Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Curr Osteoporos Rep ; 18(5): 449-459, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32860563

RESUMO

PURPOSE OF REVIEW: Compared with the current standard of implanting bone anabolics for fracture repair, bone fracture-targeted anabolics would be more effective, less invasive, and less toxic and would allow for control over what phase of fracture healing is being affected. We therefore sought to identify the optimal bone-targeting molecule to allow for systemic administration of therapeutics to bone fractures. RECENT FINDINGS: We found that many bone-targeting molecules exist, but most have been developed for the treatment of bone cancers, osteomyelitis, or osteoporosis. There are a few examples of bone-targeting ligands that have been developed for bone fractures that are selective for the bone fracture over the body and skeleton. Acidic oligopeptides have the ideal half-life, toxicity profile, and selectivity for a bone fracture-targeting ligand and are the most developed and promising of these bone fracture-targeting ligands. However, many other promising ligands have been developed that could be used for bone fractures.


Assuntos
Anabolizantes/administração & dosagem , Sistemas de Liberação de Medicamentos , Consolidação da Fratura , Fraturas Ósseas/tratamento farmacológico , Fosfatase Alcalina , Aminoácidos Acídicos , Difosfonatos , Durapatita , Humanos , Imunoglobulina G , Oligopeptídeos , Proteínas Recombinantes de Fusão , Fosfatase Ácida Resistente a Tartarato , Tetraciclina
2.
Bioconjug Chem ; 29(11): 3800-3809, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30380292

RESUMO

Approximately 6.3 million bone fractures occur annually in the United States, resulting in considerable morbidity, deterioration in quality of life, loss of productivity and wages, and sometimes death (e.g., hip fractures). Although anabolic and antiresorptive agents have been introduced for treatment of osteoporosis, no systemically administered drug has been developed to accelerate the fracture-healing process. To address this need, we have undertaken to target a bone anabolic agent selectively to fracture surfaces in order to concentrate the drug's healing power directly on the fracture site. We report here that conjugation of dasatinib to a bone fracture-homing oligopeptide via a releasable linker reduces fractured femur healing times in mice by ∼60% without causing overt off-target toxicity or remodeling of nontraumatized bones. Thus, achievement of healthy bone density, normal bone volume, and healthy bone mechanical properties at the fracture site is realized after only 3-4 weeks in dasatinib-targeted mice, but it requires ∼8 weeks in PBS-treated controls. We conclude that targeting of dasatinib to bone fracture surfaces can significantly accelerate the healing process at dasatinib concentrations that are known to be safe in oncological applications.


Assuntos
Ácido Aspártico/uso terapêutico , Dasatinibe/uso terapêutico , Consolidação da Fratura/efeitos dos fármacos , Fraturas Ósseas/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Ácido Aspártico/administração & dosagem , Ácido Aspártico/análogos & derivados , Ácido Aspártico/análise , Densidade Óssea/efeitos dos fármacos , Linhagem Celular , Dasatinibe/administração & dosagem , Dasatinibe/química , Sistemas de Liberação de Medicamentos , Feminino , Fraturas Ósseas/patologia , Camundongos , Oligopeptídeos/administração & dosagem , Oligopeptídeos/química , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Qualidade de Vida
3.
Biomacromolecules ; 16(10): 3145-53, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26331790

RESUMO

Bone fractures constitute a major cause of morbidity and mortality especially in the elderly. Complications associated with osteoporosis drugs and the age of the patient slow bone turnover and render such fractures difficult to heal. Increasing the speed of fracture repair by administration of a fracture-targeted bone anabolic agent could find considerable application. Aspartic acid oligopeptides are negatively charged molecules at physiological pH that adsorb to hydroxyapatite, the mineral portion of bone. This general adsorption is the strongest where bone turnover is highest or where hydroxyapatite is freshly exposed. Importantly, both of these conditions are prominent at fracture sites. GSK3ß inhibitors are potent anabolic agents that can promote tissue repair when concentrated in a damaged tissue. Unfortunately, they can also cause significant toxicity when administered systemically and are furthermore difficult to deliver due to their strong hydrophobicity. In this paper, we solve both problems by conjugating the hydrophobic GSK3ß inhibitor to a hydrophilic aspartic acid octapeptide using a hydrolyzable bond, thereby generating a bone fracture-targeted water-soluble form of the drug. The resulting amphiphile is shown to assemble into micelles, extending its circulation time while maintaining its fracture-targeting abilities. For measurement of pharmacokinetics, an 125I was introduced at the location of the bromine in the GSK3ß inhibitor to minimize any structural differences. Biodistribution studies demonstrate a greater than 4-fold increase in fracture accumulation over healthy bone.


Assuntos
Consolidação da Fratura/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Micelas , Inibidores de Proteínas Quinases/farmacocinética , Animais , Portadores de Fármacos , Glicogênio Sintase Quinase 3 beta , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Distribuição Tecidual , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
4.
Bioconjug Chem ; 25(11): 2012-20, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25291150

RESUMO

Osteosarcoma is a malignancy of the bone that primarily affects adolescents. Current treatments retain mortality rates, which are higher than average cancer mortality rates for the adolescent age group. We designed a micellar delivery system with the aim to increase drug accumulation in the tumor and potentially reduce side effects associated with chemotherapy. The design features are the use of the hydrophilic D-aspartic acid octapeptide as both the effective targeting agent as well as the hydrophilic micelle corona. Micelle stabilization was accomplished by binding of model drug (doxorubicin) via an acid-sensitive hydrazone bond and incorporating one to four 11-aminoundecanoic acid (AUA) moieties to manipulate the hydrophobic/hydrophilic ratio. Four micelle-forming unimers have been synthesized and their self-assembly into micelles was evaluated. Size of the micelles could be modified by changing the architecture of the unimers from linear to branched. The stability of the micelles increased with increasing content of AUA moieties. Adsorption of all micelles to hydroxyapatite occurred rapidly. Doxorubicin release occurred at pH 5.5, whereas no release was detected at pH 7.4. Cytotoxicity toward human osteosarcoma Saos-2 cells correlated with drug release data.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Osso e Ossos/metabolismo , Doxorrubicina/química , Micelas , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Osteossarcoma/tratamento farmacológico , Aminoácidos/química , Ácido Aspártico/química , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Liberação Controlada de Fármacos , Durapatita/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Cinética , Oligopeptídeos/síntese química
5.
Biomedicines ; 12(3)2024 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-38540225

RESUMO

Spinal fusions are performed to treat congenital skeletal malformations, spondylosis, degenerative disk diseases, and other pathologies of the vertebrae that can be resolved by reducing motion between neighboring vertebrae. Unfortunately, up to 100,000 fusion procedures fail per year in the United States, suggesting that efforts to develop new approaches to improve spinal fusions are justified. We have explored whether the use of an osteotropic oligopeptide to target an attached bone anabolic agent to the fusion site might be exploited to both accelerate the mineralization process and improve the overall success rate of spinal fusions. The data presented below demonstrate that subcutaneous administration of a modified abaloparatide conjugated to 20 mer of D-glutamic acid not only localizes at the spinal fusion site but also outperforms the standard of care (topically applied BMP2) in both speed of mineralization (p < 0.05) and overall fusion success rate (p < 0.05) in a posterior lateral spinal fusion model in male and female rats, with no accompanying ectopic mineralization. Because the bone-localizing conjugate can be administered ad libitum post-surgery, and since the procedure appears to improve on standard of care, we conclude that administration of a bone-homing anabolic agent for improvement of spinal fusion surgeries warrants further exploration.

6.
J Control Release ; 350: 688-697, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36030992

RESUMO

Targeted drug delivery, often referred to as "smart" drug delivery, is a process whereby a therapeutic drug is delivered to specific parts of the body in a manner that increases its concentration at the desired sites relative to others. This approach is poised to revolutionize medicine as exemplified by the recent FDA approval of Cytalux (FDA approves pioneering drug for ovarian cancer surgery - Purdue University News) which is a folate-receptor targeted intraoperative near infrared (NIR) imaging agent that was developed in our laboratories. Fracture-associated morbidities and mortality affect a significant portion of world population. United states, Canada and Europe alone spent $48 billion in treating osteoporosis related fractures although this number doesn't count the economic burden due to loss in productivity. It is estimated that by 2050 ca 21 million hip fractures would occur globally which will be leading cause of premature death and disability. Despite the need for improvement in the treatment for fracture repair, methods for treating fractures have changed little in recent decades. Systemic delivery of fracture-homing bone anabolics holds great promise as a therapeutic strategy in this regard. Here we report the design of a fracture-targeted peptide comprised of a payload that binds and activates the parathyroid hormone receptor (PTHR1) and is linked to a targeting ligand comprised of 20 D-glutamic acids (D-Glu20) that directs accumulation of the payload specifically at fracture sites. This targeted delivery results in reduction of fracture healing times to <1/2 while creating repaired bones that are >2-fold stronger than saline-treated controls in mice. Moreover, this hydroxyapatite-targeted peptide can be administered without detectable toxicity to healthy tissues or modification of healthy bones in dogs. Additionally, since similar results are obtained upon treatment of osteoporotic and diabetic fractures in mice, and pain resolution is simultaneously accelerated by this approach, we conclude that this fracture-targeted anabolic peptide displays significant potential to revolutionize the treatment of bone fractures.


Assuntos
Osteoporose , Fraturas por Osteoporose , Animais , Densidade Óssea , Cães , Ácido Fólico , Glutamatos/uso terapêutico , Hidroxiapatitas/uso terapêutico , Ligantes , Camundongos , Osteoporose/tratamento farmacológico , Fraturas por Osteoporose/tratamento farmacológico , Peptídeos/uso terapêutico , Receptor Tipo 1 de Hormônio Paratireóideo , Estados Unidos
7.
J Control Release ; 329: 570-584, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33031877

RESUMO

PURPOSE: Although more than 18,000,000 fractures occur each year in the US, methods to promote fracture healing still rely primarily on fracture stabilization, with use of bone anabolic agents to accelerate fracture repair limited to rare occasions when the agent can be applied to the fracture surface. Because management of broken bones could be improved if bone anabolic agents could be continuously applied to a fracture over the entire course of the healing process, we undertook to identify strategies that would allow selective concentration of bone anabolic agents on a fracture surface following systemic administration. Moreover, because hydroxyapatite is uniquely exposed on a broken bone, we searched for molecules that would bind with high affinity and specificity for hydroxyapatite. We envisioned that by conjugating such osteotropic ligands to a bone anabolic agent, we could acquire the ability to continuously stimulate fracture healing. RESULTS: Although bisphosphonates and tetracyclines were capable of localizing small amounts of peptidic payloads to fracture surfaces 2-fold over healthy bone, their specificities and capacities for drug delivery were significantly inferior to subsequent other ligands, and were therefore considered no further. In contrast, short oligopeptides of acidic amino acids were found to localize a peptide payload to a bone fracture 91.9 times more than the control untargeted peptide payload. Furthermore acidic oligopeptides were observed to be capable of targeting all classes of peptides, including hydrophobic, neutral, cationic, anionic, short oligopeptides, and long polypeptides. We further found that highly specific bone fracture targeting of multiple peptidic cargoes can be achieved by subcutaneous injection of the construct. CONCLUSIONS: Using similar constructs, we anticipate that healing of bone fractures in humans that have relied on immobilization alone can be greately enhanced by continuous stimulation of bone growth using systemic administration of fracture-targeted bone anabolic agents.


Assuntos
Fraturas Ósseas , Osso e Ossos , Difosfonatos , Consolidação da Fratura , Fraturas Ósseas/tratamento farmacológico , Humanos , Ligantes
8.
Arthritis Res Ther ; 21(1): 143, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31174578

RESUMO

OBJECTIVES: Most therapies for autoimmune and inflammatory diseases either neutralize or suppress production of inflammatory cytokines produced by activated macrophages (e.g., TNFα, IL-1, IL-6, IL-17, GM-CSF). However, no approved therapies directly target this activated subset of macrophages. METHODS: First, we undertook to examine whether the folate receptor beta (FR-ß) positive subpopulation of macrophages, which marks the inflammatory subset in animal models of rheumatoid arthritis, might constitute the prominent population of macrophages in inflamed lesions in humans. Next, we utilized anti-FR-ß monoclonal antibodies capable of mediating antibody-dependent cell cytotoxicity (ADCC) to treat animal models of rheumatoid arthritis and peritonitis. RESULTS: Human tissue samples of rheumatoid arthritis, Crohn's disease, ulcerative colitis, idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, chronic obstructive pulmonary disease, systemic lupus erythematosus, psoriasis, and scleroderma are all characterized by dramatic accumulation of macrophages that express FR-ß, a protein not expressed on resting macrophages or any other healthy tissues. A monoclonal antibody to FR-ß accumulates specifically in inflamed lesions of murine inflammatory disease models and successfully treats such models of rheumatoid arthritis and peritonitis. More importantly, elimination of FR-ß-positive macrophages upon treatment with an anti-FR-ß monoclonal antibody promotes the departure of other immune cells, including T cells, B cells, neutrophils, and dendritic cells from the inflamed lesions. CONCLUSIONS: These data suggest that specific elimination of FR-ß-expressing macrophages may constitute a highly specific therapy for multiple autoimmune and inflammatory diseases and that a recently developed human anti-human FR-ß monoclonal antibody (m909) might contribute to suppression of this subpopulation of macrophages.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Artrite Reumatoide/imunologia , Receptor 2 de Folato/imunologia , Imunidade Celular , Ativação de Macrófagos , Macrófagos/metabolismo , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos
9.
Nanomedicine (Lond) ; 12(3): 185-193, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28093944

RESUMO

AIM: To evaluate the fracture healing capabilities of a GSK3ß inhibitor, 6-bromoindirubin-3'-oxime, coupled with an aspartic acid octapeptide in a micellar delivery system. MATERIALS & METHODS: The efficacy of the intravenously administered micelles to accelerate healing of femoral fracture in mice was evaluated. Micro-computed tomography analysis was employed to obtain bone density, total volume, relative volume, trabecular thickness and trabecular spacing. RESULTS: Both fracture bone mineral density and volume were significantly higher in the micelle treatment groups when compared with controls. The fracture-targeted micelle demonstrates fracture-specific bone anabolism and biocompatibility in off-target tissues. CONCLUSION: Accelerated fracture healing in mice was achieved by targeting the GSK3ß inhibitor, 6-bromoindirubin-3'-oxime, to the fracture site.


Assuntos
Consolidação da Fratura/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Indóis/uso terapêutico , Oximas/uso terapêutico , Animais , Densidade Óssea/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Humanos , Indóis/administração & dosagem , Indóis/farmacologia , Masculino , Camundongos , Micelas , Estrutura Molecular , Terapia de Alvo Molecular , Osteogênese/efeitos dos fármacos , Oximas/administração & dosagem , Oximas/farmacologia , Propriedades de Superfície , Distribuição Tecidual , Microtomografia por Raio-X
10.
Biomaterials ; 35(27): 7887-95, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24930854

RESUMO

For small interfering RNA (siRNA)-based cancer therapies, we report an actively-targeted and stabilized polyion complex micelle designed to improve tumor accumulation and cancer cell uptake of siRNA following systemic administration. Improvement in micelle stability was achieved using two stabilization mechanisms; covalent disulfide cross-linking and non-covalent hydrophobic interactions. The polymer component was designed to provide disulfide cross-linking and cancer cell-targeting cyclic RGD peptide ligands, while cholesterol-modified siRNA (Chol-siRNA) provided additional hydrophobic stabilization to the micelle structure. Dynamic light scattering confirmed formation of nano-sized disulfide cross-linked micelles (<50 nm in diameter) with a narrow size distribution. Improved stability of Chol-siRNA-loaded micelles (Chol-siRNA micelles) was demonstrated by resistance to both the dilution in serum-containing medium and counter polyion exchange with dextran sulfate, compared to control micelles prepared with Chol-free siRNA (Chol-free micelles). Improved stability resulted in prolonged blood circulation time of Chol-siRNA micelles compared to Chol-free micelles. Furthermore, introduction of cRGD ligands onto Chol-siRNA micelles significantly facilitated accumulation of siRNA in a subcutaneous cervical cancer model following systemic administration. Ultimately, systemically administered cRGD/Chol-siRNA micelles exhibited significant gene silencing activity in the tumor, presumably due to their active targeting ability combined with the enhanced stability through both hydrophobic interactions of cholesterol and disulfide cross-linking.


Assuntos
Colesterol/química , Reagentes de Ligações Cruzadas/química , Dissulfetos/química , Técnicas de Transferência de Genes , Micelas , Neoplasias/metabolismo , RNA Interferente Pequeno/metabolismo , Amidinas/síntese química , Amidinas/química , Animais , Endocitose , Feminino , Células HeLa , Humanos , Íons , Luz , Camundongos Nus , Neoplasias/patologia , Oligopeptídeos/síntese química , Oligopeptídeos/química , Peptídeos Cíclicos/síntese química , Peptídeos Cíclicos/química , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polilisina/síntese química , Polilisina/química , Espectroscopia de Prótons por Ressonância Magnética , Espalhamento de Radiação
11.
Adv Drug Deliv Rev ; 64(12): 1189-204, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22316530

RESUMO

An aging population in the developing world has led to an increase in musculoskeletal diseases such as osteoporosis and bone metastases. Left untreated many bone diseases cause debilitating pain and in the case of cancer, death. Many potential drugs are effective in treating diseases but result in side effects preventing their efficacy in the clinic. Bone, however, provides a unique environment of inorganic solids, which can be exploited in order to effectively target drugs to diseased tissue. By integration of bone targeting moieties to drug-carrying water-soluble polymers, the payload to diseased area can be increased while side effects decreased. The realization of clinically relevant bone targeted polymer therapeutics depends on (1) understanding bone targeting moiety interactions, (2) development of controlled drug delivery systems, as well as (3) understanding drug interactions. The latter makes it possible to develop bone targeted synergistic drug delivery systems.


Assuntos
Doenças Ósseas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Polímeros/química , Envelhecimento , Animais , Doenças Ósseas/fisiopatologia , Osso e Ossos/metabolismo , Preparações de Ação Retardada , Portadores de Fármacos/química , Desenho de Fármacos , Interações Medicamentosas , Sinergismo Farmacológico , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA