Medicine at The University of Pennsylvania, 3450 Hamilton Walk, 309A Stemmler Hall, Philadelphia, PA 19104, United States.
| |
Collapse
|
33
|
Inhibition of immune checkpoints prevents injury-induced heterotopic ossification. Bone Res 2019; 7:33. [PMID: 31700694 PMCID: PMC6823457 DOI: 10.1038/s41413-019-0074-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/06/2019] [Accepted: 08/18/2019] [Indexed: 12/17/2022] Open
Abstract
Heterotopic ossification (HO), true bone formation in soft tissue, is closely associated with abnormal injury/immune responses. We hypothesized that a key underlying mechanism of HO might be injury-induced dysregulation of immune checkpoint proteins (ICs). We found that the earliest stages of HO are characterized by enhanced infiltration of polarized macrophages into sites of minor injuries in an animal model of HO. The non-specific immune suppressants, Rapamycin and Ebselen, prevented HO providing evidence of the central role of the immune responses. We examined the expression pattern of ICs and found that they are dysregulated in HO lesions. More importantly, loss of function of inhibitory ICs (including PD1, PD-L1, and CD152) markedly inhibited HO, whereas loss of function of stimulatory ICs (including CD40L and OX-40L) facilitated HO. These findings suggest that IC inhibitors may provide a therapeutic approach to prevent or limit the extent of HO.
Collapse
|
34
|
Valer JA, Sánchez-de-Diego C, Pimenta-Lopes C, Rosa JL, Ventura F. ACVR1 Function in Health and Disease. Cells 2019; 8:cells8111366. [PMID: 31683698 PMCID: PMC6912516 DOI: 10.3390/cells8111366] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Activin A receptor type I (ACVR1) encodes for a bone morphogenetic protein type I receptor of the TGFβ receptor superfamily. It is involved in a wide variety of biological processes, including bone, heart, cartilage, nervous, and reproductive system development and regulation. Moreover, ACVR1 has been extensively studied for its causal role in fibrodysplasia ossificans progressiva (FOP), a rare genetic disorder characterised by progressive heterotopic ossification. ACVR1 is linked to different pathologies, including cardiac malformations and alterations in the reproductive system. More recently, ACVR1 has been experimentally validated as a cancer driver gene in diffuse intrinsic pontine glioma (DIPG), a malignant childhood brainstem glioma, and its function is being studied in other cancer types. Here, we review ACVR1 receptor function and signalling in physiological and pathological processes and its regulation according to cell type and mutational status. Learning from different functions and alterations linked to ACVR1 is a key step in the development of interdisciplinary research towards the identification of novel treatments for these pathologies.
Collapse
Affiliation(s)
- José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, 08907 Barcelona, Spain.
| |
Collapse
|
35
|
Botman E, Raijmakers PGHM, Yaqub M, Teunissen B, Netelenbos C, Lubbers W, Schwarte LA, Micha D, Bravenboer N, Schoenmaker T, de Vries TJ, Pals G, Smit JM, Koolwijk P, Trotter DG, Lammertsma AA, Eekhoff EMW. Evolution of heterotopic bone in fibrodysplasia ossificans progressiva: An [ 18F]NaF PET/CT study. Bone 2019; 124:1-6. [PMID: 30858149 DOI: 10.1016/j.bone.2019.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 03/05/2019] [Accepted: 03/07/2019] [Indexed: 11/30/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare, autosomal dominant disorder characterized by heterotopic ossification (HO) in muscles, ligaments and tendons. Flare-ups often precede the formation of HO, resulting in immobilization of joints. Due to progression of the disease without signs of a flare-up, co-existence of a chronic progression of HO has been postulated, but conclusive evidence is lacking. Recently, it has been shown that [18F]NaF PET/CT is able to identify early ossifying disease activity during flare-ups. Therefore, the purpose of the present study was to assess whether [18F]NaF PET/CT might also be able to identify the possible presence of chronic progressive HO in FOP. A total of thirteen [18F]NaF PET/CT scans from five FOP patients were analysed. Scans were acquired over a period of 0.5 to 2 years. Volumes of HO and standardized uptake values (SUV) were obtained based on manual segmentation of CT images. SUVpeak values, defined as the average SUV value of a 1 mL sphere containing the hottest voxel pixels, were obtained. Two out of five patients experienced ≥1 active clinical flare-ups at the time of the [18F]NaF PET/CT scan. In addition, in four out of five patients, serial scans showed radiological progression of HO (3 to 8 cm3), as assessed by CT volume, in the absence of a clinical flare-up. This volumetric increase was present in 6/47 (12.8%) of identified HO structures and, in all cases, was accompanied by increased [18F]NaF uptake, with SUVpeak ranging from 8.4 to 17.9. In conclusion, HO may progress without signs of a flare-up. [18F]NaF PET/CT is able to identify these asymptomatic, but progressive HO lesions, thereby demonstrating the presence of chronic activity in FOP. Consequently, future drugs should not only target new HO formation, but also this chronic HO progression.
Collapse
Affiliation(s)
- Esmée Botman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Pieter G H M Raijmakers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Maqsood Yaqub
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Bernd Teunissen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - Coen Netelenbos
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Wouter Lubbers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anaesthesiology, the Netherlands
| | - Lothar A Schwarte
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Anaesthesiology, the Netherlands
| | - Dimitra Micha
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Genetics, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Nathalie Bravenboer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Chemistry, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit
| | - Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit
| | - Gerard Pals
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Clinical Genetics, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands
| | - Jan Maerten Smit
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Plastic, Reconstructive and Hand Surgery, Amsterdam Bone Center, the Netherlands
| | - Pieter Koolwijk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Physiology, the Netherlands
| | | | - Adriaan A Lammertsma
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, the Netherlands
| | - E Marelise W Eekhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Internal Medicine Section Endocrinology, Amsterdam Bone Center, Amsterdam Movement Sciences, the Netherlands.
| |
Collapse
|
36
|
Gucev Z, Tasic V, Plaseska-Karanfilska D, Dimishkovska M, Laban N, Bozinovski Z, Kostovski M, Saveski A, Polenakovic M, Towler OW, Shore EM, Kaplan FS. Severe digital malformations in a rare variant of fibrodysplasia ossificans progressiva. Am J Med Genet A 2019; 179:1310-1314. [PMID: 31012264 DOI: 10.1002/ajmg.a.61153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/23/2019] [Accepted: 04/01/2019] [Indexed: 11/06/2022]
Abstract
A 16-year-old girl with a history of nontraumatic swelling of both forearms, osteochondromas of the knees, heterotopic ossification of the neck and back, severe malformations of all digits with hypoplastic or absent nails, alopecia partialis of the scalp, and moderate cognitive impairment was seen for diagnostic evaluation. Whole exome sequencing identified an activating mutation of ACVR1 (c.983G > A; p.Gly328Glu) which confirmed a suspected FOP variant. The delayed diagnosis of an FOP variant in this patient could have been avoided if the significance of severe digital malformations had been recognized, especially in the setting of progressive heterotopic ossification.
Collapse
Affiliation(s)
- Zoran Gucev
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Velibor Tasic
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Dijana Plaseska-Karanfilska
- Research Centre for Genetic Engineering and Biotechnology "Georgi D. Efremov", Macedonian Academy of Sciences and Arts, Skopje, Republic of North Macedonia
| | - Marija Dimishkovska
- Research Centre for Genetic Engineering and Biotechnology "Georgi D. Efremov", Macedonian Academy of Sciences and Arts, Skopje, Republic of North Macedonia
| | - Nevenka Laban
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Zoran Bozinovski
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Marko Kostovski
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Alek Saveski
- Medical Faculty - Skopje, University Ss Cyril and Methodius of Skopje, Skopje, Republic of North Macedonia
| | - Momir Polenakovic
- Research Centre for Genetic Engineering and Biotechnology "Georgi D. Efremov", Macedonian Academy of Sciences and Arts, Skopje, Republic of North Macedonia
| | - O Will Towler
- Department of Orthopaedic Surgery, Center for Research in FOP & Related Disorders, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Eileen M Shore
- Department of Orthopaedic Surgery, Center for Research in FOP & Related Disorders, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Genetics, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, Center for Research in FOP & Related Disorders, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Peng K, Cheung K, Lee A, Sieberg C, Borsook D, Upadhyay J. Longitudinal Evaluation of Pain, Flare-Up, and Emotional Health in Fibrodysplasia Ossificans Progressiva: Analyses of the International FOP Registry. JBMR Plus 2019; 3:e10181. [PMID: 31485551 PMCID: PMC6715827 DOI: 10.1002/jbm4.10181] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/04/2019] [Accepted: 01/30/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is an ultra‐rare, inherited, connective tissue disease with ∼800 documented cases worldwide. The principal pathological feature of FOP is the transition of skeletal muscle, tendons, ligaments, and fascia into cartilage and bone. This heterotopic ossification (HO) is often preceded by painful soft tissue swellings or flare‐ups that may last several months. For many individuals, experiencing a flare‐up may represent a worsening of their condition and contribute to feelings of anxiety or suppressed affect, both of which are well‐recognized to exacerbate pain perception. To date, much remains unknown regarding the dynamics of pain and emotional health in FOP during flare‐up and also quiescent, non–flare‐up disease phases. In order to elucidate the occurrence and effect of pain in FOP, this study analyzed Patient‐Reported Outcomes Measurement Information System–based questionnaires completed by 99 patients participating in the international FOP Registry over a 30‐month period. We observed that although moderate to severe pain (≥4, 0 to 10 pain scale) was commonly associated with flare‐ups (56% to 67%), surprisingly, 30% to 55% of patients experienced similar pain levels during non–flare‐up states. In those patients reporting pain levels of ≥4, 45% to 74% of patients report experiencing anxiety, depression, or irritability, with 36% to 48% reporting emotional problems during no to mild pain states. Furthermore, independent of the flare‐up status, the severity of pain in FOP patients was found to be significantly anti‐correlated with emotional health, physical health, and overall quality‐of‐life. These findings strongly suggest the need for an improved understanding of pain and emotional health in FOP during flare‐up and quiescent periods. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ke Peng
- Center for Pain and the Brain Department of Anesthesiology Critical Care and Pain Medicine Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Kin Cheung
- BioSAS Consulting, Inc. Wellesley MA USA
| | - Arielle Lee
- Center for Pain and the Brain Department of Anesthesiology Critical Care and Pain Medicine Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Christine Sieberg
- Center for Pain and the Brain Department of Anesthesiology Critical Care and Pain Medicine Boston Children's Hospital Harvard Medical School Boston MA USA.,Biobehavioral Pediatric Pain Laboratory Department of Psychiatry Boston Children's Hospital Boston MA USA
| | - David Borsook
- Center for Pain and the Brain Department of Anesthesiology Critical Care and Pain Medicine Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Jaymin Upadhyay
- Center for Pain and the Brain Department of Anesthesiology Critical Care and Pain Medicine Boston Children's Hospital Harvard Medical School Boston MA USA
| |
Collapse
|
38
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor (TGF)-β family of ligands and exert most of their effects through the canonical effectors Smad1, 5, and 8. Appropriate regulation of BMP signaling is critical for the development and homeostasis of numerous human organ systems. Aberrations in BMP pathways or their regulation are increasingly associated with diverse human pathologies, and there is an urgent and growing need to develop effective approaches to modulate BMP signaling in the clinic. In this review, we provide a wide perspective on diseases and/or conditions associated with dysregulated BMP signal transduction, outline the current strategies available to modulate BMP pathways, highlight emerging second-generation technologies, and postulate prospective avenues for future investigation.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, Indiana 46222
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts 02115
| |
Collapse
|
39
|
Alessi Wolken DM, Idone V, Hatsell SJ, Yu PB, Economides AN. The obligatory role of Activin A in the formation of heterotopic bone in Fibrodysplasia Ossificans Progressiva. Bone 2018; 109:210-217. [PMID: 28629737 PMCID: PMC6706059 DOI: 10.1016/j.bone.2017.06.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/02/2017] [Accepted: 06/15/2017] [Indexed: 11/25/2022]
Abstract
Fibrodysplasia Ossificans Progressiva (FOP) is a rare genetic disorder that presents at birth with only minor patterning defects, but manifests its debilitating pathology early in life with episodic, yet progressive and cumulative, heterotopic ossification (HO) of ligaments, tendons, and a subset of major skeletal muscles. The resulting HO lesions are endochondral in nature, and appear to be linked to inflammatory stimuli arising in association with known injuries, or from inflammation linked to normal tissue repair. FOP is caused by gain-of-function mutations in ACVR1, which encodes a type I BMP receptor. Initial studies on the pathogenic mechanism of FOP-causing mutations in ACVR1 focused on the enhanced function of this receptor in response to certain BMP ligands, or independently of ligands, but did not directly address the fact that HO in FOP is episodic and inflammation-driven. Recently, we and others demonstrated that Activin A is an obligate factor for the initiation of HO in FOP, signaling aberrantly via mutant ACVR1 to transduce osteogenic signals and trigger heterotopic bone formation (Hatsell et al., 2015; Hino et al., 2015). Subsequently, we identified distinct tissue-resident mesenchymal progenitor cells residing in muscles and tendons that recognize Activin A as a pro-osteogenic signal (solely in the context of FOP-causing mutant ACVR1), and give rise to the cartilaginous anlagen that form heterotopic bone (Dey et al., 2016). During the course of these studies, we also found that the activity of FOP-causing ACVR1 mutations does not by itself explain the triggered or inflammatory nature of HO in FOP, suggesting the importance of other, inflammation-introduced, factors or processes. This review presents a synthesis of these findings with a focus on the role of Activin A and inflammation in HO, and lays out perspectives for future research.
Collapse
Affiliation(s)
- Dana M Alessi Wolken
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Vincent Idone
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Sarah J Hatsell
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Aris N Economides
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA; Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA.
| |
Collapse
|
40
|
Mucha BE, Hashiguchi M, Zinski J, Shore EM, Mullins MC. Variant BMP receptor mutations causing fibrodysplasia ossificans progressiva (FOP) in humans show BMP ligand-independent receptor activation in zebrafish. Bone 2018; 109:225-231. [PMID: 29307777 PMCID: PMC5866198 DOI: 10.1016/j.bone.2018.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/23/2017] [Accepted: 01/03/2018] [Indexed: 11/28/2022]
Abstract
The large majority of cases of the autosomal dominant human disease fibrodysplasia ossificans progressiva (FOP) are caused by gain-of-function Arg206His mutations in the BMP type I receptor ACVR1 (ALK2). The Arg206His mutation is located in the GS domain of the type I receptor. This region is normally phosphorylated by the BMP type II receptor, which activates the type I receptor to phosphorylate its substrate, the signal transducer Smad1/5/8. A small subset of patients with FOP carry variant mutations in ACVR1 altering Gly328 to Trp, Glu or Arg. Since these mutations lie outside the GS domain, the mechanism through which ACVR1 Gly328 mutations cause disease remains unclear. We used a zebrafish embryonic development assay to test the signaling of human ACVR1 Gly328 mutant receptors comparing them to the Arg206His mutant. In this assay increased or decreased BMP pathway activation alters dorsal-ventral axial patterning, providing a sensitive assay for altered BMP signaling levels. We expressed the human ACVR1 Gly328 mutant receptors in zebrafish embryos to investigate their signaling activities. We found that all ACVR1 Gly328 human mutations ventralized wild-type embryos and could partially rescue Bmp7-deficient embryos, indicating that these mutant receptors can activate BMP signaling in a BMP ligand-independent manner. The degree of ventralization or rescue was similar among all three Gly328 mutants. Smad1/5 phosphorylation, a readout of BMP receptor signaling, was mildly increased by ACVR1 Gly328 mutations. Gene expression analyses demonstrate expanded ventral and reciprocal loss of dorsal cell fate markers. This study demonstrates that Gly328 mutants increase receptor activation and BMP ligand-independent signaling through Smad phosphorylation.
Collapse
Affiliation(s)
- Bettina E Mucha
- Division of Human Genetics and Molecular Biology, and Division of Biochemical Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Universite de Montreal, Montreal, QC, Canada
| | - Megumi Hashiguchi
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph Zinski
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Eileen M Shore
- Department of Orthopedic Surgery, and the Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Genetics, and the Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Cappato S, Giacopelli F, Ravazzolo R, Bocciardi R. The Horizon of a Therapy for Rare Genetic Diseases: A "Druggable" Future for Fibrodysplasia Ossificans Progressiva. Int J Mol Sci 2018; 19:ijms19040989. [PMID: 29587443 PMCID: PMC5979309 DOI: 10.3390/ijms19040989] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 12/21/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic condition characterized by progressive extra-skeletal ossification leading to cumulative and severe disability. FOP has an extremely variable and episodic course and can be induced by trauma, infections, iatrogenic harms, immunization or can occur in an unpredictable way, without any recognizable trigger. The causative gene is ACVR1, encoding the Alk-2 type I receptor for bone morphogenetic proteins (BMPs). The signaling is initiated by BMP binding to a receptor complex consisting of type I and II molecules and can proceed into the cell through two main pathways, a canonical, SMAD-dependent signaling and a p38-mediated cascade. Most FOP patients carry the recurrent R206H substitution in the receptor Glycine-Serine rich (GS) domain, whereas a few other mutations are responsible for a limited number of cases. Mutations cause a dysregulation of the downstream BMP-dependent pathway and make mutated ACVR1 responsive to a non-canonical ligand, Activin A. There is no etiologic treatment for FOP. However, many efforts are currently ongoing to find specific therapies targeting the receptor activity and the downstream aberrant pathway at different levels or targeting cellular components and/or processes that are important in modifying the local environment leading to bone neo-formation.
Collapse
Affiliation(s)
- Serena Cappato
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy.
| | - Francesca Giacopelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy.
| | - Roberto Ravazzolo
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy.
| | - Renata Bocciardi
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 16132 Genoa, Italy.
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy.
| |
Collapse
|
42
|
Upadhyay J, Xie L, Huang L, Das N, Stewart RC, Lyon MC, Palmer K, Rajamani S, Graul C, Lobo M, Wellman TJ, Soares EJ, Silva MD, Hesterman J, Wang L, Wen X, Qian X, Nannuru K, Idone V, Murphy AJ, Economides AN, Hatsell SJ. The Expansion of Heterotopic Bone in Fibrodysplasia Ossificans Progressiva Is Activin A-Dependent. J Bone Miner Res 2017; 32:2489-2499. [PMID: 28782882 DOI: 10.1002/jbmr.3235] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/23/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare autosomal dominant disorder that is characterized by episodic yet cumulative heterotopic ossification (HO) in skeletal muscles, tendons, and ligaments over a patient's lifetime. FOP is caused by missense mutations in the type I bone morphogenetic protein (BMP) receptor ACVR1. We have determined that the formation of heterotopic bone in FOP requires activation of mutant ACVR1 by Activin A, in part by showing that prophylactic inhibition of Activin A blocks HO in a mouse model of FOP. Here we piece together a natural history of developing HO lesions in mouse FOP, and determine where in the continuum of HO Activin A is required, using imaging (T2-MRI, μCT, 18 F-NaF PET/CT, histology) coupled with pharmacologic inhibition of Activin A at different times during the progression of HO. First, we show that expansion of HO lesions comes about through growth and fusion of independent HO events. These events tend to arise within a neighborhood of existing lesions, indicating that already formed HO likely triggers the formation of new events. The process of heterotopic bone expansion appears to be dependent on Activin A because inhibition of this ligand suppresses the growth of nascent HO lesions and stops the emergence of new HO events. Therefore, our results reveal that Activin A is required at least up to the point when nascent HO lesions mineralize and further demonstrate the therapeutic utility of Activin A inhibition in FOP. These results provide evidence for a model where HO is triggered by inflammation but becomes "self-propagating" by a process that requires Activin A. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
| | - LiQin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Lily Huang
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Lili Wang
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Xialing Wen
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | | | | | | | - Aris N Economides
- Regeneron Pharmaceuticals, Tarrytown, NY, USA.,Regeneron Genetics Center, Tarrytown, NY, USA
| | | |
Collapse
|
43
|
Kaplan FS, Al Mukaddam M, Pignolo RJ. A cumulative analogue joint involvement scale (CAJIS) for fibrodysplasia ossificans progressiva (FOP). Bone 2017; 101:123-128. [PMID: 28465250 DOI: 10.1016/j.bone.2017.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/26/2017] [Accepted: 04/26/2017] [Indexed: 11/13/2022]
Abstract
BACKGROUND Fibrodysplasia ossificans progressiva (FOP) is a catastrophic genetic disorder of progressive heterotopic ossification (HO). Assessment of functional mobility in FOP will be essential to support clinical trials of investigational agents. RESULTS Of necessity, we developed a simple, rapidly-administered, cumulative analogue joint involvement scale (CAJIS) for FOP based on assessments in 144 individuals worldwide with classic FOP. CONCLUSIONS CAJIS scores correlated with patient age, activities of daily living, and ambulatory function with excellent inter-rater variability. We show here that the CAJIS score provides an accurate and reproducible snapshot of total body and regional mobility burden in FOP.
Collapse
Affiliation(s)
- Frederick S Kaplan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; The Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Mona Al Mukaddam
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; The Center for Research in FOP and Related Disorders, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
44
|
Eekhoff EMW, Netelenbos JC, de Graaf P, Hoebink M, Bravenboer N, Micha D, Pals G, de Vries TJ, Lammertsma AA, Raijmakers PG, van Es RJ. Flare-Up After Maxillofacial Surgery in a Patient With Fibrodysplasia Ossificans Progressiva: An [ 18F]-NaF PET/CT Study and a Systematic Review. JBMR Plus 2017; 2:55-58. [PMID: 30283890 PMCID: PMC6124206 DOI: 10.1002/jbm4.10008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/21/2017] [Accepted: 05/22/2017] [Indexed: 01/18/2023] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder leading to progressive heterotopic ossifications (HO) of muscles, tendons, and ligaments, which can be induced by trauma or by surgery. Despite strong medical advice to the contrary, an FOP patient insisted on surgery to alleviate her complete trismus, which caused an unbearable impact on her quality of life (QOL). The entire trismus history of this FOP patient is presented. [18F]‐NaF position emission tomography/computed tomography (PET/CT) scans were introduced as an imaging method for heterotopic bone formation activity. To place our findings into context, a systematic review on jaw surgery in FOP was performed. After falling down the stairs, a 9‐year‐old patient developed mobility impairment of her left‐sided jaw. During the following 13 years bone scintigraphy showed persistent activity of the disease leading to progressive left‐sided zygomatico‐mandibular fusion by HO, resulting in complete trismus. Within 1 month after HO removal on the left side and a matching right coronoidectomy, [18F]‐NaF PET/CT demonstrated a substantial flare‐up activity followed by new HO in both masseter and temporalis muscles. Despite recurrent HO and trismus her QOL increased due to a stable increased interincisal opening of 5.5 mm. Although systematic review reveals a 100% risk of HO recurrence after jaw surgery, information on improved QOL is scarce. In conclusion, surgery in FOP may be beneficial for QOL despite new HO formation. Assessment of disease activity using [18F]‐NaF PET/CT is possible before HO is evident on CT and may serve as a new and quantitative marker of the disease. © 2017 The Authors. JBMR Plus Published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- E Marelise W Eekhoff
- Department of Internal Medicine Section Endocrinology VU University Medical Center Amsterdam The Netherlands
| | - J Coen Netelenbos
- Department of Internal Medicine Section Endocrinology VU University Medical Center Amsterdam The Netherlands
| | - Pim de Graaf
- Department of Radiology & Nuclear Medicine VU University Medical Center Amsterdam The Netherlands
| | - Max Hoebink
- Department of Internal Medicine Section Endocrinology VU University Medical Center Amsterdam The Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Genetics and Bone Histomorphology VU University Medical Center Amsterdam The Netherlands
| | - Dimitra Micha
- Department of Clinical Genetics and Bone Histomorphology VU University Medical Center Amsterdam The Netherlands
| | - Gerard Pals
- Department of Clinical Genetics and Bone Histomorphology VU University Medical Center Amsterdam The Netherlands
| | - Teun J de Vries
- Department Periodontology Academic Centre for Dentistry Amsterdam (ACTA) University of Amsterdam and VU University Amsterdam The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine VU University Medical Center Amsterdam The Netherlands
| | - Pieter Ghm Raijmakers
- Department of Radiology & Nuclear Medicine VU University Medical Center Amsterdam The Netherlands
| | - Robert Jj van Es
- Department of Oral and Maxillofacial Surgery Utrecht University Medical Center Utrecht The Netherlands
| |
Collapse
|
45
|
Di Rocco M, Baujat G, Bertamino M, Brown M, De Cunto CL, Delai PLR, Eekhoff EMW, Haga N, Hsiao E, Keen R, Morhart R, Pignolo RJ, Kaplan FS. International physician survey on management of FOP: a modified Delphi study. Orphanet J Rare Dis 2017; 12:110. [PMID: 28606101 PMCID: PMC5468985 DOI: 10.1186/s13023-017-0659-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/19/2017] [Indexed: 11/10/2022] Open
Abstract
Fibrodysplasia ossificans progressiva (FOP), a disabling disorder of progressive heterotopic ossification (HEO), is caused by heterozygous gain-of- function mutations in Activin receptor A, type I (ACVR1, also known as ALK2), a bone morphogenetic protein (BMP) type I receptor. Presently, symptomatic management is possible, but no definitive treatments are available. Although extensive guidelines for symptomatic management are widely used, regional preferences exist. In order to understand if there was worldwide consensus among clinicians treating FOP patients, an expert panel of physicians directly involved in FOP patient care was convened. Using a modified Delphi method, broad international consensus was reached on four main topics: diagnosis, prevention of flare-ups, patient and family-centered care and general clinical management issues. This study of physician preferences provides a basis for standardization of clinical management for FOP.
Collapse
Affiliation(s)
- Maja Di Rocco
- Department of Pediatrics, Unit of Rare Diseases, Giannina Gaslini Institute, Largo Gaslini 5, 16147, Genoa, Italy.
| | - Genevieve Baujat
- Service of Medical Genetics CHU Paris - Hôpital Necker-Enfants Malades, 149 rue de Sèvres, 75743, Paris, France
| | - Marta Bertamino
- Department of Pediatrics, Unit of Rare Diseases, Giannina Gaslini Institute, Largo Gaslini 5, 16147, Genoa, Italy
| | - Matthew Brown
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Woolloongabba, QLD, 4069, Australia
| | - Carmen L De Cunto
- Department of Pediatrics, Pediatric Rheumatology Section, Hospital Italiano de Buenos Aires, Gascón 450, 1181, Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia L R Delai
- Orthopaedic Department of Santa Casa de Misericórdia de São Paulo, School of Medicine Faculdade de Ciências Médicas da Santa Casa de São Paulo, Rua Pedro de Toledo 129 cj 121, Vila Clementino, 04039-001, São Paulo, Brazil
| | - Elisabeth M W Eekhoff
- Department of Internal Medicine/Section Endocrinology, VU Medical Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Nobuhiko Haga
- Department of Rehabilitation Medicine Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Edward Hsiao
- Department of Endocrinology, Faculty Practice University of California-San Francisco, 400 Parnassus Ave., San Francisco, CA, 94143-1222, USA
| | - Richard Keen
- University College London Hospitals, London, NW1 2PQ, UK
| | - Rolf Morhart
- Department of Pediatrics Klinikum Garmisch-Partenkirchen GmbH, Auenstraße 6, 82467, Garmisch-Partenkirchen, Germany
| | - Robert J Pignolo
- Department of Medicine, Division of Geriatric Medicine & Gerontology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, USA
| | - Frederick S Kaplan
- Department of Orthopaedic Surgery, Center for Research in FOP & Related Disorders, The Perelman School of Medicine, The University of Pennsylvania, 3737 Market Street, Philadelphia, PA, 19104, USA
| |
Collapse
|
46
|
Anomalie réductionnelle transverse et fibrodysplasie ossifiante progressive atypique, à propos d’un cas de diagnostic tardif. Arch Pediatr 2017; 24:547-551. [DOI: 10.1016/j.arcped.2017.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 12/01/2016] [Accepted: 03/14/2017] [Indexed: 11/23/2022]
|
47
|
The congenital great toe malformation of fibrodysplasia ossificans progressiva? - A close call. Eur J Med Genet 2017; 60:399-402. [PMID: 28473268 DOI: 10.1016/j.ejmg.2017.04.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/03/2017] [Accepted: 04/29/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Congenital bilateral hallux valgus with associated absence or fusion of the interphalangeal joint is a classic diagnostic feature of fibrodysplasia ossificans progressiva (FOP), a human genetic disease of extra-skeletal bone formation caused in nearly all cases by a gain-of-function mutation in Activin A Receptor I/Activin-like Kinase 2 (ACVR1/ALK2), which encodes a bone morphogenetic protein (BMP) Type 1 receptor. This toe malformation prompts the suspicion of FOP even before the appearance of extra-skeletal bone. Here we report the case of a four-month-old child who was suspected of having FOP on the basis of a great toe malformation identical to that seen in children with the disease. METHODS The patient's genomic DNA of the coding region of ACVR1 was sequenced and analyzed for mutations known to cause FOP and novel mutations. Subsequent comparative genomic hybridization (CGH) and single nucleotide polymorphism (SNP) analyses were performed to detect mutations elsewhere in the genome. RESULTS Genetic testing exonerated ACVR1 as culpable for the patient's toe malformation. CGH and SNP analyses identified a large intragenic deletion in a different BMP Type 1 receptor gene, BMP Receptor 1B/Activin-like kinase 6 (BMPR1B/ALK6), a gene associated with a variable spectrum of autosomal dominant brachydactyly phenotypes. CONCLUSIONS This report illustrates that while toe morphology remains the earliest indicator of FOP, toe morphology alone is not an unequivocal clinical diagnostic feature of FOP, and supports that embryonic development of the great toe is highly sensitive to dysregulated signaling from at least two BMP type I receptors.
Collapse
|
48
|
Sferopoulos NK, Kotakidou R, Petropoulos AS. Myositis ossificans in children: a review. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2017; 27:491-502. [DOI: 10.1007/s00590-017-1932-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 02/16/2017] [Indexed: 12/29/2022]
|
49
|
Yamamoto Y, Ihara M. Disruption of transforming growth factor-β superfamily signaling: A shared mechanism underlying hereditary cerebral small vessel disease. Neurochem Int 2016; 107:211-218. [PMID: 28034724 DOI: 10.1016/j.neuint.2016.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 11/20/2022]
Abstract
Cerebral small vessel disease (SVD) is not only one of the leading causes of cognitive impairment but also an important contributory factor in Alzheimer's disease. SVD and related white matter changes are common in the elderly, but the underlying pathogenic mechanism remains unclear. The end-stage pathology of SVD often involves replacement of vascular smooth muscle cells with collagenous or other nontensile fibrillary material. Recent studies on hereditary SVD have revealed a close relationship between small vessel pathology and disruption of transforming growth factor-β (TGF-β) superfamily signaling. TGF-β superfamily members, such as TGF-β and bone morphogenetic proteins, are multifunctional proteins that regulate production of extracellular matrix proteins, which in turn control the bioavailability of TGF-β superfamily members and modulate their signaling activities. This article reviews hereditary disorders with small vessel pathology and their relation to TGF-β superfamily signaling.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishirodai, Suita, Osaka, 565-8565, Japan.
| |
Collapse
|
50
|
Mucke HA. Drug Repurposing Patent Applications July–September 2016. Assay Drug Dev Technol 2016; 14:577-582. [DOI: 10.1089/adt.2016.29053.pq3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|