1
|
Mineta K, Nishisho T, Okada M, Kamada M, Sairyo K. Real-world safety and effectiveness of romosozumab following daily or weekly administration of teriparatide in primary and secondary osteoporosis. Bone 2025; 193:117392. [PMID: 39826700 DOI: 10.1016/j.bone.2025.117392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Romosozumab is an anti-sclerostin antibody that increases bone formation and decreases bone resorption, and it became available for patients at high risk of osteoporotic fractures in Japan in 2019. The aim of this study was to clarify the clinical effects, safety, and predictors of the effectiveness of 12 months of romosozumab therapy following daily or weekly administration of teriparatide. The study had an observational pre-post design and included 171 female patients. Romosozumab was administered at a dose of 210 mg subcutaneously every 4 weeks for 12 months following daily or weekly administration of teriparatide. The incidence of new fractures, safety, and changes in bone mineral density (BMD) and bone turnover markers were recorded. New fractures occurred in 3 cases (2.2 %). Four patients (2.3 %) with secondary osteoporosis experienced cardiovascular events, which were fatal in 1 patient (0.6 %). The percent changes in BMD at the spine and total hip at 12 months from baseline were + 7.9 % and + 2.4 %, respectively. The percent change in spine BMD did not significantly differ according to whether daily or weekly teriparatide was given as previous treatment. Romosozumab following teriparatide showed greater effectiveness in patients with primary osteoporosis, high P1NP level at 1 month, and low percent changes in TRACP-5b after 12 months of treatment. Romosozumab after treatment with daily or weekly teriparatide was relatively safe and more effective in patients with primary osteoporosis than in those with secondary osteoporosis.
Collapse
Affiliation(s)
- Kazuaki Mineta
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Toshihiko Nishisho
- Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Masahiko Okada
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Mitsuhiro Kamada
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Koichi Sairyo
- Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
2
|
Mäkinen VN, Sølling AS, McClung M, Langdahl BL. Romosozumab for the treatment of osteoporosis - a systematic review. J Endocrinol Invest 2025; 48:547-572. [PMID: 39487940 DOI: 10.1007/s40618-024-02469-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/07/2024] [Indexed: 11/04/2024]
Abstract
INTRODUCTION Romosozumab, a new treatment of osteoporosis, is a monoclonal antibody that targets sclerostin and thereby exhibits a dual mechanism of action by stimulating bone formation and inhibiting bone resorption. This systematic review aims to assess the clinical efficacy and safety of romosozumab for treatment of primary and secondary osteoporosis. METHODS A comprehensive literature search was conducted in October 2023 across multiple databases including Embase, PubMed and Cochrane Library. Randomized controlled trials (RCTs) and observational studies evaluating the impact of romosozumab on BMD, bone turnover markers (BTM), fracture outcomes, and its safety profile were included. Data extraction and quality assessment were performed independently by two reviewers in accordance with PRISMA guidelines. RESULTS A total of 36 articles met the inclusion criteria. Romosozumab significantly increased BMD at the lumbar spine, total hip, and femoral neck compared to placebo and active comparators in patients with primary osteoporosis. Sequential therapy with romosozumab followed by antiresorptives maintained or further increased BMD and reduced fracture risk. Romosozumab was generally well tolerated, however, an imbalance in cardiovascular adverse event was observed in one large clinical trial. Observational studies supported these findings. Specific subgroups of patients with secondary osteoporosis were assessed, demonstrating overall positive outcomes with romosozumab treatment. CONCLUSION Romosozumab effectively increases BMD and reduces fracture risk, particularly when used as initial therapy in high fracture-risk patients. Sequential therapy with subsequent antiresorptive treatment optimizes long-term benefits. While generally well-tolerated, its cardiovascular safety profile requires further long-term studies to ensure its safety in clinical practice. Additional studies are needed to confirm efficacy and safety in patients with secondary osteoporosis.
Collapse
Affiliation(s)
- V-N Mäkinen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | - A S Sølling
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - M McClung
- Oregon Osteoporosis Center, Portland, OR, USA
| | - B L Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Fuggle N, Laslop A, Rizzoli R, Al-Daghri N, Alokail M, Balkowiec-Iskra E, Beaudart C, Bruyère O, Bemden ABV, Burlet N, Cavalier E, Cerreta F, Chandran M, Cherubini A, da Silva Rosa MMC, Conaghan P, Cortet B, Jentoft AC, Curtis EM, D'Amelio P, Dawson-Hughes B, Dennison EM, Hiligsmann M, Kaufman JM, Maggi S, Matijevic R, McCloskey E, Messina D, Pinto D, Yerro MCP, Radermecker RP, Rolland Y, Torre C, Veronese N, Kanis JA, Cooper C, Reginster JY, Harvey NC. Treatment of Osteoporosis and Osteoarthritis in the Oldest Old. Drugs 2025; 85:343-360. [PMID: 39969778 PMCID: PMC11891106 DOI: 10.1007/s40265-024-02138-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 02/20/2025]
Abstract
Osteoporosis and osteoarthritis are key diseases of musculoskeletal ageing and are increasing in prevalence and burden with the progressively ageing population worldwide. These conditions are thus particularly common in 'the oldest old', and there are complexities of managing them within the context of extensive multimorbidity, physical and mental disability, and polypharmacy, the rates for all of which are high in this population. In this narrative review, we explore the epidemiology of osteoporosis and osteoarthritis in the oldest old before examining trials and real-world data relating to the pharmacological treatment of these diseases in older adults, including anti-resorptives and bone-forming agents in osteoporosis and symptomatic slow-acting drugs for osteoarthritis, paracetamol, and non-steroidal anti-inflammatory drugs in osteoarthritis, recognising that the oldest old are usually excluded from clinical trials. We then review the potential benefits of nutritional interventions and exercise therapy before highlighting the health economic benefits of interventions for osteoporosis and osteoarthritis. The high prevalence of risk factors for both disease and adverse events associated with treatment in the oldest old mean that careful attention must be paid to the potential benefits of intervention (including fracture risk reduction and improvements in osteoarthritis pain and function) versus the potential harms and adverse effects. Further direct evidence relating to such interventions is urgently needed from future research.
Collapse
Affiliation(s)
- Nicholas Fuggle
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
| | - Andrea Laslop
- Scientific Office, Austrian Medicines and Medical Devices Agency, Vienna, Austria
| | - René Rizzoli
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Nasser Al-Daghri
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Majed Alokail
- Protein Research Chair, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Ewa Balkowiec-Iskra
- Department of Experimental and Clinical Pharmacology, Medical University of Warsaw, Warsaw, Poland
- The Office for Registration of Medicinal Products, Medical Devices and Biocidal Products, Warsaw, Poland
| | - Charlotte Beaudart
- Clinical Pharmacology and Toxicology Research Unit, Department of Biomedical Sciences, Faculty of Medicine, NARILIS, University of Namur, Namur, Belgium
| | - Olivier Bruyère
- Research Unit in Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
| | | | - Nansa Burlet
- The European Society for Clinical and Economic Aspects of Osteoporosis, Osteoarthritis and Musculoskeletal Diseases (ESCEO), Liege, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CIRM, University of Liège, CHU de Liège, Liège, Belgium
| | | | - Manju Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, Singapore, Singapore
- DUKE NUS Medical School, Singapore, Singapore
| | - Antonio Cherubini
- Geriatria, Accettazione geriatrica e Centro di ricerca per l'invecchiamento, IRCCS INRCA, Ancona, Italy
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | | | - Philip Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Bernard Cortet
- Department of Rheumatology, University of Lille, Lille, France
| | - Alfonso Cruz Jentoft
- Servicio de Geriatría. Hospital Universitario Ramón y Cajal (IRYIS), Madrid, Spain
| | - Elizabeth M Curtis
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
| | - Patrizia D'Amelio
- Department of Geriatrics and Geriatric Rehabilitation, Lausanne University Hospital, Lausanne, Switzerland
| | - Bess Dawson-Hughes
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Elaine M Dennison
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
| | - Mickaël Hiligsmann
- Department of Health Services Research, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Jean-Marc Kaufman
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | | | - Radmila Matijevic
- Faculty of Medicine, Clinic for Orthopedic Surgery and Traumatology, Clinical Center of Vojvodina, University of Novi Sad, Novi Sad, Serbia
| | - Eugene McCloskey
- Division of Clinical Medicine, School of Medicine and Population Health, Centre for Integrated Research in Musculoskeletal Ageing, University of Sheffield, Sheffield, UK
| | - Daniel Messina
- IRO Investigaciones Reumatologicas y Osteologicas SRL Collaborating Centre WHO, University of Buenos Aires, Buenos Aires, Argentina
| | - Daniel Pinto
- Department of Physical Therapy, Marquette University, Milwaukee, WI, USA
| | | | - Régis Pierre Radermecker
- Department of Diabetes, Nutrition and Metabolic disorders, Clinical pharmacology, University of Liège, CHU de Liège, Liège, Belgium
| | - Yves Rolland
- IHU Health Age, CHU Toulouse, INSERM 1295, Toulouse, France
| | - Carla Torre
- Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines of the University of Lisbon (iMED.ULisboa), Avenida Professor Gama Pinto, 1649-003, Lisbon, Portugal
| | - Nicola Veronese
- Chair for Biomarkers of Chronic Diseases, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
- Geriatric Unit, Department of Medicine, University of Palermo, 90127, Palermo, Italy
| | - John A Kanis
- Division of Clinical Medicine, School of Medicine and Population Health, Centre for Integrated Research in Musculoskeletal Ageing, University of Sheffield, Sheffield, UK
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jean-Yves Reginster
- Protein Research Chair, Biochemistry Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
- Research Unit in Public Health, Epidemiology and Health Economics, University of Liège, Liège, Belgium
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, SO16 6YD, UK.
- NIHR Southampton Biomedical Research Centre, University of Southampton, Southampton, UK.
| |
Collapse
|
4
|
Cheng SH, Chu W, Chou WH, Chu WC, Kang YN. Cardiovascular Safety of Romosozumab Compared to Commonly Used Anti-osteoporosis Medications in Postmenopausal Osteoporosis: A Systematic Review and Network Meta-analysis of Randomized Controlled Trials. Drug Saf 2025; 48:7-23. [PMID: 39227560 PMCID: PMC11711713 DOI: 10.1007/s40264-024-01475-9] [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: 08/06/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION The aim of this study was to investigate the cardiovascular safety of romosozumab in postmenopausal women with osteoporosis. Romosozumab, a monoclonal antibody targeting sclerostin, has been shown to increase bone mineral density and reduce the risk of osteoporotic fractures. However, in previous studies, romosozumab therapy was identified as a potential risk factor for cardiovascular events, particularly in patients with predisposing cardiovascular disease. METHODS A systematic literature search was performed in the Cochrane Library, Embase, PubMed, and Web of Science databases to identify randomized controlled trials (RCTs) comparing the safety and efficacy of romosozumab versus alendronate, teriparatide, denosumab, or placebo in postmenopausal women with osteoporosis. Contrast-based network meta-analysis was performed using a random-effects model. The pooled estimates are presented as risk ratios with 95% confidence intervals. RESULTS Of the 5282 articles retrieved, 25 RCTs were included in this review (n = 24,942), and 18 randomized controlled trials (n = 16,777) were included in the network meta-analysis. The results indicated no significant differences in cardiovascular mortality rate between romosozumab and placebo. Regarding the risk of major cardiovascular events, no significant differences were found in the direct evidence or the network meta-analysis with placebo as the reference. CONCLUSION Romosozumab might be a safe option for treating postmenopausal women with osteoporosis. The cardiovascular concerns associated with this treatment seem less significant than previously suggested, although additional real-world data are required to confirm this conclusion.
Collapse
Affiliation(s)
- Shih-Hao Cheng
- Department of Biomedical Engineering, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
- Department of Orthopedics, Wan Fang Hospital, Medical University Hospital, Taipei, Taiwan
| | - William Chu
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Wen-Hsiang Chou
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Woei-Chyn Chu
- Department of Biomedical Engineering, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.
| | - Yi-No Kang
- National Taipei University of Nursing and Health Sciences, Taipei, Taiwan.
- Evidence-Based Medicine Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Institute of Health Policy and Management, College of Public Health, National Taiwan University, Taipei, Taiwan.
- Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
5
|
Baroudi M, Daher M, Maheshwari K, Singh M, Nassar JE, McDonald CL, Diebo BG, Daniels AH. Surgical Management of Adult Spinal Deformity Patients with Osteoporosis. J Clin Med 2024; 13:7173. [PMID: 39685632 DOI: 10.3390/jcm13237173] [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: 10/26/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Adult spinal deformity (ASD) commonly affects older adults, with up to 68% prevalence in those over 60, and is often complicated by osteoporosis, which reduces bone mineral density (BMD) and increases surgical risks. Osteoporotic patients undergoing ASD surgery face higher risks of complications like hardware failure, pseudoarthrosis, and proximal junctional kyphosis (PJK). Medical management with antiresorptive medications (e.g., bisphosphonates, SERMs, and denosumab) and anabolic agents (e.g., teriparatide, abaloparatide, and romosozumab) can improve BMD and reduce complications. While bisphosphonates reduce fracture risk, teriparatide and newer agents like romosozumab show promise in increasing bone density and improving fusion rates. Surgical adaptations such as consideration of age-adjusted alignment, fusion level selection, cement augmentation, and the use of expandable screws or tethers enhance surgical outcomes in osteoporotic patients. Specifically, expandable screws and cement augmentation have been shown to improve fixation stability. However, further research is needed to evaluate the effectiveness of these treatments, specifically in osteoporotic ASD patients.
Collapse
Affiliation(s)
- Makeen Baroudi
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Mohammad Daher
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Krish Maheshwari
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Manjot Singh
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Joseph E Nassar
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Christopher L McDonald
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Bassel G Diebo
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Alan H Daniels
- Department of Orthopedic Surgery, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| |
Collapse
|
6
|
Wong RMY, Wong PY, Liu C, Wong HY, Fong MK, Zhang N, Cheung WH, Law SW. Treatment effects, adverse outcomes and cardiovascular safety of romosozumab - Existing worldwide data: A systematic review and meta-analysis. J Orthop Translat 2024; 48:107-122. [PMID: 39189010 PMCID: PMC11345130 DOI: 10.1016/j.jot.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Background Romosozumab is a novel monoclonal antibody that binds to sclerostin, and has dual effects of increasing bone formation and decreasing bone resorption, giving it a unique mechanism of action. The objective of this study was to perform a systematic review and meta-analysis based on existing worldwide data on treatment effects and safety of romosozumab in randomized controlled trials. Methods A systematic search was carried out on four databases including PubMed, Embase, Web of Science and Cochrane Central Register of Controlled Trials (CENTRAL). The keywords used for search was "(romosozumab) AND (osteoporosis OR safety)". Randomized controlled trial or post-hoc studies of the included randomized controlled trial which studied the effects and safety of romosozumab were included. The quality of selected studies was assessed with the Cochrane collaboration tool and the PEDro scale. Results 20 studies were included for qualitative analysis. 14 studies were included for meta-analysis. In total, there were 13,507 (n = 13,507) participants with 637 men and 12,870 women from original cohorts. The overall mean difference was in favor of romosozumab treatment for lumbar spine (10.04 (95 % confidence interval (CI) = 7.51-12.57; p < 0.00001)), total hip (4.04 (95 % CI = 3.10-4.99; p < 0.00001)) and femoral neck bone mineral density (3.77 (95 % CI = 2.90-4.64; p < 0.00001)) at 12 months. There was significantly less likelihood of new vertebral fractures with romosozumab compared to control (odds ratio (OR) 0.42 (95 % CI = 0.20-0.89); p = 0.02) at 12 months of treatment. There was significantly less likelihood of new vertebral fracture at 24 months with 12 months of romosozumab followed by sequential treatment with anti-resorptive compared to control with only anti-resorptive agent use (OR 0.36 (95 % CI = 0.18-0.71); p = 0.003). There was no significant difference in serious adverse events and fatal adverse events with use of romosozumab compared with control in our meta-analyses. There were no significant differences in serious cardiovascular events in Asian population of romosozumab with control group with 12 months of romosozumab treatment followed by 24 months of anti-resorptive agent with OR 1.09 (95 % CI = 0.40-2.96; P = 0.86). There was no significant difference between romosozumab group and control group for the median time to radiographic healing. Our qualitative analysis on Quantitative Computed Tomography (QCT), Finite element analysis (FEA) and bone biopsy analyses demonstrated that romosozumab improved parameters and measures in these domains as well. Conclusion In conclusion, our study showed that romosozumab was an effective agent to treat osteoporosis with high quality evidence. There were no significant differences in the adverse events, serious adverse events, fatal adverse events identified. Further subgroup analysis of cardiovascular events and cardiovascular death in the total population showed no differences either. The translational potential of this article Given the results, romosozumab is an effective agent to treat patients with very-high risk of osteoporotic fractures.
Collapse
Affiliation(s)
- Ronald Man Yeung Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pui Yan Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chaoran Liu
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hei Yuet Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Man Ki Fong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ning Zhang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Hoi Cheung
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sheung Wai Law
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Kobayashi T, Hara M, Shimanoe C, Morimoto T, Masaaki M, Ito K, Shimazaki T. Efficacy and safety of romosozumab: a meta-analysis of placebo-controlled trials. J Bone Miner Metab 2024; 42:492-502. [PMID: 38977437 DOI: 10.1007/s00774-024-01531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/17/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION We aimed to comprehensively compile placebo-controlled trials on the efficacy and safety of romosozumab (210 mg, subcutaneously, once monthly) in postmenopausal women and men with osteoporosis. MATERIALS AND METHODS PubMed, Google Scholar, and ClinicalTrials.gov were searched for relevant placebo-controlled trials (as of January 1, 2024). Percent change in bone mineral density (BMD), falls, fractures, and adverse events (AEs) after drug administration were collected. Risk ratios (RRs) and mean differences (MDs) with 95% confidence intervals (CIs) were calculated. RESULTS Six trials (7990 patients; follow-up period, 6-12 months) were included. Compared with placebo, romosozumab significantly increased lumbar spine BMD (MD = 12.69; 95% CI 11.10-14.29), total hip BMD (MD = 4.42; 95% CI 3.03-5.80), and femoral neck BMD (MD = 3.99; 95% CI 2.42-5.57) at 12 months. Romosozumab significantly decreased falls (RR = 0.80; 95% CI 0.68-0.93) and major osteoporotic fractures (RR = 0.37; 95% CI 0.25-0.54), but increased injection-site reactions (RR = 1.83; 95% CI 1.46-2.30) within 12 months. No significant differences were observed in other AEs (including cardiovascular AEs) within 12 months. CONCLUSION Romosozumab treatment resulted in a significant BMD gain, reduced falls and major osteoporotic fractures. It was generally well-tolerated, including the cardiovascular aspects. However, clinicians should consider the occurrence of minor AEs (e.g., injection-site reactions).
Collapse
Affiliation(s)
- Takaomi Kobayashi
- Department of Orthopaedic Surgery, Taku City Hospital, Saga, Japan.
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan.
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan.
- Department of Clinical Research, Amagi Chuo Hospital, Fukuoka, Japan.
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Chisato Shimanoe
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
- Department of Pharmacy, Saga University Hospital, Saga, Japan
| | - Tadatsugu Morimoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Mawatari Masaaki
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Koji Ito
- Department of Orthopaedic Surgery, Taku City Hospital, Saga, Japan
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Takafumi Shimazaki
- Department of Orthopaedic Surgery, Taku City Hospital, Saga, Japan
- Department of Orthopaedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
8
|
Mineta K, Nishisho T, Okada M, Kamada M, Sairyo K. Real-world effects, safety, and predictors of the effectiveness of romosozumab in primary and secondary osteoporosis: An observational study. Bone 2024; 186:117164. [PMID: 38871265 DOI: 10.1016/j.bone.2024.117164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
Romosozumab is an anti-sclerostin antibody that increases bone formation and decreases bone resorption. It became available for patients at high risk of osteoporotic fractures in Japan in 2019. The aim of this study was to clarify the clinical effects, safety, and predictors of the effectiveness of 12 months of romosozumab therapy. The study had an observational pre-post design and included 460 patients. Romosozumab was administered at a dose of 210 mg subcutaneously every 4 weeks for 12 months. The incidence of new fractures, safety, and changes in bone mineral density (BMD) and bone turnover markers were recorded. New fractures occurred in 11 cases (3.0 %). Nine patients (2.0 %) experienced cardiovascular events, which were fatal in 3 (0.65 %). Percent changes in BMD at the spine and total hip at 12 months from baseline were +7.7 % and +1.8 %, respectively. Romosozumab had better effects in patients with good renal function, low spine BMD, and high TRACP-5b at baseline and low TRACP-5b or high P1NP after 1 month of treatment. The percent change in spine BMD at 12 months was significantly lower in patients transitioning from denosumab than in those not previously treated with other anti-osteoporosis agents. Romosozumab is considered to be relatively safe in patients with primary osteoporosis compared to those with secondary osteoporosis. Romosozumab resulted in larger increases in spine BMD in patients with primary osteoporosis who were not previously treated with other anti-osteoporosis therapies and those with low spine BMD at the start of treatment.
Collapse
Affiliation(s)
- Kazuaki Mineta
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Toshihiko Nishisho
- Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan.
| | - Masahiko Okada
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Mitsuhiro Kamada
- Department of Orthopaedic Surgery, Tokushima Kensei Hospital, 4-9 Shimosuketo-cho, Tokushima 770-8547, Japan
| | - Koichi Sairyo
- Department of Orthopedics, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
9
|
Gao G, Cui J, Xie Y, Dong J. Effects of romosozumab combined with routine therapy on pain relief, disease progression and adverse reactions in patients with postmenopausal osteoporosis: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1440948. [PMID: 39206178 PMCID: PMC11349545 DOI: 10.3389/fmed.2024.1440948] [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: 05/30/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Background Postmenopausal osteoporosis (PMOP) increases fracture risk in women. Though traditional treatments are slow to act, combining romosozumab with conventional therapy shows promise. Despite its growing use, studies on effectiveness are limited. This study aims to systematically evaluate the combined therapy's impact on pain relief, disease progression, and adverse reactions in PMOP patients. Methods Databases including PubMed, EMBASE, ScienceDirect, and the Cochrane Library were searched from their inception to September 2023 to identify randomized controlled trials (RCTs) evaluating the role of romosozumab in PMOP. Random or fixed effect models were employed for statistical analysis. Two reviewers independently assessed the quality of the included studies and extracted the data. The meta-analysis was conducted using RevMan 5.4 software. Results Six RCTs with a total sample size of 17,985 cases were included. The incidence of vertebral fractures was compared and analyzed after 12 and 24 months of treatment. Romosozumab significantly reduced the incidence of vertebral fractures at 24 months (OR = 0.36; 95% CI: 0.35-0.52) but not at 12 months (OR = 0.39; 95% CI: 0.14-1.05). It was also associated with a decreased incidence of nonvertebral fractures (OR = 0.79; 95% CI: 0.66-0.94) and clinical fractures at 24 months (OR = 0.70; 95% CI: 0.59-0.82) compared to standard therapy. Romosozumab demonstrated a significant improvement in percentage change in bone mineral density (BMD) [mean difference (MD) = 10.38; 95% CI: 4.62-16.14] and in hip joint BMD (MD = 4.24; 95% CI: 2.92-5.56). There was no notable difference in adverse reactions compared to standard care (p > 0.05). Funnel plots displayed a predominantly symmetrical pattern, suggesting no evidence of publication bias in the selected literature. Conclusion Combining romosozumab with conventional therapy effectively treats PMOP, significantly reducing vertebral, non-vertebral, and clinical fractures while increasing BMD in the hip, femoral neck, and lumbar spine. However, further high-quality studies are needed for validation.
Collapse
Affiliation(s)
- Ge Gao
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jian Cui
- The First Affiliated Hospital of Shandong First Medical University (Shandong Qianfo Mountain Hospital), Jinan, China
| | - Yuanyuan Xie
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Dong
- The First Affiliated Hospital of Shandong First Medical University (Shandong Qianfo Mountain Hospital), Jinan, China
| |
Collapse
|
10
|
Miyauchi A, Hamaya E, Shimauchi J, Yoshinaga Y, Nishi K. Effectiveness of romosozumab in patients with osteoporosis at high fracture risk: a Japanese real-world study. J Bone Miner Metab 2024; 42:77-89. [PMID: 38086988 DOI: 10.1007/s00774-023-01477-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/25/2023] [Indexed: 02/10/2024]
Abstract
INTRODUCTION To describe the real-world use of romosozumab in Japan, we conducted a chart review of > 1000 Japanese patients with osteoporosis (OP) at high risk of fracture, across multiple medical institutions. MATERIALS AND METHODS Treatment-naïve and prior OP-treatment patients who received romosozumab for 12 months followed by ≥ 6 months of sequential OP treatment were included. The primary objective described the baseline demographics and clinical characteristics; secondary objectives evaluated changes in bone mineral density (BMD) and bone turnover markers in all patients and effectiveness of romosozumab in a sub-group of treatment-naïve patients using the fracture risk assessment tool (FRAX®). RESULTS Of the 1027 patients (92.4% female), 45.0% were treatment-naïve. The mean ± SD age of treatment-naïve versus prior OP-treatment patients was 76.8 ± 8.5 and 77.1 ± 8.5 years. The most frequent prior OP treatment was bisphosphonates (45.0%). Romosozumab treatment for 12 months increased BMD at the lumbar spine in all groups; the median percent change from baseline in lumbar spine BMD was higher in the treatment-naïve (13.4%) versus prior OP-treatment group (bisphosphonates [9.2%], teriparatide [11.3%], denosumab [DMAb, 4.5%]). DMAb, bisphosphonates, or teriparatide after romosozumab maintained the BMD gains at all skeletal sites at month 18 in treatment-naïve patients. Most treatment-naïve patients were at high risk of fracture, BMD increased consistently with romosozumab regardless of the baseline fracture risk assessed by FRAX. CONCLUSION This large-scale, multicenter chart review provides clinically relevant insights into the profiles of patients initiating romosozumab, effectiveness of real-world romosozumab use, and sequential therapy in Japanese patients at high risk of fracture.
Collapse
Affiliation(s)
| | - Etsuro Hamaya
- Amgen K.K., Midtown Tower 9-7-1 Akasaka, Minato-ku, Tokyo, 107-6239, Japan.
| | | | - Yoko Yoshinaga
- Amgen K.K., Midtown Tower 9-7-1 Akasaka, Minato-ku, Tokyo, 107-6239, Japan
| | - Kiyoshi Nishi
- Amgen K.K., Midtown Tower 9-7-1 Akasaka, Minato-ku, Tokyo, 107-6239, Japan
| |
Collapse
|
11
|
Yavropoulou MP, Kasdagli MI, Makras P, Diomatari KM, Anastasilakis AD, Mitsikostas DD, Kassi E, Sfikakis PP, Kravvariti E. Nocebo-associated treatment discontinuation with subcutaneous anti-osteoporotic drugs. A systematic review and meta-analysis of placebo-arm dropouts in randomized-controlled trials. Maturitas 2024; 179:107874. [PMID: 37976923 DOI: 10.1016/j.maturitas.2023.107874] [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: 05/29/2023] [Revised: 09/22/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Nocebo is a concept of therapeutics referring to unpleasant symptoms attributed by a patient to a drug, due to negative anticipation. Patients receiving oral anti-osteoporotic drugs in randomized controlled trials (RCT) can experience adverse events leading to dropout, implying that nocebo contributes to treatment discontinuation for these drugs. In this study we aim to investigate the nocebo effect of subcutaneous anti-osteoporotic drugs with a higher compliance rate than orally administered drugs. STUDY DESIGN We searched MEDLINE, EMBASE, SCOPUS, and Cochrane databases for double-blind trials investigating subcutaneous anti-osteoporotic drugs for osteoporosis (namely, denosumab, teriparatide, abaloparatide and romosozumab) published up to May 2023. MAIN OUTCOME MEASURE Dropouts due to reported adverse events in the placebo arms ("nocebo dropouts"). RESULTS Data from 17 trials were extracted. Among 10,529 placebo-treated patients the pooled nocebo-dropout percentage was 3 % for denosumab (average: 0.03; 95 % CI: 0.01-0.05), 1 % for romosozumab (average: 0.01; 95 % CI: 0.00-0.03) and 6 % for teriparatide and abaloparatide (average: 0.06; 95 % CI: 0.05-0.07). Nocebo-dropouts were significantly higher in men than women (6 % vs. 3 %, respectively, p = 0.012), in older (mean age >68 years) than in younger patients (5 % vs. 1 %, respectively, p = 0.017) and in those with more severe osteoporosis (based on the percentage of participants with prior fragility-related fractures in the study cohort) compared with patients with no prior fracture history (4 % vs. 1 %, respectively, p = 0.046). CONCLUSION Nocebo responses may contribute to treatment discontinuation with subcutaneous anti-osteoporotic drugs in clinical practice. Higher nocebo-related dropout rates in the higher-risk RCT population (older patients, males, those with prior fractures) show that nocebo mechanisms have the potential to hinder therapeutic efforts to specific populations who would benefit most. Prospero registration number CRD42020212843.
Collapse
Affiliation(s)
- Maria P Yavropoulou
- Endocrinology Unit, 1st Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece.
| | - Maria-Iosifina Kasdagli
- Department of Hygiene and Epidemiology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Polyzois Makras
- Department of Medical Research, 251 Hellenic Air Force & VA General Hospital, Athens, Greece
| | - Konstantina-Maria Diomatari
- Postgraduate Medical Studies in the Physiology of Aging and Geriatric Syndromes, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Dimos D Mitsikostas
- 1st Neurology Department, Aeginition Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Eva Kassi
- Endocrinology Unit, 1st Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece
| | - Petros P Sfikakis
- Endocrinology Unit, 1st Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece; 1st Department of Propaedeutic and Internal Medicine, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece
| | - Evrydiki Kravvariti
- Endocrinology Unit, 1st Department of Propaedeutic and Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece; 1st Department of Propaedeutic and Internal Medicine, Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Laikon University Hospital of Athens, 11527 Athens, Greece
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW The role of wnt signalling in atherogenesis raises the possibility that the wnt inhibitor, sclerostin, provides a natural defence to this process, and that anti-sclerostin antibodies might increase the risk of atherosclerosis and associated conditions such as CVD. This article aims to triangulate evidence concerning possible adverse effects of sclerostin inhibition on CVD risk. RECENT FINDINGS Randomised controlled trials of treatment with the anti-sclerostin antibody, romosozumab, have yielded conflicting evidence with respect to possible adverse effects of sclerostin inhibition on CVD risk. To further examine the causal relationship between sclerostin inhibition and CVD risk, three Mendelian randomisation (MR) studies have examined effects of sclerostin lowering on CVD outcomes, using common genetic variants in the SOST gene which produces sclerostin, to mimic effects of a randomised trial. Concordant findings were seen in two studies, comprising an effect of sclerostin lowering on increased risk of MI and type II diabetes mellitus. One study also suggested that sclerostin lowering increases coronary artery calcification. Triangulation of evidence from different sources provides some suggestion that sclerostin lowering increases MI risk, supporting the need for CVD risk assessment when considering treatment with romosozumab.
Collapse
Affiliation(s)
- Jonathan H Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
13
|
Wu D, Li L, Wen Z, Wang G. Romosozumab in osteoporosis: yesterday, today and tomorrow. J Transl Med 2023; 21:668. [PMID: 37759285 PMCID: PMC10523692 DOI: 10.1186/s12967-023-04563-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
Osteoporosis is a systemic bone disease characterized by low bone mass, microarchitectural deterioration, increased bone fragility, and fracture susceptibility. It commonly occurs in older people, especially postmenopausal women. As global ageing increases, osteoporosis has become a global burden. There are a number of medications available for the treatment of osteoporosis, categorized as anabolic and anti-resorptive. Unfortunately, there is no drugs which have dual influence on bone, while all drugs have limitations and adverse events. Some serious adverse events include jaw osteonecrosis and atypical femoral fracture. Recently, a novel medication has appeared that challenges this pattern. Romosozumab is a novel drug monoclonal antibody to sclerostin encoded by the SOST gene. It has been used in Japan since 2019 and has achieved promising results in treating osteoporosis. However, it is also accompanied by some controversy. While it promotes rapid bone growth, it may cause serious adverse events such as cardiovascular diseases. There has been scepticism about the drug since its inception. Therefore, the present review comprehensively covered romosozumab from its inception to its clinical application, from animal studies to human studies, and from safety to cost. We hope to provide a better understanding of romosozumab for its clinical application.
Collapse
Affiliation(s)
- Dong Wu
- Department of Orthopeadics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China
| | - Lei Li
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhun Wen
- Department of Orthopaedics, Zhuanghe Central Hospital, Zhuanghe City, 116499, Liaoning Province, China.
| | - Guangbin Wang
- Department of Orthopeadics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China.
| |
Collapse
|
14
|
Nugraha AP, Ramadhani NF, Riawan W, Ihsan IS, Ernawati DS, Ridwan RD, Narmada IB, Saskianti T, Rezkita F, Sarasati A, Noor TNEBTA, Inayatillah B, Nugraha AP, Joestandari F. Gingival Mesenchymal Stem Cells Metabolite Decreasing TRAP, NFATc1, and Sclerostin Expression in LPS-Associated Inflammatory Osteolysis In Vivo. Eur J Dent 2023; 17:881-888. [PMID: 35728613 PMCID: PMC10569879 DOI: 10.1055/s-0042-1748529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE Bone is a dynamic tissue that undergoes remodeling. During bone remodeling, there are transcription factors such as nuclear factor-activated T cells-1 (NFATc1), sclerostin, and tartrate-resistant acid phosphatase (TRAP) that are released for bone resorption. Metabolite from gingival mesenchymal stem cells (GMSCs) has the ability to activate proliferation, migration, immunomodulation, and tissue regeneration of bone cells and tissues. Furthermore, the aim of this study is to investigate the metabolite of GMSCs' effect on expression of NFATc1, TRAP, and sclerostin in calvaria bone resorption of Wistar rats. MATERIALS AND METHODS Twenty male healthy Wistar rats (Rattus norvegicus), 1 to 2 months old, 250 to 300 g body were divided into four groups, namely group 1 (G1): 100 µg phosphate-buffered saline day 1 to 7; group 2 (G2): 100 μg lipopolysaccharide (LPS) day 1 to 7; group 3 (G3): 100 μg LPS + 100 μg GMSCs metabolite day 1 to 7; and group 4 (G4): 100 μg GMSCs metabolite day 1 to 7. Escherichia coli LPS was used to induce inflammatory osteolysis on the calvaria with subcutaneous injection. GMSCs metabolite was collected after passage 4 to 5, then injected subcutaneously on the calvaria. All samples were sacrificed on the day 8 through cervical dislocation. The expression of TRAP, NFATc1, and sclerostin of osteoclast in the calvaria was observed with 1,000× magnification. STATISTICAL ANALYSIS One-way analysis of variance and Tukey honest significant different were conducted to analyze differences between groups (p < 0.05). RESULTS The administration of GMSCs metabolite can significantly decrease TRAP, NFATc1, and sclerostin expression (p < 0.05) in LPS-associated inflammatory osteolysis calvaria in Wistar rats (R. norvegicus). There were significantly different TRAP, NFATc1, and sclerostin expressions between groups (p < 0.05). CONCLUSION GMSCs metabolite decrease TRAP, NFATc1, and sclerostin expression in LPS-associated osteolysis calvaria in Wistar rats (R. norvegicus) as documented immunohistochemically.
Collapse
Affiliation(s)
- Alexander Patera Nugraha
- Dental Regenerative Research Group, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Nastiti Faradilla Ramadhani
- Dental Regenerative Research Group, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Dentomaxillofacial Radiology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Wibi Riawan
- Department of Biomolecular Biochemistry, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Igo Syaiful Ihsan
- Stem Cell Research and Development Center, Universitas Airlangga Surabaya, Surabaya, Indonesia
| | - Diah Savitri Ernawati
- Department of Oral Medicine, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Rini Devijanti Ridwan
- Department of Oral Biology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ida Bagus Narmada
- Dental Regenerative Research Group, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Tania Saskianti
- Department of Pediatric Dentistry, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Fianza Rezkita
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Andari Sarasati
- Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Bilqis Inayatillah
- Department of Basic Medical of Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | | |
Collapse
|
15
|
Tominaga A, Wada K, Okazaki K, Nishi H, Terayama Y, Shimamoto S, Kodama Y, Kato Y. Nonresponder Considerations for Romosozumab Treatment. Calcif Tissue Int 2023:10.1007/s00223-023-01087-y. [PMID: 37138124 DOI: 10.1007/s00223-023-01087-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/21/2023] [Indexed: 05/05/2023]
Abstract
Romosozumab can increase bone mineral density (BMD) in patients with osteoporosis, but some patients do not respond to it. This study aimed to identify risk factors for being a nonresponder to romosozumab treatment. This retrospective observational study included 92 patients. Romosozumab (210 mg) was subcutaneously administered to the participants every 4 weeks over 12 months. We excluded patients who previously underwent treatment for osteoporosis to assess the impact of romosozumab alone. We evaluated the proportion of patients who did not respond to romosozumab treatment to the lumbar spine and hip with increased BMD. Nonresponders were defined as those with a bone density change of < 3% after 12 months of treatment. We compared demographics and biochemical markers between responders and nonresponders. We found that 11.5% of patients were nonresponders at the lumbar spine, and 56.8% were nonresponders at the hip. A risk factor for nonresponse at the spine was low type I procollagen N-terminal propeptide (P1NP) values at 1 month. The cutoff value for P1NP at month 1 was 50 ng/ml. We found that 11.5% and 56.8% of patients experienced no significant improvement in the lumbar spine and hip BMD, respectively. Clinicians should use nonresponse risk factors to inform decisions about romosozumab treatment for patients with osteoporosis.
Collapse
Affiliation(s)
- Ayako Tominaga
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan.
| | - Keiji Wada
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Okazaki
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | - Shuji Shimamoto
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuteru Kodama
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | | |
Collapse
|
16
|
Dai Z, Zhu R, Sheng Z, Qin G, Luo X, Qin Q, Song C, Li L, Jin P, Yang G, Cheng Y, Peng D, Zou C, Wang L, Shentu J, Zhang Q, Zhang Z, Yan X, Fang P, Yan Q, Yang L, Fan X, Liu W, Wu B, Cui R, Wu X, Xie Y, Shu C, Shen K, Wei W, Lu W, Chen H, Zhou Z. Multiple doses of SHR-1222, a sclerostin monoclonal antibody, in postmenopausal women with osteoporosis: A randomized, double-blind, placebo-controlled, dose-escalation phase 1 trial. Front Endocrinol (Lausanne) 2023; 14:1168757. [PMID: 37091850 PMCID: PMC10116854 DOI: 10.3389/fendo.2023.1168757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/22/2023] [Indexed: 04/25/2023] Open
Abstract
SHR-1222, a novel humanized monoclonal antibody targeting sclerostin, has been shown to induce bone formation and decrease bone resorption at a single dose ranging 50-400 mg in our previous phase 1 trial. This study was a randomized, double-blind, placebo-controlled, dose-escalation phase 1 trial, which further investigated the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of multiple ascending doses of SHR-1222 in women with postmenopausal osteoporosis (POP). A total of 105 women with POP were enrolled and randomly assigned. Twenty-one received placebo and eighty-four received SHR-1222 sequentially (100 mg QM, n=4; 200 or 300 mg QM, n=20; and 400 or 600 mg Q2M, n=20). The most common adverse events included increased blood parathyroid hormone, increased low-density lipoprotein, increased blood alkaline phosphatase, increased blood cholesterol, back pain, and arthralgia, the majority of which were mild in severity without noticeable safety concerns. Serum SHR-1222 exposure (Cmax,ss and AUC0-tau,ss) increased in a greater than dose-proportional manner. Following multiple doses of SHR-1222, the bone formation markers (terminal propeptide of type I procollagen, bone-specific alkaline phosphatase, and osteocalcin) increased in a dose-dependent manner, whereas the bone resorption marker (β-C-telopeptide) was downregulated. Accordingly, BMD gains in the lumbar spine, total hip, and femoral neck were observed. The maximum BMD increase from baseline at the lumbar spine was detected in the 300 mg QM cohort (14.6% vs. 0.6% in the placebo group on day 169). Six (6/83; 7.2%) subjects developed anti-SHR-1222 antibodies with no discernible effects on PKs, PDs, and safety. Thus, multiple doses of SHR-1222 showed an acceptable safety profile and dose-dependent plasma exposure in women with POP, and could improve their BMD rapidly and prominently by promoting bone formation and inhibiting bone resorption. These findings further support SHR-1222 as a potential alternative agent for the treatment of POP.
Collapse
Affiliation(s)
- Zhijie Dai
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ronghua Zhu
- Phase I Clinical Trial Center and Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhifeng Sheng
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, and Health Management Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Guijun Qin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianghang Luo
- Department of Endocrinology, Xiangya Hospital of Central South University, Changsha, China
| | - Qun Qin
- National Agency for Clinical Trial of Medicines, Xiangya Hospital of Central South University, Changsha, China
| | - Chunli Song
- Orthopedics Department, Peking University Third Hospital, Beijing, China
| | - Liping Li
- Endocrine Department, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Ping Jin
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yanxiang Cheng
- Department of Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danhong Peng
- Department of Gynaecology and Obstetrics, Zhongda Hospital Southeast University, Nanjing, China
| | - Chong Zou
- Department of Clinical Pharmacology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Lijuan Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jianzhong Shentu
- Clinical Pharmacy, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qin Zhang
- Department of Geriatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhe Zhang
- Endocrinology and Metabolism, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang Yan
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Pingfei Fang
- Phase I Clinical Trial Center and Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiangyong Yan
- Phase I Clinical Trial Center and Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lingfeng Yang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiao Fan
- Phase I Clinical Trial Center and Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Bo Wu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Rongrong Cui
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiyu Wu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuting Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chang Shu
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Kai Shen
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Wenhua Wei
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Wei Lu
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Hong Chen
- Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Zhiguang Zhou,
| |
Collapse
|
17
|
Alcorta-Sevillano N, Infante A, Macías I, Rodríguez CI. Murine Animal Models in Osteogenesis Imperfecta: The Quest for Improving the Quality of Life. Int J Mol Sci 2022; 24:ijms24010184. [PMID: 36613624 PMCID: PMC9820162 DOI: 10.3390/ijms24010184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Osteogenesis imperfecta is a rare genetic disorder characterized by bone fragility, due to alterations in the type I collagen molecule. It is a very heterogeneous disease, both genetically and phenotypically, with a high variability of clinical phenotypes, ranging from mild to severe forms, the most extreme cases being perinatal lethal. There is no curative treatment for OI, and so great efforts are being made in order to develop effective therapies. In these attempts, the in vivo preclinical studies are of paramount importance; therefore, serious analysis is required to choose the right murine OI model able to emulate as closely as possible the disease of the target OI population. In this review, we summarize the features of OI murine models that have been used for preclinical studies until today, together with recently developed new murine models. The bone parameters that are usually evaluated in order to determine the relevance of new developing therapies are exposed, and finally, current and innovative therapeutic strategies attempts considered in murine OI models, along with their mechanism of action, are reviewed. This review aims to summarize the in vivo studies developed in murine models available in the field of OI to date, in order to help the scientific community choose the most accurate OI murine model when developing new therapeutic strategies capable of improving the quality of life.
Collapse
Affiliation(s)
- Natividad Alcorta-Sevillano
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, 48940 Leioa, Spain
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
| | - Iratxe Macías
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
| | - Clara I. Rodríguez
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Plaza de Cruces S/N, 48903 Barakaldo, Spain
- Correspondence:
| |
Collapse
|
18
|
Lim SY, Bolster MB. Clinical Utility of Romosozumab in the Management of Osteoporosis: Focus on Patient Selection and Perspectives. Int J Womens Health 2022; 14:1733-1747. [PMID: 36544862 PMCID: PMC9762257 DOI: 10.2147/ijwh.s315184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/05/2022] [Indexed: 12/23/2022] Open
Abstract
As one of the most potent osteoanabolic agents with a unique mechanism of action, romosozumab has high efficacy for osteoporosis treatment. It is a monoclonal antibody against sclerostin, a natural inhibitor of the Wnt signaling pathway, and by inhibiting sclerostin, activation of Wnt signaling occurs with a cascade of changes ultimately leading to bone mineral density (BMD) gains. Romosozumab stimulates bone modeling and has a dual effect of activating bone formation while inhibiting bone resorption. With this unique mechanism of action, treatment with romosozumab leads to a rapid and significant gain in BMD; these gains are higher than seen with bisphosphonates, denosumab, or parathyroid hormone (PTH) analogs. The FRAME and ARCH studies represent two pivotal trials demonstrating the efficacy of romosozumab in treating osteoporosis. Treatment with romosozumab should be followed by an antiresorptive agent, as this approach has demonstrated maintenance of or greater increases in BMD and reduced fracture risk even after finishing romosozumab treatment. As an osteoanabolic agent, romosozumab has shown superiority to alendronate in reducing fracture risk, increasing bone density, and potentially more rapid fracture risk reduction. Recent data have suggested that romosozumab prior to antiresorptive therapy may be the ideal treatment sequence, especially in high-risk patients and patients at imminent risk of fracture. Carrying a black box warning, romosozumab should be avoided in patients who have had myocardial infarction or stroke in the past year. Further studies are needed to clarify the increased cardiovascular risk attributed to this drug. Romosozumab has expanded our osteoporosis armamentarium and has enabled novel approaches, including "treat to target." Future studies are needed to evaluate the optimal use sequence and to assess its safety, especially in patients with cardiovascular risk factors.
Collapse
Affiliation(s)
- Sian Yik Lim
- Hawaii Pacific Health Medical Group, Honolulu, HI, USA,Department of Family Medicine, John E Burns School of Medicine, University of Hawaii, Honolulu, HI, USA,Correspondence: Sian Yik Lim, Bone and Joint Center, Straub Clinic, 800 S. King Street, Honolulu, HI, 96813, USA, Tel +1 808-522-4232, Fax +1 808-522-4401, Email
| | - Marcy B Bolster
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Associate Professor of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Langdahl B, Hofbauer LC, Ferrari S, Wang Z, Fahrleitner-Pammer A, Gielen E, Lakatos P, Czerwinski E, Gimeno EJ, Timoshanko J, Oates M, Libanati C. Romosozumab efficacy and safety in European patients enrolled in the FRAME trial. Osteoporos Int 2022; 33:2527-2536. [PMID: 36173415 PMCID: PMC9652294 DOI: 10.1007/s00198-022-06544-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022]
Abstract
UNLABELLED In this post hoc analysis, we assessed romosozumab efficacy and safety in European patients enrolled in FRAME. Romosozumab treatment through 12 months, followed by denosumab for a further 24 months, resulted in early and sustained risk reduction for major fracture categories, associated with large gains in bone mineral density. INTRODUCTION In the multinational FRAME phase 3 trial of romosozumab in postmenopausal women with osteoporosis, marked differences between clinical and non-vertebral fracture outcomes were observed among patients from Central and Southern America versus rest of world. This post hoc analysis assessed romosozumab efficacy and safety in European patients enrolled in the FRAME trial and extension study. METHODS In FRAME (NCT01575834), patients were randomised 1:1 to romosozumab 210 mg or placebo monthly (QM) for 12 months, followed by open-label denosumab 60 mg Q6M to month 36, including a 12-month extension study. We report incidence of major fracture outcomes, bone mineral density (BMD) change from baseline and safety for European patients enrolled in FRAME. RESULTS In FRAME, 3013/7180 (41.96%) patients were European; 1494 received romosozumab and 1519 received placebo. Through 12 months, romosozumab reduced fracture risk versus placebo for non-vertebral fracture (1.4% versus 3.0%; p = 0.004), clinical fracture (1.4% versus 3.6%; p < 0.001), new vertebral fracture (0.4% versus 2.1%; p < 0.001) and major osteoporotic fracture (0.9% versus 2.8%; p < 0.001), with results sustained through 36 months following transition to denosumab. Hip fractures were numerically reduced with romosozumab at month 12 (0.2% versus 0.6%; p = 0.092). Romosozumab increased BMD versus placebo at month 12; all patients in the romosozumab and placebo groups experienced further increases by month 36 after transition to denosumab. Adverse events were balanced between groups. CONCLUSIONS Among European patients in FRAME, romosozumab resulted in early and sustained risk reduction for all major fracture categories, associated with large BMD gains that continued after transition to denosumab.
Collapse
Affiliation(s)
- Bente Langdahl
- Department of Endocrinology, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Clouse G, Penman S, Hadjiargyrou M, Komatsu DE, Thanos PK. Examining the role of cannabinoids on osteoporosis: a review. Arch Osteoporos 2022; 17:146. [PMID: 36401719 DOI: 10.1007/s11657-022-01190-x] [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] [Received: 08/02/2022] [Accepted: 11/11/2022] [Indexed: 11/20/2022]
Abstract
PURPOSE Prior research studies have shown that the endocannabinoid system, influenced by CBD and THC, plays a role in bone remodeling. As both the research on cannabis and use of cannabis continue to grow, novel medicinal uses of both its constituents as well as the whole plant are being discovered. This review examines the role of cannabinoids on osteoporosis, more specifically, the endocannabinoid system and its role in bone remodeling and the involvement of the cannabinoid receptors 1 and 2 in bone health, as well as the effects of Δ9-tetrahydrocannabinol (THC), cannabidiol (CBD), and synthetic cannabinoids on bone. METHODS A comprehensive literature search of online databases including PUBMED was utilized. RESULTS A total of 29 studies investigating the effects of cannabis and/or its constituents as well as the activation or inactivation of cannabinoid receptors 1 and 2 were included and discussed. CONCLUSION While many of the mechanisms are still not yet fully understood, both preclinical and clinical studies show that the effects of cannabis mediated through the endocannabinoid system may prove to be an effective treatment option for individuals with osteoporosis.
Collapse
Affiliation(s)
- Grace Clouse
- Behavioral Neuropharmacology and Neuroimaging Laboratory On Addictions (BNNLA), Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Samantha Penman
- Behavioral Neuropharmacology and Neuroimaging Laboratory On Addictions (BNNLA), Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David E Komatsu
- Department of Orthopedics, Stony Brook University, Stony Brook, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory On Addictions (BNNLA), Research Institute On Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA. .,Department of Psychology, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
21
|
Chen H, Zhang Y, Yu T, Song G, Xu T, Xin T, Lin Y, Han B. Nano-Based Drug Delivery Systems for Periodontal Tissue Regeneration. Pharmaceutics 2022; 14:2250. [PMID: 36297683 PMCID: PMC9612159 DOI: 10.3390/pharmaceutics14102250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/15/2022] Open
Abstract
Periodontitis is a dysbiotic biofilm-induced and host-mediated inflammatory disease of tooth supporting tissues that leads to progressive destruction of periodontal ligament and alveolar bone, thereby resulting in gingival recession, deep periodontal pockets, tooth mobility and exfoliation, and aesthetically and functionally compromised dentition. Due to the improved biopharmaceutical and pharmacokinetic properties and targeted and controlled drug release, nano-based drug delivery systems have emerged as a promising strategy for the treatment of periodontal defects, allowing for increased efficacy and safety in controlling local inflammation, establishing a regenerative microenvironment, and regaining bone and attachments. This review provides an overview of nano-based drug delivery systems and illustrates their practical applications, future prospects, and limitations in the field of periodontal tissue regeneration.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yunfan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tingting Yu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Guangying Song
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianmin Xu
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Tianyi Xin
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| | - Yifan Lin
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Bing Han
- Department of Orthodontics, School and Hospital of Stomatology, Peking University, Beijing 100081, China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing 100081, China
| |
Collapse
|
22
|
Zhou H, Jiang J, Chen X, Zhang Z. Differentially expressed genes and miRNAs in female osteoporosis patients. Medicine (Baltimore) 2022; 101:e29856. [PMID: 35839011 PMCID: PMC11132388 DOI: 10.1097/md.0000000000029856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 06/07/2022] [Indexed: 11/26/2022] Open
Abstract
Osteoporosis is characterized by lowing bone mineral density. This study aimed to investigate the genes, miRNAs, pathways, and miRNA-gene interaction pairs involved in the pathogenesis of female osteoporosis. The differentially expressed genes (DEGs, GSE62402), differentially expressed miRNAs (DEmiRNAs, GSE63446), and differentially methylated genes (GSE62588) between females with low- and high-hip bone mineral density were identified. Genes common to DEGs, differentially methylated genes, DEmiRNAs' targets, and osteoporosis-related genes were retained and used to construct the miRNA-mRNA-pathway regulatory network. The expression of hub nodes was validated in microarray datasets (genes in GSE56116 and miRNAs in GSE93883). Thirty-four DEmiRNAs and 179 DEGs with opposite expression-methylation profiles were identified. Functional enrichment analysis showed that DEGs were associated with pathways including "hsa00380:Tryptophan metabolism," "hsa04670:Leukocyte transendothelial migration," "hsa04630:Jak-STAT signaling pathway," and "hsa04062:Chemokine signaling pathway." The miRNA-mRNA-pathway network included 10 DEGs, 9 miRNAs, and 4 osteoporosis-related pathways. The miRNA-gene-pathway axes including hsa-miR-27b-5p/3p-IFNAR1-hsa04630, hsa-miR-30a-5p/3p-IFNAR1-hsa04630, hsa-miR-30a-5p/3p-ALDH2-hsa00380, and hsa-miR-194-5p/3p-NCF2-hsa04670 were included in the network. Microarray validation showed that IFNAR1, NCF2, and ALDH2 were upregulated, and hsa-miR-30a-3p/5p, hsa-miR-194-3p/5p, hsa-miR-27b-3p/5p, and hsa-miR-34a-3p were downregulated in osteoporotic samples compared with control. Axes including hsa-miR-27b/30a-IFNAR1-Jak-STAT signaling pathway, hsa-miR-30a-ALDH2-Tryptophan metabolism, and hsa-miR-194-NCF2-Leukocyte transendothelial migration were involved in osteoporosis pathogenesis.
Collapse
Affiliation(s)
- Hailong Zhou
- Department of Integrated Traditional Chinese and Western Medicine, the First People’s Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang Province, PR China
| | - Jianmin Jiang
- Department of Internal Medicine, the First People’s Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang Province, PR China
| | - Xiaohua Chen
- Department of Orthopaedics, the First People’s Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang Province, PR China
| | - Zhiwei Zhang
- Department of Orthopaedics, the First People’s Hospital of Fuyang Hangzhou, Hangzhou, Zhejiang Province, PR China
| |
Collapse
|
23
|
Abstract
Sclerostin is most recognized for its role in controlling bone formation but is also expressed in the heart, aorta, coronary, and peripheral arteries. This review summarizes research on sclerostin's role in cardiovascular disease. Rodent studies have found sclerostin to be expressed at sites of arterial calcification. In contrast, aortic sclerostin was reported to be downregulated in a mouse model of abdominal aortic aneurysm, and transgenic upregulation or administration of sclerostin was found to prevent abdominal aortic aneurysm and atherosclerosis formation. Sclerostin deficiency was reported to stimulate cardiac rupture in one rodent model. In humans, 7 of 11 studies reported a significant association between high serum sclerostin and high carotid intima media thickness. Ten of 15 studies reported a significant association between high serum sclerostin and severe arterial calcification. Twelve of 14 studies reported a significant association between high serum sclerostin and high arterial stiffness or atherosclerosis severity. Four of 9 studies reported a significant association between high serum sclerostin and high risk of cardiovascular events. A meta-analysis of randomized controlled trials suggested that administration of the sclerostin blocking antibody romosozumab did not significantly increase the risk of major adverse cardiovascular events (risk ratio, 1.14 [95% CI, 0.83-1.57]; P=0.54) or cardiovascular death (risk ratio, 0.92 [95% CI, 0.53-1.59]; P=0.71). Human genetic studies reported variants predisposing to low arterial sclerostin expression were associated with a high risk of cardiovascular events. Overall, past research suggests a cardiovascular protective role of sclerostin but findings have been inconsistent, possibly due to variations in study design, the unique populations and models studied, and the heterogeneous methods used.
Collapse
Affiliation(s)
- Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.G., S.T.), James Cook University, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine J.G.' S.T.), James Cook University, Townsville, Queensland, Australia
| | - Shivshankar Thanigaimani
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry (J.G., S.T.), James Cook University, Townsville, Queensland, Australia.,The Australian Institute of Tropical Health and Medicine J.G.' S.T.), James Cook University, Townsville, Queensland, Australia.,The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
24
|
Poutoglidou F, Samoladas E, Raikos N, Kouvelas D. Efficacy and safety of anti-sclerostin antibodies in the treatment of osteoporosis: A meta-analysis and systematic review. J Clin Densitom 2022; 25:401-415. [PMID: 34920938 DOI: 10.1016/j.jocd.2021.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 11/19/2022]
Abstract
Osteoporosis is a chronic disease with an increasing prevalence. Anti-sclerostin antibodies are being investigated for the treatment of osteoporosis. The aim of this systematic review and meta-analysis is to evaluate the efficacy and safety of antis-sclerostin antibodies compared to placebo and conventional therapies (alendronate and teriparatide) in the treatment of osteoporosis. Randomized controlled trials were searched from PubMed, EMBASE and Cochrane Central Register of Controlled Trails (CENTRAL) from their inception up to June 2021 by using Medical Subject Headings terms "anti-sclerostin antibody", "romosozumab", "blosozumab", "AMG 785″, "LY2541546", and "osteoporosis". Two investigators independently screened eligible studies, assessed the risk of bias and extracted the data from each study. The I2 index was used to assess heterogeneity. Meta-analysis was conducted using the Review Manager Software (RevMan, Version 5.4). The GRADE approach was used to rate the quality of evidence for all the pooled outcomes. 8 RCTs with 12,416 patients met the inclusion criteria. Anti-sclerostin antibodies significantly increased lumbar spine, total hip and femoral neck bone mineral density compared to placebo, alendronate and teriparatide at both 6 and 12 mo. Adverse events were comparable between anti-sclerostin antibodies and other treatments, except for the incidence of injection-site reactions that was higher in the anti-sclerostin antibody groups. Anti-sclerostin antibodies represent a valid theurapeutic option in the treatment of osteoporosis. Further studies with longer duration and follow-up are needed to confirm the results of this meta-analysis.
Collapse
Affiliation(s)
- Frideriki Poutoglidou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Efthimios Samoladas
- Orthopeadics Division of Genimatas Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Raikos
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
25
|
Lu J, Hu D, Ma C, Shuai B. Advances in Our Understanding of the Mechanism of Action of Drugs (including Traditional Chinese Medicines) for the Intervention and Treatment of Osteoporosis. Front Pharmacol 2022; 13:938447. [PMID: 35774616 PMCID: PMC9237325 DOI: 10.3389/fphar.2022.938447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis (OP) is known as a silent disease in which the loss of bone mass and bone density does not cause obvious symptoms, resulting in insufficient treatment and preventive measures. The losses of bone mass and bone density become more severe over time and an only small percentage of patients are diagnosed when OP-related fractures occur. The high disability and mortality rates of OP-related fractures cause great psychological and physical damage and impose a heavy economic burden on individuals and society. Therefore, early intervention and treatment must be emphasized to achieve the overall goal of reducing the fracture risk. Anti-OP drugs are currently divided into three classes: antiresorptive agents, anabolic agents, and drugs with other mechanisms. In this review, research progress related to common anti-OP drugs in these three classes as well as targeted therapies is summarized to help researchers and clinicians understand their mechanisms of action and to promote pharmacological research and novel drug development.
Collapse
|
26
|
Mochizuki T, Yano K, Ikari K, Hiroshima R, Okazaki K. Comparison of Romosozumab Versus Denosumab Treatment on Bone Mineral Density After One Year in Rheumatoid Arthritis Patients with Severe Osteoporosis: A Randomized Clinical Pilot Study. Mod Rheumatol 2022; 33:490-495. [PMID: 35689558 DOI: 10.1093/mr/roac059] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVES To investigate the effect of romosozumab versus denosumab treatment on bone mineral density (BMD), disease activity, and joint damage in patients with rheumatoid arthritis (RA) and severe osteoporosis. METHODS Fifty-one postmenopausal women were enrolled and randomized equally into two groups to receive either romosozumab or the denosumab. Changes (Δ) in the BMD (at lumbar spine, total hip, and femoral neck), disease activity score in 28 joints (DAS28)-erythrocyte sedimentation rate (ESR), and van der Heijde-modified Total Sharp Score (TSS) from baseline to 12 months after treatment were evaluated. RESULTS The ΔBMD at 12 months in the romosozumab and denosumab groups were 10.2 ± 5.6% and 5.0 ± 3.1% (p = 0.002) for the lumbar spine, 3.7 ± 4.9% and 3.5 ± 3.0% (p = 0.902) for total hip, and 3.6 ± 4.7% and 3.2 ± 4.9% (p = 0.817) for femoral neck, respectively. The ΔDAS28-ESR at 12 months in the romosozumab and denosumab groups was 0.14 and 0.22 (p = 0.643), respectively, whereas, the ΔTSS at 12 months was 0.33 and 0.29 (p = 0.927), respectively. CONCLUSIONS Our results suggest that romosozumab treatment was more effective in increasing the BMD at the lumbar spine than denosumab, and may be selected for patients who require a significant increase in the lumbar spine BMD. Moreover, romosozumab may be not affect disease activity and joint damage in patients with RA.
Collapse
Affiliation(s)
- Takeshi Mochizuki
- Department of Orthopaedic Surgery, Kamagaya General Hospital, Chiba, Japan
| | - Koichiro Yano
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan.,Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University, Tokyo, Japan
| | - Ryo Hiroshima
- Department of Orthopaedic Surgery, Kamagaya General Hospital, Chiba, Japan
| | - Ken Okazaki
- Department of Orthopaedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
27
|
Inose H, Ariga A, Motoyoshi T, Fukushima K, Tomizawa S, Kato T, Takahashi K, Yoshii T, Okawa A. The real‐world effect of 12 months of romosozumab treatment on patients with osteoporosis with a high risk of fracture and factors predicting the rate of bone mass increase: A multicenter retrospective study. JBMR Plus 2022; 6:e10637. [PMID: 35866147 PMCID: PMC9289984 DOI: 10.1002/jbm4.10637] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 11/25/2022] Open
Abstract
Excluding clinical trials, there is limited evidence on the effect of 12 months of romosozumab treatment on bone mineral density (BMD) increase in real‐world clinical practice because its use has only been approved recently. Thus, this study aimed to investigate the real‐world effect of 12 months of romosozumab treatment on BMD increase and identify factors that predict the rate of BMD increase after 12 months of romosozumab treatment. We retrospectively investigated 106 patients who completed a 12‐month romosozumab treatment for osteoporosis with a high risk of fractures at four hospitals from March 2020 to March 2022. The univariate and multiple regression analyses were performed to analyze the concurrent effects of various factors on the BMD increase after the 12‐month romosozumab treatment. After 1 year of treatment, the lumbar spine BMD increased by 14.6%, and femoral neck BMD increased by 5.1%. Univariate regression analysis found that male sex, high tartrate‐resistant acid phosphatase 5b (TRACP‐5b) value before romosozumab administration, absence of osteoporosis medications before romosozumab administration, and low baseline lumbar spine BMD were associated with the extent of lumbar spine BMD increase. Moreover, stepwise multiple regression analysis found that the TRACP‐5b value before romosozumab administration was a significant predictor of the rate of lumbar spine BMD increase after 1 year of romosozumab administration. In conclusion, our results demonstrated the effectiveness of the 12‐month romosozumab treatment for osteoporosis with a high risk of fractures and the TRACP‐5b value before romosozumab administration was a significant predictor of the rate of lumbar spine BMD increase after 1 year of romosozumab administration. Our findings could help establish more efficient treatment strategies for patients with osteoporosis at a high risk of fracture. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Hiroyuki Inose
- Department of Orthopedic and Trauma Research Tokyo Medical and Dental University, 1‐5‐45 Yushima, Bunkyo‐ku Tokyo Japan
| | - Akane Ariga
- Department of Orthopedics Saku Central Hospital Advanced Care Center, 3400‐28, Nakagomi, Saku‐city Nagano Japan
| | - Takayuki Motoyoshi
- Department of Orthopedics Saku Central Hospital Advanced Care Center, 3400‐28, Nakagomi, Saku‐city Nagano Japan
| | - Kazuyuki Fukushima
- Department of Orthopedics Saku Central Hospital Advanced Care Center, 3400‐28, Nakagomi, Saku‐city Nagano Japan
| | - Shoji Tomizawa
- Department of Orthopedics Tokyobay UrayasuIchikawa Medical Center, 3‐4‐32 Todaijima, Urayasushi Chiba Japan
| | - Tsuyoshi Kato
- Department of Orthopaedics Ome Municipal General Hospital, 4‐16‐5 Higashiome, Ome‐shi Tokyo Japan
| | - Kunihiko Takahashi
- Department of Biostatistics, M&D Data Science Center Tokyo Medical and Dental University, 1‐5‐45 Yushima, Bunkyo‐ku Tokyo Japan
| | - Toshitaka Yoshii
- Department of Orthopaedics Graduate School, Tokyo Medical and Dental University, 1‐5‐45 Yushima, Bunkyo‐ku Tokyo Japan
| | - Atsushi Okawa
- Department of Orthopaedics Graduate School, Tokyo Medical and Dental University, 1‐5‐45 Yushima, Bunkyo‐ku Tokyo Japan
| |
Collapse
|
28
|
Appelman-Dijkstra NM, Oei HLDW, Vlug AG, Winter EM. The effect of osteoporosis treatment on bone mass. Best Pract Res Clin Endocrinol Metab 2022; 36:101623. [PMID: 35219602 DOI: 10.1016/j.beem.2022.101623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Over the last two decades there have been significant developments in the pharmacotherapy of osteoporosis. The therapeutic arsenal has expanded with monoclonal antibodies which have been developed based on discoveries of the molecular mechanisms underlying bone resorption and bone formation. Denosumab, the antibody binding RANKL, inhibits bone resorption, and romosozumab, the antibody binding sclerostin, inhibits bone resorption and stimulates bone formation as well. Both antibodies have shown potent anti-fracture efficacy in randomized clinical trials and this review will discuss the preclinical and clinical studies focusing on the effects on bone mass. After discontinuation of these antibodies, bone mineral density quickly returns to baseline and in the case of denosumab, discontinuation can not only induce rebound bone loss, but also the occurrence of vertebral fractures. Therefore, sequential antiresorptive therapy to maintain bone mass gains and anti-fracture efficacy is of utmost importance and will also be discussed in this review.
Collapse
Affiliation(s)
- Natasha M Appelman-Dijkstra
- Department of Internal Medicine; Division Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands.
| | - H Ling D W Oei
- Department of Internal Medicine; Division Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands; Department of Internal Medicine, Jan van Goyen Medical Center, Amsterdam, the Netherlands.
| | - Annegreet G Vlug
- Department of Internal Medicine; Division Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands; Department of Internal Medicine, Jan van Goyen Medical Center, Amsterdam, the Netherlands.
| | - Elizabeth M Winter
- Department of Internal Medicine; Division Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
29
|
Singh S, Dutta S, Khasbage S, Kumar T, Sachin J, Sharma J, Varthya SB. A systematic review and meta-analysis of efficacy and safety of Romosozumab in postmenopausal osteoporosis. Osteoporos Int 2022; 33:1-12. [PMID: 34432115 PMCID: PMC9003152 DOI: 10.1007/s00198-021-06095-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/08/2021] [Indexed: 11/29/2022]
Abstract
The study was conducted to illustrate the effect of Romosozumab in postmenopausal osteoporosis patients. Romosozumab decreased the incidence of vertebral, nonvertebral, and clinical fractures significantly. In addition, decreased incidence of falls and increased bone mineral density at lumbar spine, total hip, and femoral neck was observed. Romosozumab is a monoclonal antibody that acts against the sclerostin pathway leading to enhanced bone formation and reduced bone resorption in patients with osteoporosis. Electronic search was performed on Medline (via PubMed), The Cochrane Central Register of Controlled Trials, and clinicaltrials.gov, till May 2020, for RCTs evaluating the effectiveness of Romosozumab in postmenopausal osteoporosis. RCTs evaluating the effect of Romosozumab on fractures and bone mineral density in postmenopausal osteoporosis patients. Meta-analysis was performed by Cochrane review manager 5 (RevMan) version 5.3. Cochrane risk of bias 2.0 tool and GRADE pro-GDT were applied for methodological quality and overall evidence quality, respectively. One hundred seventy-nine studies were screened, and 10 eligible studies were included in the analysis, with a total of 6137 patients in romosozumab group and 5732 patients in control group. Romosozumab significantly reduced the incidence of vertebral fractures [OR = 0.43 (95%CI = 0.35-0.52), High-quality evidence], nonvertebral fractures [OR = 0.78 (95%CI = 0.66-0.92), High quality], and clinical fractures [OR = 0.70 (95%CI = 0.60-0.82), High quality] at 24 months. Significant reduction in incidence risk of falls [OR = 0.87 (95%CI = 0.78-0.96), High quality] was observed with romosozumab. Bone mineral density was significantly increased in the romosozumab treated groups at lumbar spine [MD = 12.66 (95%CI = 12.66-12.67), High quality], total hip [MD = 5.69 (95%CI = 5.68 - 5.69), Moderate quality], and femoral neck [MD = 5.18 (95%CI = 5.18-5.19), Moderate quality] at 12 months. The total adverse events [RR = 0.98(95%CI = 0.96-1.01), Moderate quality] and serious adverse events [RR = 0.98(95%CI = 0.88-1.08), Moderate quality] with romosozumab were comparable to the control group. The current analysis with evidence on efficacy and safety of Romosozumab, authors opine to recommend the use of Romosozumab treatment for post-menopausal osteoporosis.Systematic review registration: PROSPERO registration number: CRD42019112196.
Collapse
Affiliation(s)
- S Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India
| | - S Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India.
| | - S Khasbage
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, India
| | - T Kumar
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India
| | - J Sachin
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India
| | - J Sharma
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India
| | - S B Varthya
- Department of Pharmacology, All India Institute of Medical Sciences, Rajasthan, 342005, Jodhpur, India
| |
Collapse
|
30
|
Miller SA, St Onge EL, Whalen KL. Romosozumab: A Novel Agent in the Treatment for Postmenopausal Osteoporosis. J Pharm Technol 2021; 37:45-52. [PMID: 34752536 DOI: 10.1177/8755122520967632] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective: To review the safety and efficacy of romosozumab (Evenity) in the treatment of osteoporosis in women. Data Sources: An English-language search of PubMed and Medline (1966 to August 2020) was conducted using the keywords romosozumab, sclerostin inhibitor, AMG785, and osteoporosis. Manufacturer prescribing information, abstracts, fda.gov, and ClinicalTrials.gov data were incorporated for additional materials. In addition, a review of bibliographies of retrieved articles was performed to identify additional references. Study Selection/Data Extraction: Articles selected included those that described clinical studies of pharmacokinetics, efficacy, or safety of romosozumab. Data Synthesis: Romosozumab is a human monoclonal antibody that inhibits the action of sclerostin and is the first agent in its class to reach Phase III trials. Significant increases in bone mineral density and decreases in vertebral and hip fractures are demonstrated in Phase III trials. Favorable results led to its marketing approval in several countries. Major adverse cardiac events were observed in one clinical trial. Other adverse effects include arthralgia, headache, and injection site reactions. Place in Therapy: Romosozumab is the first agent to inhibit bone resorption and stimulate bone formation. Romosozumab should be reserved for postmenopausal women at highest risk for fracture and should be followed by an anti-resportive agent to maintain or further increase bone mineral density. This injectable agent should not be considered for women with a history of or at high risk of cardiovascular disease.
Collapse
|
31
|
The Effects of Osteoporotic and Non-osteoporotic Medications on Fracture Risk and Bone Mineral Density. Drugs 2021; 81:1831-1858. [PMID: 34724173 PMCID: PMC8578161 DOI: 10.1007/s40265-021-01625-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/26/2022]
Abstract
Osteoporosis is a highly prevalent bone disease affecting more than 37.5 million individuals in the European Union (EU) and the United States of America (USA). It is characterized by low bone mineral density (BMD), impaired bone quality, and loss of structural and biomechanical properties, resulting in reduced bone strength. An increase in morbidity and mortality is seen in patients with osteoporosis, caused by the approximately 3.5 million new osteoporotic fractures occurring every year in the EU. Currently, different medications are available for the treatment of osteoporosis, including anti-resorptive and osteoanabolic medications. Bisphosphonates, which belong to the anti-resorptive medications, are the standard treatment for osteoporosis based on their positive effects on bone, long-term experience, and low costs. However, not only medications used for the treatment of osteoporosis can affect bone: several other medications are suggested to have an effect on bone as well, especially on fracture risk and BMD. Knowledge about the positive and negative effects of different medications on both fracture risk and BMD is important, as it can contribute to an improvement in osteoporosis prevention and treatment in general, and, even more importantly, to the individual's health. In this review, we therefore discuss the effects of both osteoporotic and non-osteoporotic medications on fracture risk and BMD. In addition, we discuss the underlying mechanisms of action.
Collapse
|
32
|
Mochizuki T, Yano K, Ikari K, Okazaki K. Effects of romosozumab or denosumab treatment on the bone mineral density and disease activity for 6 months in patients with rheumatoid arthritis with severe osteoporosis: An open-label, randomized, pilot study. Osteoporos Sarcopenia 2021; 7:110-114. [PMID: 34632114 PMCID: PMC8486617 DOI: 10.1016/j.afos.2021.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Accepted: 08/15/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives To investigate effects of romosozumab treatment on disease activity and bone mineral density (BMD) in patients with rheumatoid arthritis (RA) and severe osteoporosis in comparison with effects of denosumab treatment. Methods A total of 50 women were enrolled in this study. The subjects were randomized equally into 2 groups: the romosozumab group or the denosumab group. Disease activity score in 28 joints (DAS28)-erythrocyte sedimentation rate (ESR) and BMD at lumbar spine were evaluated. Results The percent changes (Δ) in the BMD values at 3 and 6 months for the lumbar spine were as follows: romosozumab; 4.9% and 5.2%, denosumab: 2.3% and 3.2%. The ΔBMD for the lumbar spine at 3 months was significantly higher in the romosozumab group than in the denosumab group (P = 0.044). The DAS28-ESR at baseline, 3 and 6 months in the romosozumab group were 2.88, 2.60 (P = 0.427) and 2.58 (P = 0.588), respectively. The change from baseline in DAS28-ESR did not differ significantly between these 2 groups at any time point. Conclusions The present study revealed that romosozumab treatment is more effective than denosumab treatment in increasing BMD of the lumbar spine at 3 months. Furthermore, the present study suggested that romosozumab treatment has no effects on the disease activity of RA in patients with RA and severe osteoporosis for 6 months.
Collapse
Affiliation(s)
- Takeshi Mochizuki
- Department of Orthopedic Surgery, Kamagaya General Hospital, Chiba, Japan
- Corresponding author. Department of Orthopedic Surgery, Kamagaya General Hospital, 929-6 Hatsutomi, Kamagaya, Chiba, 273-0121, Japan.
| | - Koichiro Yano
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsunori Ikari
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
- Division of Multidisciplinary Management of Rheumatic Diseases, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Okazaki
- Department of Orthopedic Surgery, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
33
|
Shimizu T, Arita K, Murota E, Hiratsuka S, Fujita R, Ishizu H, Asano T, Takahashi D, Takahata M, Iwasaki N. Effects after starting or switching from bisphosphonate to romosozumab or denosumab in Japanese postmenopausal patients. J Bone Miner Metab 2021; 39:868-875. [PMID: 33847831 PMCID: PMC8042469 DOI: 10.1007/s00774-021-01226-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/23/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE We aimed to investigate the longitudinal changes in bone metabolic markers and bone mineral density (BMD) after starting or switching from bisphosphonate (BP) to romosozumab (ROMO) or denosumab (DENO) therapies over 12 months and to determine predictors that establish associations with changes in BMD among the patients received the ROMO therapy. METHODS Postmenopausal osteoporosis patients with a high risk of fracture-154 in total-were recruited; their therapies were switched to ROMO or DENO from BP/naïve or vitamin D (ND) (ND-ROMO: 43, BP-ROMO: 38, ND-DENO: 38, and BP-DENO: 35). Longitudinal changes in bone metabolic markers and BMD were evaluated. RESULTS ROMO groups showed significant increases in BMD of the lumbar spine at 6 and 12 months and femoral neck at 12 months compared to the DENO groups. Although BP-ROMO showed significant increase in the lumbar spine BMD compared to BP-DENO, there were no significant differences in femoral neck and total hip BMDs between BP-ROMO and BP-DENO. Among the ROMO groups, % changes of BMD from baseline to 12 months were associated with bone metabolic markers at baseline and changes in TRACP-5b from baseline to 3 months. CONCLUSIONS ROMO continuously increased BMD for 12 months and performed better than DENO. On the other hand, effects of ROMO switched from BP on BMD of femoral neck and total hip were almost same with DENO. Bone metabolic markers at baseline and changes in TRACP-5b from baseline to 3 months may predict the efficacy of ROMO after 12 months of administration.
Collapse
Affiliation(s)
- Tomohiro Shimizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Kosuke Arita
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Eihiro Murota
- Department of Orthopaedic Surgery, Wajyokai Sapporo Hospital, Sapporo, Japan
| | - Shigeto Hiratsuka
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Orthopaedic Surgery, Wajyokai Sapporo Hospital, Sapporo, Japan
| | - Ryo Fujita
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
- Department of Orthopaedic Surgery, Hokkaido Orthopaedic Memorial Hospital, Sapporo, Japan
| | - Hotaka Ishizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Tsuyoshi Asano
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Daisuke Takahashi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Masahiko Takahata
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| |
Collapse
|
34
|
Ishizu H, Arita K, Terkawi MA, Shimizu T, Iwasaki N. Risks vs. benefits of switching therapy in patients with postmenopausal osteoporosis. Expert Rev Endocrinol Metab 2021; 16:217-228. [PMID: 34310233 DOI: 10.1080/17446651.2021.1956902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Introduction: Osteoporosis is characterized by the fragility of bones, leading to fractures and, consequently, the deterioration of functional capacity and quality of life. Postmenopausal women, in particular, are prone to osteoporosis and often require anti-osteoporosis treatment. In the last few decades, various anti-osteoporosis drugs have been approved for clinical use. In an aging society, osteoporosis cannot be treated using a single agent; therefore, switching therapy is an important treatment strategy.Areas covered: This review covers switching therapy in patients with postmenopausal osteoporosis. It's extremely important to understand the characteristics of each drug including; limitations on the duration of use, side effects due to long-term use (such as atypical femur fracture and osteonecrosis of the jaw) or discontinuation (such as rebound phenomenon), compliance, and ability to prevent fractures. We review and summarize the risks and benefits of switching therapy.Expert opinion: When switching therapy, the order of drug administration is important. Routine monitoring should be continued after switching treatments. We recommend first using osteoanabolic agents in postmenopausal women with severe osteoporosis. In addition, identifying predictors of the efficacy and side effects of treatment may help prevent the inappropriate use of drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Hotaka Ishizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Kosuke Arita
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Tomohiro Shimizu
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo, Japan
| |
Collapse
|
35
|
Ginsberg C, Ix JH. Diagnosis and Management of Osteoporosis in Advanced Kidney Disease: A Review. Am J Kidney Dis 2021; 79:427-436. [PMID: 34419519 DOI: 10.1053/j.ajkd.2021.06.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Osteoporosis and fractures are common in persons with advanced chronic kidney disease (CKD) and on maintenance dialysis. Although the diagnosis of osteoporosis in this population can be difficult, imaging, especially with dual-energy x-ray absorptiometry (DXA), is helpful in identifying persons with CKD at the highest risk of fracture. Although blood biomarkers including parathyroid hormone and bone-specific alkaline phosphatase concentrations can aid in assessing bone turnover state, bone biopsy remains the gold standard in determining bone turnover in persons with advanced kidney disease and osteoporosis. With the increasing armamentarium of osteoporosis drugs, it now may be possible to prevent many fractures in advanced CKD. Unfortunately, data on these drugs are limited in persons with advanced CKD. Clinicians, aided by advances in imaging, biomarkers, and bone biopsy can now use these novel agents to target bone turnover abnormalities such as adynamic bone disease and high bone turnover disease. This review will discuss the most recent literature surrounding the diagnosis, management, and monitoring of osteoporosis and fractures in persons with advanced CKD or on maintenance dialysis.
Collapse
Affiliation(s)
- Charles Ginsberg
- Division of Nephrology-Hypertension, Department of Medicine, University of California-San Diego, San Diego, California.
| | - Joachim H Ix
- Division of Nephrology-Hypertension, Department of Medicine, University of California-San Diego, San Diego, California; Nephrology Section, Veterans Affairs San Diego Healthcare System (JHI), San Diego, California
| |
Collapse
|
36
|
Prather C, Adams E, Zentgraf W. Romosozumab: A first-in-class sclerostin inhibitor for osteoporosis. Am J Health Syst Pharm 2021; 77:1949-1956. [PMID: 32880646 DOI: 10.1093/ajhp/zxaa285] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE The purpose of this article is to review the pharmacology, efficacy, and safety of the sclerostin inhibitor romosozumab for the treatment of osteoporosis, including data from clinical trials of the drug. SUMMARY A review of the literature was performed by searching PubMed and MEDLINE for all relevant articles published between January 2014 and February 2020 using the keywords romosozumab, romosozumab-aqqg, osteoporosis, and fracture. All relevant English-language articles evaluating the pharmacology, efficacy, or safety of romosozumab for the treatment of osteoporosis in humans were included; poster presentations were excluded. Romosozumab has been approved by the Food and Drug Administration and is considered both safe and effective for the treatment of osteoporosis in high-risk postmenopausal females. Phase 2 and phase 3 clinical trials have shown a statistically significant decrease in new vertebral fractures and an increase in bone mineral density with romosozumab use, as compared with both placebo use and use of alternative osteoporosis therapies. The primary safety concern is a potential risk of cardiovascular events; additionally, hypocalcemia must be corrected prior to initiation. Romosozumab is the first anabolic medication that both increases bone formation and decreases bone resorption. Data suggest that romosozumab is more effective than oral bisphosphonates in preventing osteoporotic fractures, though cost and safety concerns must be considered. CONCLUSION Romosozumab is a novel, 12-month treatment option for postmenopausal women at high risk for osteoporotic fracture that both increases bone formation and decreases bone resorption.
Collapse
Affiliation(s)
| | - Erin Adams
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Winchester, VA
| | - Whitney Zentgraf
- Bernard J. Dunn School of Pharmacy, Shenandoah University, Winchester, VA
| |
Collapse
|
37
|
Kobayakawa T, Suzuki T, Nakano M, Saito M, Miyazaki A, Takahashi J, Nakamura Y. Real-world effects and adverse events of romosozumab in Japanese osteoporotic patients: A prospective cohort study. Bone Rep 2021; 14:101068. [PMID: 33981812 PMCID: PMC8085670 DOI: 10.1016/j.bonr.2021.101068] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 11/27/2022] Open
Abstract
Real-world data on the new anti-sclerostin antibody drug, romosozumab, remain scarce. There is a strong need to accumulate and analyze data on romosozumab treatment for such conditions as osteoporosis. The purpose of this study was to investigate the therapeutic and adverse effects of romosozumab for osteoporosis treatment in clinical practice. Of the 230 osteoporosis patients prescribed romosozumab from September 2019 in this prospective multicenter cohort study, 204 patients completed 12 months of treatment. The primary outcome of interest was the rate of change in bone mineral density (BMD) of the lumbar spine, total hip, and femoral neck as measured by dual-energy X-ray absorptiometry. Secondary outcomes included changes in bone turnover markers and serum-corrected calcium level as well as the incidence of adverse events. At 6 and 12 months of romosozumab treatment, the respective percentage change in BMD from baseline was 7.4% and 12.2% for the lumbar spine, 1.8% and 5.8% for the total hip, and 2.9% and 6.0% for the femoral neck, all of which were significantly higher (P < 0.001) than baseline values. Patients who switched from another osteoporosis regimen exhibited significantly lower lumbar spine BMD gains versus treatment-naïve patients, especially for cases switching from denosumab. P1NP was significantly increased at 6 months (58.9%; P < 0.01), while TRACP-5b was significantly decreased at 6 months (-14.7%; P < 0.001) and 12 months (-18.8%; P < 0.001) versus baseline values. The largest rate of decrease in serum-corrected calcium was 3.7% at 12 months. Sixty-four (27.8%) of 230 patients experienced an adverse event, and 7 (3.0%) new fractures were recorded. In sum, romosozumab treatment for 12 months significantly improved lumbar spine, total hip, and femoral neck BMD according to real-world data.
Collapse
Affiliation(s)
- Tomonori Kobayakawa
- Kobayakawa Orthopedic and Rheumatologic Clinic, 1969 Kunou, Fukuroi, Shizuoka 437-0061, Japan
| | - Takako Suzuki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Department of Human Nutrition, Faculty of Human Nutrition, Tokyo Kasei Gakuin University, 22 Sanban-cho, Chiyoda-ku, Tokyo 102-8341, Japan
| | - Masaki Nakano
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Makoto Saito
- Department of Clinical Support Office, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkagome, Bunkyou-ku, Tokyo 113-8677, Japan
| | - Akiko Miyazaki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Jun Takahashi
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yukio Nakamura
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| |
Collapse
|
38
|
Abstract
Skeletal integrity is maintained by a meticulous balance between bone resorption and bone formation, and recent studies have revealed the essential role of canonical Wnt signaling pathways in maintaining skeletal homeostasis. The SOST gene, which encodes sclerostin, a member of Dan family glycoproteins, was originally identified as the gene responsible for two sclerosing bone dysplasias, sclerosteosis and van Buchem disease. Sclerostin is highly expressed by osteocytes, negatively regulates canonical Wnt signaling pathways by binding to low-density lipoprotein receptor-related protein (LRP) 5/6, and suppresses osteoblast differentiation and/or function. Romosozumab, a specific anti-sclerostin antibody, inhibits sclerostin-LRP5/6 interactions and indirectly activates canonical Wnt signaling pathways and bone formation. This review focuses on the mechanism of action of sclerostin and summarizes clinical studies that demonstrated the efficacy of romosozumab to increase bone mineral density and reduce osteoporotic fractures, as well as its cardiovascular safety.
Collapse
Affiliation(s)
- Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
39
|
Aljohani H, Stains JP, Majumdar S, Srinivasan D, Senbanjo L, Chellaiah MA. Peptidomimetic inhibitor of L-plastin reduces osteoclastic bone resorption in aging female mice. Bone Res 2021; 9:22. [PMID: 33837180 PMCID: PMC8035201 DOI: 10.1038/s41413-020-00135-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/22/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
L-plastin (LPL) was identified as a potential regulator of the actin-bundling process involved in forming nascent sealing zones (NSZs), which are precursor zones for mature sealing zones. TAT-fused cell-penetrating small molecular weight LPL peptide (TAT- MARGSVSDEE, denoted as an inhibitory LPL peptide) attenuated the formation of NSZs and impaired bone resorption in vitro in osteoclasts. Also, the genetic deletion of LPL in mice demonstrated decreased eroded perimeters and increased trabecular bone density. In the present study, we hypothesized that targeting LPL with the inhibitory LPL peptide in vivo could reduce osteoclast function and increase bone density in a mice model of low bone mass. We injected aging C57BL/6 female mice (36 weeks old) subcutaneously with the inhibitory and scrambled peptides of LPL for 14 weeks. Micro-CT and histomorphometry analyses demonstrated an increase in trabecular bone density of femoral and tibial bones with no change in cortical thickness in mice injected with the inhibitory LPL peptide. A reduction in the serum levels of CTX-1 peptide suggests that the increase in bone density is associated with a decrease in osteoclast function. No changes in bone formation rate and mineral apposition rate, and the serum levels of P1NP indicate that the inhibitory LPL peptide does not affect osteoblast function. Our study shows that the inhibitory LPL peptide can block osteoclast function without impairing the function of osteoblasts. LPL peptide could be developed as a prospective therapeutic agent to treat osteoporosis.
Collapse
Affiliation(s)
- Hanan Aljohani
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
- Department of Oral Medicine and Diagnostics Sciences, King Saud University, School of Dentistry, Riyadh, Kingdom of Saudi Arabia
| | - Joseph P Stains
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sunipa Majumdar
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Deepa Srinivasan
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Linda Senbanjo
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA
| | - Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
40
|
McClung MR. Role of bone-forming agents in the management of osteoporosis. Aging Clin Exp Res 2021; 33:775-791. [PMID: 33594648 DOI: 10.1007/s40520-020-01708-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
Recent evidence confirms the superiority of osteoanabolic therapy compared to anti-remodeling drugs for rapid improvement in bone density and fracture risk reduction, providing strong justification for the use of these anabolic agents as the initial therapy in high-risk patients, to be followed by anti-remodeling therapy. This review will highlight the results of recent studies and define the current status of osteoanabolic therapy for osteoporosis.
Collapse
Affiliation(s)
- Michael R McClung
- Oregon Osteoporosis Center, Portland, OR, USA.
- Mary MacKillop Center for Health Research, Australian Catholic University, Melbourne, VIC, Australia.
| |
Collapse
|
41
|
Osteoporosis Treatment with Anti-Sclerostin Antibodies-Mechanisms of Action and Clinical Application. J Clin Med 2021; 10:jcm10040787. [PMID: 33669283 PMCID: PMC7920044 DOI: 10.3390/jcm10040787] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/30/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis is characterized by reduced bone mass and disruption of bone architecture, resulting in increased risk of fragility fractures and significant long-term disability. Although both anti-resorptive treatments and osteoanabolic drugs, such as parathyroid hormone analogues, are effective in fracture prevention, limitations exist due to lack of compliance or contraindications to these drugs. Thus, there is a need for novel potent therapies, especially for patients at high fracture risk. Romosozumab is a monoclonal antibody against sclerostin with a dual mode of action. It enhances bone formation and simultaneously suppresses bone resorption, resulting in a large anabolic window. In this opinion-based narrative review, we highlight the role of sclerostin as a critical regulator of bone mass and present human diseases of sclerostin deficiency as well as preclinical models of genetically modified sclerostin expression, which led to the development of anti-sclerostin antibodies. We review clinical studies of romosozumab in terms of bone mass accrual and anti-fracture activity in the setting of postmenopausal and male osteoporosis, present sequential treatment regimens, and discuss its safety profile and possible limitations in its use. Moreover, an outlook comprising future translational applications of anti-sclerostin antibodies in diseases other than osteoporosis is given, highlighting the clinical significance and future scopes of Wnt signaling in these settings.
Collapse
|
42
|
Wnt signaling: An attractive target for periodontitis treatment. Biomed Pharmacother 2020; 133:110935. [PMID: 33227711 DOI: 10.1016/j.biopha.2020.110935] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
Periodontitis is the most common chronic inflammatory disease, and a leading cause of tooth loss. Characterized by resorption of alveolar process and destruction of periodontal ligaments, periodontitis can impact not only periodontal tissues but also systemic diseases, such as diabetes, cardiovascular diseases, and respiratory infections. Currently, it is a hotspot to manage destruction and gain regeneration of periodontal tissues. Increasing evidence indicates that the Wnt signaling plays an important role in homeostasis of periodontal tissues, functions of periodontal derived cells, and progression of periodontitis. Its molecule expressions were abnormal in periodontitis. As such, modulators targeting the Wnt signaling may be an adjuvant therapy for periodontitis treatment. This review elucidates the role of Wnt signaling and its molecules, with a view to develop a potential application of drugs targeting the Wnt signaling for periodontitis treatment.
Collapse
|
43
|
Wu CH, Hung WC, Chang IL, Tsai TT, Chang YF, McCloskey EV, Watts NB, McClung MR, Huang CF, Chen CH, Wu KL, Tsai KS, Chan DC, Chen JF, Tu ST, Hwang JS, Xia W, Matsumoto T, Chung YS, Cooper C, Kanis JA, Yang RS, Chan WP. Pharmacologic intervention for prevention of fractures in osteopenic and osteoporotic postmenopausal women: Systemic review and meta-analysis. Bone Rep 2020; 13:100729. [PMID: 33195764 PMCID: PMC7645632 DOI: 10.1016/j.bonr.2020.100729] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2020] [Accepted: 10/25/2020] [Indexed: 02/03/2023] Open
Abstract
Objectives Emerging evidence has indicated a role for pharmacologic agents in the primary prevention of osteoporotic fracture, but have not yet been systematically reviewed for meta-analysis. We conducted a meta-analysis to evaluate the efficacy of pharmacologic interventions in reducing fracture risk and increasing bone mineral density (BMD) in postmenopausal women with osteopenia or osteoporosis but without prevalent fragility fracture. Method The Medline, EMBASE, and CENTRAL databases were searched from inception to September 30, 2019. Only randomized placebo-controlled trials evaluating postmenopausal women with −1.0 > bone mineral density (BMD) T-score > −2.5 (low bone mass) and those with BMD T-score ≤ −2.5 (osteoporosis) but without baseline fractures, who were receiving anti-osteoporotic agents, providing quantitative outcomes data and evaluating risk of vertebral and/or non-vertebral fragility fracture at follow-up. The PRISMA guidelines were followed, applying a random-effects model. The primary endpoint was the effect of anti-osteoporotic regimens in reducing the incidence of vertebral fractures. Secondary endpoints were percentage changes in baseline BMD at the lumbar spine and total hip at 1 and 2 years follow up. Results Full-text review of 144 articles yielded, 20 for meta-analysis. Bisphosphonates reduced the risk of vertebral fracture (pooled OR = 0.50, 95%CIs = 0.36–0.71) and significantly increased lumbar spine BMD after 1 year, by 4.42% vs placebo (95%CIs = 3.70%–5.14%). At the hip, this value was 2.94% (95%CIs = 2.13%–3.75%). Overall results of limited studies for non-bisphosphonate drugs showed increased BMD and raloxifene significantly decreases the risk of subsequent clinical vertebral fractures. Conclusion The bisphosphonates are efficacious and most evident for the primary prevention of osteoporotic vertebral fractures, reducing their incidence and improving BMD in postmenopausal women with osteopenia or osteoporosis. Bisphosphonates reduced the risk of vertebral fracture in postmenopausal women with osteopenia or osteoporosis but without fracture. Bisphosphonates increased BMD in postmenopausal women with osteopenia or osteoporosis but without fracture. Limited studies for non-bisphosphonate drugs showed increased BMD in postmenopausal women with osteopenia or osteoporosis but without fracture. Raloxifene decreased the risk of clinical vertebral fractures in postmenopausal women with osteopenia or osteoporosis but without fracture.
Collapse
Affiliation(s)
- Chih-Hsing Wu
- Department of Family Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Geriatrics, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Chieh Hung
- Department of Family Medicine, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan.,Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan.,Medicine for International Student, I-Shou University, Kaohsiung, Taiwan
| | - Ing-Lin Chang
- Department of Orthopaedics, ChangHua Christian Hospital, ChangHua, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopedics, Chang Gung Memorial Hospital, Chang Gung University, Linkou, Taiwan
| | - Yin-Fan Chang
- Department of Family Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Eugene V McCloskey
- Academic Unit of Bone Metabolism, University of Sheffield, Sheffield, UK
| | - Nelson B Watts
- Mercy Health Osteoporosis and Bone Health Services, Cincinnati, OH, USA
| | | | - Chun-Feng Huang
- Department of Family Medicine, National Yang Ming University Hospital, I-Lan, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Centre, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Orthopaedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City, Taiwan.,Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Kun-Ling Wu
- Department of Family Medicine, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan City, Taiwan.,Outpatient Clinic Department, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan City, Taiwan
| | - Keh-Sung Tsai
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ding-Cheng Chan
- Superintendent Office, National Taiwan University Hospital Chu-Tung Branch, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Geriatrics and Gerontology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Fu Chen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shih-Te Tu
- Division of Endocrinology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Jawl-Shan Hwang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Linkou, Taiwan
| | - Weibo Xia
- Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, University of Tokushima, Japan
| | - Yoon-Sok Chung
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, South Korea
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.,Oxford National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John A Kanis
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, Australia.,Centre for Metabolic Bone Diseases, University of Sheffield Medical School, UK
| | - Rong-Sen Yang
- Department of Orthopaedics, College of Medicine, National Taiwan University & Hospital, Taipei, Taiwan
| | - Wing P Chan
- Department of Radiology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Kobza AO, Papaioannou A, Lau AN, Adachi JD. Romosozumab in the treatment of osteoporosis. Immunotherapy 2020; 12:965-981. [DOI: 10.2217/imt-2020-0158] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis is a disease characterized by weakening of the bone architecture, which leads to an increased risk of fracture. There has been interest in the development of osteoanabolic agents that can increase bone mass and reverse the deteriorating architecture of osteoporotic bone. Romosozumab is a new agent for osteoporosis that both promotes bone formation and inhibits bone resorption. It is a monoclonal antibody that inhibits the activity of sclerostin, which allows the Wnt pathway to promote osteoblastogenesis and inhibit the activity of bone-resorbing osteoclasts. In clinical trials, it has proven to be superior to other agents in terms of increasing bone mineral density and reducing the incidence of fractures. This review will highlight the pharmacology, clinical efficacy and safety profile of romosozumab and suggest where this medication may fit within our current management of osteoporosis.
Collapse
Affiliation(s)
- Alexandra O Kobza
- Department of Medicine, Division of Rheumatology, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Alexandra Papaioannou
- Department of Medicine, Division of Geriatric Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Department of Health Research Methods, Evidence & Impact, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Arthur N Lau
- Department of Medicine, Division of Rheumatology, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jonathan D Adachi
- Department of Medicine, Division of Rheumatology, McMaster University, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
45
|
Hamaya E. [Pharmacological characteristics and clinical study results of romosozumab (EVENITY ®; genetical recombination), a drug with novel mechanism of action to treat osteoporosis at high risk of fracture]. Nihon Yakurigaku Zasshi 2020; 155:258-267. [PMID: 32612041 DOI: 10.1254/fpj.20007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Romosozumab (EVENITY®) is a humanized monoclonal antibody designed to target sclerostin. Sclerostin is a glycoprotein that is secreted by osteocytes and that inhibits Wnt signaling in osteoblast lineage cells, leading to decreased bone formation by osteoblasts and increased bone resorption by osteoclasts. Romosozumab, by binding and inhibiting sclerostin, increases bone formation and decreases bone resorption. Romosozumab is known to mainly enable increase in modeling-based bone formation. In studies using ovariectomized (OVX) models of rats and cynomolgus monkeys, those administered romosozumab showed dose-dependently increased bone mass and strength. In addition, the bone-forming effect of romosozumab decreased after continued administration. In rats, romosozumab caused almost no focal osteoblast hyperplasia or benign or malignant bone tumors, presumably owing to the time-dependent decrease in the bone-forming effect. Clinical studies demonstrated inhibition of new vertebral fractures at 12 months of treatment, and increased bone mineral density at 6 months of treatment. With a dual effect on bone, increasing bone formation and decreasing bone resorption, romosozumab is expected to become a new treatment option, and was approved in January 2019 for the indication of "patients with osteoporosis at high risk for fracture".
Collapse
|
46
|
Bovijn J, Krebs K, Chen CY, Boxall R, Censin JC, Ferreira T, Pulit SL, Glastonbury CA, Laber S, Millwood IY, Lin K, Li L, Chen Z, Milani L, Smith GD, Walters RG, Mägi R, Neale BM, Lindgren CM, Holmes MV. Evaluating the cardiovascular safety of sclerostin inhibition using evidence from meta-analysis of clinical trials and human genetics. Sci Transl Med 2020; 12:eaay6570. [PMID: 32581134 PMCID: PMC7116615 DOI: 10.1126/scitranslmed.aay6570] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/26/2019] [Accepted: 05/26/2020] [Indexed: 12/23/2022]
Abstract
Inhibition of sclerostin is a therapeutic approach to lowering fracture risk in patients with osteoporosis. However, data from phase 3 randomized controlled trials (RCTs) of romosozumab, a first-in-class monoclonal antibody that inhibits sclerostin, suggest an imbalance of serious cardiovascular events, and regulatory agencies have issued marketing authorizations with warnings of cardiovascular disease. Here, we meta-analyze published and unpublished cardiovascular outcome trial data of romosozumab and investigate whether genetic variants that mimic therapeutic inhibition of sclerostin are associated with higher risk of cardiovascular disease. Meta-analysis of up to three RCTs indicated a probable higher risk of cardiovascular events with romosozumab. Scaled to the equivalent dose of romosozumab (210 milligrams per month; 0.09 grams per square centimeter of higher bone mineral density), the SOST genetic variants were associated with lower risk of fracture and osteoporosis (commensurate with the therapeutic effect of romosozumab) and with a higher risk of myocardial infarction and/or coronary revascularization and major adverse cardiovascular events. The same variants were also associated with increased risk of type 2 diabetes mellitus and higher systolic blood pressure and central adiposity. Together, our findings indicate that inhibition of sclerostin may elevate cardiovascular risk, warranting a rigorous evaluation of the cardiovascular safety of romosozumab and other sclerostin inhibitors.
Collapse
Affiliation(s)
- Jonas Bovijn
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Kristi Krebs
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| | - Chia-Yen Chen
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ruth Boxall
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Jenny C Censin
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Ferreira
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Sara L Pulit
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- Department of Genetics, University Medical Center Utrecht, 3584 CX Utrecht, Netherlands
| | - Craig A Glastonbury
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
| | - Samantha Laber
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Peking University, Beijing 100191, China
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Lili Milani
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Barley House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu 51010, Estonia
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Cecilia M Lindgren
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Michael V Holmes
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7FZ, UK.
- Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit (MRC PHRU), Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
47
|
Davis S, Simpson E, Hamilton J, James MMS, Rawdin A, Wong R, Goka E, Gittoes N, Selby P. Denosumab, raloxifene, romosozumab and teriparatide to prevent osteoporotic fragility fractures: a systematic review and economic evaluation. Health Technol Assess 2020; 24:1-314. [PMID: 32588816 PMCID: PMC7357239 DOI: 10.3310/hta24290] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Fragility fractures are fractures that result from mechanical forces that would not ordinarily result in fracture. OBJECTIVES The objectives were to evaluate the clinical effectiveness, safety and cost-effectiveness of non-bisphosphonates {denosumab [Prolia®; Amgen Inc., Thousand Oaks, CA, USA], raloxifene [Evista®; Daiichi Sankyo Company, Ltd, Tokyo, Japan], romosozumab [Evenity®; Union Chimique Belge (UCB) S.A. (Brussels, Belgium) and Amgen Inc.] and teriparatide [Forsteo®; Eli Lilly and Company, Indianapolis, IN, USA]}, compared with each other, bisphosphonates or no treatment, for the prevention of fragility fracture. DATA SOURCES For the clinical effectiveness review, nine electronic databases (including MEDLINE, EMBASE and the World Health Organization International Clinical Trials Registry Platform) were searched up to July 2018. REVIEW METHODS A systematic review and network meta-analysis of fracture and femoral neck bone mineral density were conducted. A review of published economic analyses was undertaken and a model previously used to evaluate bisphosphonates was adapted. Discrete event simulation was used to estimate lifetime costs and quality-adjusted life-years for a simulated cohort of patients with heterogeneous characteristics. This was done for each non-bisphosphonate treatment, a strategy of no treatment, and the five bisphosphonate treatments previously evaluated. The model was populated with effectiveness evidence from the systematic review and network meta-analysis. All other parameters were estimated from published sources. An NHS and Personal Social Services perspective was taken, and costs and benefits were discounted at 3.5% per annum. Fracture risk was estimated from patient characteristics using the QFracture® (QFracture-2012 open source revision 38, Clinrisk Ltd, Leeds, UK) and FRAX® (web version 3.9, University of Sheffield, Sheffield, UK) tools. The relationship between fracture risk and incremental net monetary benefit was estimated using non-parametric regression. A probabilistic sensitivity analysis and scenario analyses were used to assess uncertainty. RESULTS Fifty-two randomised controlled trials of non-bisphosphonates were included in the clinical effectiveness systematic review and an additional 51 randomised controlled trials of bisphosphonates were included in the network meta-analysis. All treatments had beneficial effects compared with placebo for vertebral, non-vertebral and hip fractures, with hazard ratios varying from 0.23 to 0.94, depending on treatment and fracture type. The effects on vertebral fractures and the percentage change in bone mineral density were statistically significant for all treatments. The rate of serious adverse events varied across trials (0-33%), with most between-group differences not being statistically significant for comparisons with placebo/no active treatment, non-bisphosphonates or bisphosphonates. The incremental cost-effectiveness ratios were > £20,000 per quality-adjusted life-year for all non-bisphosphonate interventions compared with no treatment across the range of QFracture and FRAX scores expected in the population eligible for fracture risk assessment. The incremental cost-effectiveness ratio for denosumab may fall below £30,000 per quality-adjusted life-year at very high levels of risk or for high-risk patients with specific characteristics. Raloxifene was dominated by no treatment (resulted in fewer quality-adjusted life-years) in most risk categories. LIMITATIONS The incremental cost-effectiveness ratios are uncertain for very high-risk patients. CONCLUSIONS Non-bisphosphonates are effective in preventing fragility fractures, but the incremental cost-effectiveness ratios are generally greater than the commonly applied threshold of £20,000-30,000 per quality-adjusted life-year. STUDY REGISTRATION This study is registered as PROSPERO CRD42018107651. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 24, No. 29. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Sarah Davis
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Emma Simpson
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Jean Hamilton
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Marrissa Martyn-St James
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Andrew Rawdin
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Ruth Wong
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Edward Goka
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Neil Gittoes
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Peter Selby
- School of Medical Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
48
|
Efficacy and safety of Romosozumab in treatment for low bone mineral density: a systematic review and meta-analysis. Clin Rheumatol 2020; 39:3261-3276. [PMID: 32385757 DOI: 10.1007/s10067-020-04948-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 01/12/2020] [Accepted: 01/16/2020] [Indexed: 01/27/2023]
Abstract
Osteoporosis is a chronic skeletal disease with an increasing prevalence. Romosozumab, as a monoclonal anti-sclerostin antibody with a dual function, has been produced. In this meta-analysis, we aimed to examine the efficacy of Romosozumab in patients with low bone mineral density. A systematic search was conducted in the most important electronic search engines like Cochrane Library, PubMed, Web of Science, Scopus, Google Scholar, and ClinicalTrials.gov at the end of July 2019 to retrieve randomized controlled trials (RCTs), which evaluated the effect of Romosozumab in patients with osteoporosis and/or low bone mineral density. After evaluating the quality of articles with the Cochrane checklist, data related to the outcomes of bone mineral density (BMD) of lumbar spine, femoral neck, and total hip, risk of clinical, vertebral and non-vertebral fractures, and risk of adverse events were extracted. Quality of evidence was assessed according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Heterogeneity between studies was evaluated by I2 and Q statistics. The meta-analysis was performed using CMA v.2.0 software. Of all the 671 initially retrieved articles, seven articles were entered into the meta-analysis after removing duplicates and reviewing papers with inclusion and exclusion criteria. The results of the meta-analysis showed that Romosozumab 210, 140, and 70 mg compared with Alendronate, Teriparatide, and placebo can increase the bone mineral density in the lumbar spine, femoral neck, and total hip. The risk of adverse events like adjudicated cardiovascular serious adverse events and adjudicated cardiovascular death was more in Romosozumab 210 mg in comparison with placebo. However, this difference was not statistically significant. Treatment with anti-sclerostin antibodies can be a proper therapeutic option in patients with osteoporosis and low bone mineral density. Based on the results of this meta-analysis, it seems that Romosozumab, with its dual function, has a positive role in the treatment of osteoporosis and low bone mineral density.
Collapse
|
49
|
Takada J, Dinavahi R, Miyauchi A, Hamaya E, Hirama T, Libanati C, Nakamura Y, Milmont CE, Grauer A. Relationship between P1NP, a biochemical marker of bone turnover, and bone mineral density in patients transitioned from alendronate to romosozumab or teriparatide: a post hoc analysis of the STRUCTURE trial. J Bone Miner Metab 2020; 38:310-315. [PMID: 31707465 DOI: 10.1007/s00774-019-01057-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Procollagen type I N-terminal propeptide (P1NP), a bone formation marker, reportedly predicts bone mineral density (BMD) response to teriparatide treatment in treatment-naive patients with osteoporosis. Results from a randomized, phase 3, open-label, active-controlled trial- STRUCTURE-showed that in patients previously treated with bisphosphonates, romosozumab led to gains in hip BMD, which were not observed with teriparatide. This post hoc analysis investigated the comparative utility of early changes in P1NP in predicting BMD response in patients who participated in the STRUCTURE trial, which enrolled patients who switched treatment from bisphosphonates to romosozumab/teriparatide. MATERIALS AND METHODS Postmenopausal women (aged 55-90 years) with osteoporosis who had previously taken bisphosphonates were randomized to receive open-label subcutaneous romosozumab (210 mg once monthly; n = 218) or teriparatide (20 µg once daily; n = 218) for 12 months. BMD was assessed by dual-energy X-ray absorptiometry at the proximal femur and lumbar spine (LS) at baseline and months 6 and 12. To assess the utility of P1NP, the positive predictive value of increase from baseline in P1NP of > 10 µg/L at month 1 and achievement of various thresholds of percent change from baseline in BMD at month 12 were evaluated. RESULTS Overall, 95% (191/202) of patients in the romosozumab group and 91% (183/201) in the teriparatide group demonstrated an increase in P1NP of > 10 µg/L from baseline at month 1. Among these patients, 18% and 3% of romosozumab-treated patients versus 60% and 12% of teriparatide-treated patients showed no increase from baseline (i.e., ≤ 0%) in total hip and LS BMD, respectively, at month 12. These data indicate that in patients switching from bisphosphonates to a bone-forming therapy, increases in P1NP do not help predict the hip BMD response. Although most patients treated with either teriparatide or romosozumab showed an increase in P1NP, the majority of patients on romosozumab showed an increase in hip BMD, while more than half of the patients on teriparatide did not. Teriparatide therapy did not increase total hip BMD in the majority of patients who transitioned from bisphosphonates to teriparatide. CONCLUSIONS Thus, increases in P1NP were not predictive of BMD response in the teriparatide group because in approximately 60% of the patients who were administered teriparatide, the hip BMD decreased independent of the change in P1NP levels.
Collapse
|
50
|
Martin M, Sansalone V, Cooper DML, Forwood MR, Pivonka P. Assessment of romosozumab efficacy in the treatment of postmenopausal osteoporosis: Results from a mechanistic PK-PD mechanostat model of bone remodeling. Bone 2020; 133:115223. [PMID: 31935526 DOI: 10.1016/j.bone.2020.115223] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/03/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Abstract
This paper introduces a theoretical framework for the study of the efficacy of romosozumab, a humanized monoclonal antibody targeting sclerostin for the treatment of osteoporosis. We developed a comprehensive mechanistic pharmacokinetic-pharmacodynamic (PK-PD) model of the effect of drug treatment on bone remodeling in postmenopausal osteoporosis (PMO). We utilized a one-compartment PK model to represent subcutaneous injections of romosozumab and subsequent absorption into serum. The PD model is based on a recently-developed bone cell population model describing the bone remodeling process at the tissue scale. The latter accounts for mechanical feedback by incorporating nitric oxide (NO) and sclerostin (Scl) as biochemical feedback molecules. Utilizing a competitive binding model, where Wnt and Scl compete for binding to LRP5/6, allows to regulate anabolic bone remodeling responses. Here, we extended this model with respect to romosozumab binding to sclerostin. For the currently approved monthly injections of 210 mg, the model predicted a 6.59%, 10.38% and 15.25% increase in BMD at the lumbar spine after 6, 12 and 24 months, respectively. These results are in good agreement with the data reported in the literature. Our model is also able to distinguish the bone-site specific drug effects. For instance, at the femoral neck, our model predicts a BMD increase of 3.85% after 12 months of 210 mg injections, which is consistent with literature observations. Finally, our simulations indicate rapid bone loss after treatment discontinuation, indicating that some additional interventions such as use of bisphosphonates are required to maintain bone mass.
Collapse
Affiliation(s)
- Madge Martin
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, 2 George St, Brisbane, QLD 4000, Australia; Laboratoire Modélisation et Simulation Multi-Echelle (MSME), UMR CNRS 8208, Université Paris-Est Créteil, 61 avenue du Général de Gaulle, Créteil 94010, France.
| | - Vittorio Sansalone
- Laboratoire Modélisation et Simulation Multi-Echelle (MSME), UMR CNRS 8208, Université Paris-Est Créteil, 61 avenue du Général de Gaulle, Créteil 94010, France
| | - David M L Cooper
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, Canada
| | - Mark R Forwood
- School of Medical Science, Griffith University, Gold Goast, QLD 4222, Australia
| | - Peter Pivonka
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, 2 George St, Brisbane, QLD 4000, Australia
| |
Collapse
|