Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Ther ; 40(4): 1530-1545, 2023 04.
Article in English | MEDLINE | ID: mdl-36719566

ABSTRACT

INTRODUCTION: X-linked hypophosphataemia (XLH) is a rare, genetic renal phosphate-wasting disease, resulting from excess fibroblast growth factor 23 (FGF23) activity, which has a progressive and profound impact on patients throughout life. The monoclonal anti-FGF23 antibody, burosumab, is a subcutaneous injection indicated for the treatment of XLH in children and adults. Originally, burosumab was approved to be administered by a healthcare professional (HCP), but the option of self-administration would enable patient independence and easier access to treatment. Two open-label, single-arm clinical trials, conducted in Japan and Korea, have assessed the safety and efficacy of self-administration of burosumab in both children and adults with XLH. METHODS: In KRN23-003 (n = 15 children aged 1-12 years) and KRN23-004 (n = 5 children aged 3-13 years, n = 4 adults aged 21-65 years), children initially received 0.8 mg/kg of burosumab every 2 weeks and adults initially received 1.0 mg/kg of burosumab every 4 weeks. Self-administration was permitted from Week 4, and patients or carers were provided with training to inject correctly. RESULTS: In both trials, burosumab had an acceptable safety profile with mainly mild-to-moderate adverse events. Following self-administration, no patients reported serious treatment-emergent adverse events ≥ grade 3, injection-site reactions or hypersensitivity reactions related to burosumab. Serum phosphate and active vitamin D levels increased from baseline in children and adults. CONCLUSIONS: These results indicated that the efficacy and safety of burosumab when administered either by a carer or patient are similar to that when administered by an HCP and show that self-administration is a viable option for patients with XLH. TRIAL REGISTRATION NUMBERS: NCT03233126 and NCT04308096.


Subject(s)
Antibodies, Monoclonal , Familial Hypophosphatemic Rickets , Humans , Adult , Child , Antibodies, Monoclonal/adverse effects , Familial Hypophosphatemic Rickets/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Phosphates/therapeutic use
2.
J Endocr Soc ; 6(5): bvac021, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35356008

ABSTRACT

Objective: Burosumab, an anti-fibroblast growth factor 23 antibody, was recently approved for the treatment of X-linked hypophosphatemia (XLH).We evaluated the safety and efficacy of burosumab in pediatric XLH patients. Methods: This open-label, phase 3/4 trial of ≤ 124 weeks' duration was conducted at 4 Japanese medical centers. Fifteen children aged 1 to 12 years with XLH were included. All had previously been treated with phosphorus or vitamin D. Subcutaneous burosumab was administered every 2 weeks, starting with 0.8 mg/kg, and adjusted based on serum phosphorus levels and any safety concerns (maximum 2 mg/kg). Safety assessments included the frequency of treatment-emergent adverse events (TEAEs). Efficacy of burosumab on biochemical markers, clinical markers of rickets, motor function, and growth was also evaluated. Results: The average treatment duration was 121.7 weeks. Frequently reported TEAEs were nasopharyngitis (46.7%), dental caries (40.0%), and influenza (33.3%). At baseline, patients had low serum phosphorus concentrations (2.6 ±â€…0.3 mg/dL) and low-to-normal 1,25-dihydroxyvitamin D concentrations (24.7 ±â€…12.7 pg/mL), which increased with burosumab treatment and were maintained during the study period. Alkaline phosphatase decreased continuously. At baseline, the mean ±â€…SD total Thacher Rickets Severity Score (RSS) was 1.3 ±â€…1.2, and 4 patients (26.7%) had an RSS ≥ 2.0. Mean Radiographic Global Impression of Change and RSS tended to improve, particularly in patients with higher baseline RSS. There was a trend toward increased 6-minute walk test distance. No apparent changes in growth rate were observed. Conclusion: Burosumab has a good safety profile and is effective in pediatric patients with XLH.

3.
J Bone Miner Res ; 36(2): 262-270, 2021 02.
Article in English | MEDLINE | ID: mdl-32967046

ABSTRACT

Patients with tumor-induced osteomalacia (TIO), an acquired paraneoplastic condition characterized by osteomalacia due to hypophosphatemia, exhibit a similar clinical picture to those with X-linked hypophosphatemic rickets/osteomalacia (XLH). The human monoclonal anti-fibroblast growth factor 23 (FGF23) antibody burosumab (KRN23) increases serum phosphate and improves bone turnover, fracture healing, pain, and physical function in XLH patients by inhibiting circulating FGF23; thus, burosumab is expected to be an effective treatment for TIO. We report here an interim analysis of a multicenter, open-label, intraindividual dose-adjustment study of burosumab (0.3 to 2.0 mg/kg every 4 weeks) in Japanese and Korean TIO patients. The primary endpoint was the fasting serum phosphate level at each visit. Key secondary endpoints were changes over time in bone biomarkers, pharmacodynamic markers, bone histomorphometric parameters, motor function, and patient-reported outcomes. Safety was assessed based on treatment-emergent adverse events (TEAEs). Thirteen patients received burosumab treatment, of whom 4 underwent bone biopsy. The mean dose after week 112 was approximately 1.0 mg/kg. After the first burosumab administration, mean serum phosphate levels increased and remained above the lower limit of normal and in the normal range from weeks 14 to 112. Bone biomarkers initially increased, reaching maximum values at week 16 or 24, and then gradually decreased. After burosumab treatment, patients were able to walk further (evaluated by the 6-minute walk test), reported decreased pain levels, and showed a tendency toward healing of baseline fractures and pseudofractures. Two patients discontinued, one each due to disease progression and consent withdrawal. Burosumab was generally well tolerated, with no treatment-related TEAEs of grade ≥3 and no treatment-related serious AEs. In conclusion, the interim results of this first study of burosumab to treat TIO patients indicate that this drug has the potential to provide clinical benefit for patients with unresectable tumors. The full study results are eagerly anticipated. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)..


Subject(s)
Familial Hypophosphatemic Rickets , Neoplasms, Connective Tissue , Osteomalacia , Antibodies, Monoclonal, Humanized , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Humans , Paraneoplastic Syndromes
4.
Chemistry ; 17(44): 12372-84, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21954012

ABSTRACT

The rate constants of intermolecular photoinduced electron transfer from triplet excited states of metalloporphyrins to a series of p-benzoquinone derivatives in benzonitrile were determined to examine the effects of the driving force, the metal, and the conformational distortion of the porphyrin ring on the reorganization energies (λ) of electron transfer by laser flash photolysis. The λ values were evaluated from the determined rate constants on the basis of the Marcus theory of electron transfer. The λ values of planar metalloporphyrins, [Al(TPP)(PhCOO)] and [Zn(TPP)] (TPP(2-)=tetraphenylporphyrin dianion), are approximately the same, but they are 0.27 eV smaller than those of the corresponding nonplanar (saddle-distorted) metalloporphyrins [Al(DPP)(PhCOO)] and [Zn(DPP)] (DPP(2-)=dodecaphenylporphyrin dianion) when they are compared for the same driving force of photoinduced electron transfer. The axial ligand PhCOO(-) of [Al(TPP)](+) and [Al(DPP)](+) was replaced by anthraquinone-2-carboxylate (AqCOO(-)) to afford the electron donor-acceptor complexes [Al(TPP)(AqCOO)] and [Al(DPP)(AqCOO)], respectively. The X-ray crystal structure of [Al(TPP)(AqCOO)] revealed strong coordination of AqCOO(-) to the Al(3+) ion of [Al(TPP)](+) and the existence of π-π interactions between AqCOO(-) and the porphyrin ring. In the case of the saddle-distorted [Al(DPP)(AqCOO)], however, the AqCOO(-) moiety is nearly perpendicular to the porphyrin ring. The photodynamics of intracomplex photoinduced electron transfer from the singlet excited state of [Al(TPP)](+) and [Al(DPP)](+) to the AqCOO(-) moiety were also examined in comparison with the intermolecular photoinduced electron-transfer reactions, and the determined rate constants were evaluated in light of the Marcus theory of electron transfer to reveal that the electron transfer is adiabatic in each case.


Subject(s)
Benzoquinones/chemistry , Metalloporphyrins/chemistry , Quinones/chemistry , Crystallography, X-Ray , Electrochemistry , Electron Transport , Models, Molecular , Oxidation-Reduction
5.
Exp Cell Res ; 314(17): 3232-44, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18775697

ABSTRACT

When damaged, skeletal muscle regenerates. In the early phases of regeneration, inflammatory cells such as neutrophils/granulocytes and macrophages infiltrate damaged muscle tissue. To reveal the roles of macrophages during skeletal muscle regeneration, we injected an antibody, AFS98 that blocks the binding of M-CSF to its receptor into normal mice that received muscle damages. Anti-M-CSF receptor administration suppressed macrophage but not neutrophil infiltration. Histological study indicated that suppression of macrophages function leads to the incomplete muscle regeneration. In addition FACS and immunohistochemical study showed that the acute lack of macrophages delayed proliferation and differentiation of muscle satellite cells in vivo. Furthermore, mice injected with the anti-M-CSF receptor antibody exhibited not only adipogenesis, but also significant collagen deposition, i.e., fibrosis and continuous high expression of connective tissue growth factor. Finally we indicate that these fibrosis markers were strongly enriched in CD90(+) cells that do not include myogenic cells. These results indicate that macrophages directly affect satellite cell proliferation and that a macrophage deficiency severely impairs skeletal muscle regeneration and causes fibrosis.


Subject(s)
Fibrosis/pathology , Macrophages/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiology , Regeneration , Animals , Antibodies, Monoclonal/metabolism , Antigens, CD/metabolism , Biomarkers/metabolism , Cadherins/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Macrophages/cytology , Mice , Mice, Inbred C3H , Muscle, Skeletal/cytology , Random Allocation , Rats , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...