Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Int J Mol Sci ; 22(14)2021 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-34299008

RESUMO

Angiogenesis has a pivotal role in tumor growth and the metastatic process. Molecular imaging was shown to be useful for imaging of tumor-induced angiogenesis. A great variety of radiolabeled peptides have been developed to target αvß3 integrin, a target structure involved in the tumor-induced angiogenic process. The presented study aimed to synthesize deferoxamine (DFO)-based c(RGD) peptide conjugate for radiolabeling with gallium-68 and perform its basic preclinical characterization including testing of its tumor-imaging potential. DFO-c(RGDyK) was labeled with gallium-68 with high radiochemical purity. In vitro characterization including stability, partition coefficient, protein binding determination, tumor cell uptake assays, and ex vivo biodistribution as well as PET/CT imaging was performed. [68Ga]Ga-DFO-c(RGDyK) showed hydrophilic properties, high stability in PBS and human serum, and specific uptake in U-87 MG and M21 tumor cell lines in vitro and in vivo. We have shown here that [68Ga]Ga-DFO-c(RGDyK) can be used for αvß3 integrin targeting, allowing imaging of tumor-induced angiogenesis by positron emission tomography.


Assuntos
Desferroxamina/química , Radioisótopos de Gálio/química , Glioblastoma/diagnóstico por imagem , Integrina alfaVbeta3/metabolismo , Neovascularização Patológica/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Animais , Linhagem Celular Tumoral , Desferroxamina/análogos & derivados , Desferroxamina/síntese química , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Tecidual , Tomografia Computadorizada por Raios X/métodos , Transplante Heterólogo
2.
ACS Appl Mater Interfaces ; 13(8): 9729-9738, 2021 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-33599495

RESUMO

Intracerebral hemorrhage (ICH) will be accompanied by the overload of iron and reactive oxygen species (ROS) following hematoma clearance. Although deferoxamine (DFO) has been widely utilized as a clinical first-line siderophore to remove the iron overload, the ROS-inducing damage still greatly limits the therapeutic effect of DFO. To address this issue, we designed and fabricated a series of dual-functional macromolecular nanoscavengers featuring high-density DFO units and catechol moieties. Note that the former units could effectively remove the iron overload, while the latter ones could efficiently deplete the ROS. The resulting nanoscavengers efficiently down-regulate the iron and ROS levels as well as significantly reduce the cell death in both iron-overloaded RAW 264.7 cells and the ICH mice model. This work suggests a novel clue for the ICH-ameliorated iron-depleting interventional therapeutic regimen.


Assuntos
Antioxidantes/uso terapêutico , Hemorragia Cerebral/tratamento farmacológico , Sobrecarga de Ferro/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Polímeros/uso terapêutico , Sideróforos/uso terapêutico , Animais , Antioxidantes/síntese química , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Catecóis/síntese química , Catecóis/uso terapêutico , Hemorragia Cerebral/induzido quimicamente , Hemorragia Cerebral/complicações , Hemorragia Cerebral/fisiopatologia , Colagenases , Desferroxamina/análogos & derivados , Desferroxamina/uso terapêutico , Ferro/metabolismo , Sobrecarga de Ferro/etiologia , Sobrecarga de Ferro/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Neuroprotetores/síntese química , Polímeros/síntese química , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Sideróforos/síntese química
3.
Molecules ; 27(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35011419

RESUMO

Desferrioxamine (DFO) is the current "gold standard" chelator for 89Zr4+, which is used to label monoclonal antibodies for applications in immunopositron emission tomography. Recently, controversial data have been reported regarding the speciation and the stability of the complexes formed by DFO with Zr4+ in solution. To shed some light on this point, we studied the coordination properties in solution ofa chromophoric DFO derivative bearing a substituted pyrimidine residue (DFO-Pm) toward several metal ions (Zr4+, Cu2+, Zn2+, Mg2+, Ca2+, Na+, K+). Potentiometric titrations showed that DFO-Pm and pristine DFO form complexes with very similar stoichiometry and stability. DFO-Pm, which can consequently be taken as a model system for DFO, provides a photochemical response to metal coordination that can be used to further define the complexes formed. In the critical case of Zr4+, spectrophotometric measurements allowed the verification of the formation of 1:1 and 2:3 complexes that, together with 2:2 complexes form the coordination model that was obtained through the use of our potentiometric measurements. Additionally, mass spectrometry measurements verified the formation of 1:1 and 2:3 complexes and showed that 1:2 species can be easily generated through the fragmentation of the 2:3 species. In conclusion, the results obtained with DFO-Pm validate the complexation model of Zr4+/DFO composed of 1:1, 2:2, and 2:3 metal-to-ligand complexes. Convergences and conflicts with other works are addressed.


Assuntos
Complexos de Coordenação/química , Desferroxamina/análogos & derivados , Desferroxamina/química , Quelantes/química , Fenômenos Químicos , Modelos Teóricos , Estrutura Molecular , Espectrometria de Massas por Ionização por Electrospray , Análise Espectral , Termodinâmica , Zircônio/química
4.
Nucl Med Biol ; 90-91: 23-30, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32957056

RESUMO

INTRODUCTION: Radiolabeling of stem cells with a positron emitting radioisotope represents a major advancement in regenerative biotherapy enabling non-invasive imaging. To assess the value of such an approach in a clinically relevant scenario, the tolerability and therapeutic aptitude of [89Zr]zirconium-p-isothiocyanatobenzyl-desferrioxamine ([89Zr]Zr-DBN) labeled human cardiopoietic stem cells (CPs) were evaluated in a model of ischemic heart failure. METHODS AND RESULTS: [89Zr]Zr-DBN based radiolabeling of human CPs yielded [89Zr]Zr-DBN-CPs with radioactivity yield of 0.70 ± 0.20 MBq/106 cells and excellent label stability. Compared to unlabeled cell counterparts, [89Zr]Zr-DBN-CPs maintained morphology, viability, and proliferation capacity with characteristic expression of mesodermal and pro-cardiogenic transcription factors defining the cardiopoietic phenotype. Administered in chronically infarcted murine hearts, [89Zr]Zr-DBN-CPs salvaged cardiac pump failure, documented by improved left ventricular ejection fraction not inferior to unlabeled CPs and notably superior to infarcted hearts without cell treatment. CONCLUSION: The present study establishes that [89Zr]Zr-DBN labeling does not compromise stem cell identity or efficacy in the setting of heart failure, offering a non-invasive molecular imaging platform to monitor regenerative biotherapeutics post-transplantation.


Assuntos
Desferroxamina/análogos & derivados , Insuficiência Cardíaca/patologia , Isotiocianatos/química , Radioisótopos/química , Células-Tronco/metabolismo , Zircônio/química , Animais , Desferroxamina/química , Camundongos , Tomografia por Emissão de Pósitrons , Coloração e Rotulagem , Células-Tronco/patologia , Volume Sistólico
5.
Int J Mol Sci ; 21(14)2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32679799

RESUMO

Proteins, as a major component of organisms, are considered the preferred biomaterials for drug delivery vehicles. Hemoglobin (Hb) has been recently rediscovered as a potential drug carrier, but its use for biomedical applications still lacks extensive investigation. To further explore the possibility of utilizing Hb as a potential tumor targeting drug carrier, we examined and compared the biodistribution of Hb in healthy and lung tumor-bearing mice, using for the first time 89Zr labelled Hb in a positron emission tomography (PET) measurement. Hb displays a very high conjugation yield in its fast and selective reaction with the maleimide-deferoxamine (DFO) bifunctional chelator. The high-resolution X-ray structure of the Hb-DFO complex demonstrated that cysteine ß93 is the sole attachment moiety to the αß-protomer of Hb. The Hb-DFO complex shows quantitative uptake of 89Zr in solution as determined by radiochromatography. Injection of 0.03 mg of Hb-DFO-89Zr complex in healthy mice indicates very high radioactivity in liver, followed by spleen and lungs, whereas a threefold increased dosage results in intensification of PET signal in kidneys and decreased signal in liver and spleen. No difference in biodistribution pattern is observed between naïve and tumor-bearing mice. Interestingly, the liver Hb uptake did not decrease upon clodronate-mediated macrophage depletion, indicating that other immune cells contribute to Hb clearance. This finding is of particular interest for rapidly developing clinical immunology and projects aiming to target, label or specifically deliver agents to immune cells.


Assuntos
Portadores de Fármacos/farmacocinética , Sistemas de Liberação de Medicamentos , Hemoglobinas/farmacocinética , Neoplasias Pulmonares/metabolismo , Pulmão/metabolismo , Animais , Linhagem Celular Tumoral , Complexos de Coordenação/química , Complexos de Coordenação/farmacocinética , Desferroxamina/análogos & derivados , Desferroxamina/farmacocinética , Portadores de Fármacos/química , Feminino , Hemoglobinas/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Radioisótopos/química , Radioisótopos/farmacocinética , Distribuição Tecidual , Zircônio/química , Zircônio/farmacocinética
6.
PLoS One ; 15(1): e0223814, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31910217

RESUMO

INTRODUCTION: Chimeric antigen receptor (CAR) T-cells have been recently developed and are producing impressive outcomes in patients with hematologic malignancies. However, there is no standardized method for cell trafficking and in vivo CAR T-cell monitoring. We assessed the feasibility of real-time in vivo 89Zr-p-Isothiocyanatobenzyl-desferrioxamine (Df-Bz-NCS, DFO) labeled CAR T-cell trafficking using positron emission tomography (PET). RESULTS: The 89Zr-DFO radiolabeling efficiency of Jurkat/CAR and human peripheral blood mononuclear cells (hPBMC)/CAR T-cells was 70%-79%, and cell radiolabeling activity was 98.1-103.6 kBq/106 cells. Cell viability after radiolabeling was >95%. Cell proliferation was not significantly different during the early period after radiolabeling, compared with unlabeled cells; however, the proliferative capacity decreased over time (day 7 after labeling). IL-2 or IFN-γ secretion was not significantly different between unlabeled and labeled CAR T-cells. PET/magnetic resonance imaging in the xenograft model showed that most of the 89Zr-DFO-labeled Jurkat/CAR T-cells were distributed in the lung (24.4% ± 3.4%ID) and liver (22.9% ± 5.6%ID) by one hour after injection. The cells gradually migrated from the lung to the liver and spleen by day 1, and remained stable in these sites until day 7 (on day 7: lung 3.9% ± 0.3%ID, liver 36.4% ± 2.7%ID, spleen 1.4% ± 0.3%ID). No significant accumulation of labeled cells was identified in tumors. A similar pattern was observed in ex vivo biodistributions on day 7 (lung 3.0% ± 1.0%ID, liver 19.8% ± 2.2%ID, spleen 2.3% ± 1.7%ID). 89Zr-DFO-labeled hPBMC/CAR T-cells showed a similar distribution, compared with Jurkat/CAR T-cells, on serial PET images. CAR T cell distribution was cross-confirmed by flow cytometry, Alu polymerase chain reaction, and immunohistochemistry. CONCLUSION: Real-time in vivo cell trafficking is feasible using PET imaging of 89Zr-DFO-labeled CAR T-cells. This can be used to investigate cellular kinetics, initial in vivo biodistribution, and safety profiles in future CAR T-cell development.


Assuntos
Desferroxamina/análogos & derivados , Isotiocianatos/farmacologia , Radioisótopos/farmacologia , Receptores de Antígenos de Linfócitos T/isolamento & purificação , Receptores de Antígenos Quiméricos/isolamento & purificação , Zircônio/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desferroxamina/farmacologia , Neoplasias Hematológicas/tratamento farmacológico , Neoplasias Hematológicas/patologia , Humanos , Imunoconjugados/farmacologia , Marcação por Isótopo , Células Jurkat , Leucócitos Mononucleares/química , Leucócitos Mononucleares/efeitos dos fármacos , Tomografia por Emissão de Pósitrons , Radioisótopos/química , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/uso terapêutico , Receptores de Antígenos Quiméricos/química , Receptores de Antígenos Quiméricos/uso terapêutico , Linfócitos T/química , Linfócitos T/imunologia , Distribuição Tecidual
7.
Mol Pharm ; 16(5): 1950-1957, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30986347

RESUMO

Platelet-derived growth factor receptor ß (PDGFRß) is overexpressed in a variety of malignant cancers, plays a critical role in tumor angiogenesis, and has been proven as a valuable target for cancer treatment. In this pilot study, a dimeric affibody molecule, ZPDGFRß, was prepared and radiolabeled with positron emission radionuclide zirconium-89 for PET imaging of colorectal tumors by targeting PDGFRß expression in vivo. The PDGFRß-binding capability of dimeric affibody was evaluated by flow cytometry, immunofluorescent staining, and whole-body optical imaging. Then, ZPDGFRß was conjugated with DFO-Bn-NCS and radiolabeled with 89Zr. Targeted binding capability of 89Zr-DFO-ZPDGFRß to PDGFRß expressing cells was investigated by cellular assay in vitro and microPET/CT imaging in vivo. Dimeric ZPDGFRß affibody had specifically higher binding capability with PDGFRß expressing pericytes rather than LS-174T cancer cells, and well colocalized with tumor neovasculature by flow cytometry and immunofluorescent assay. ZPDGFRß was successfully labeled with 89Zr by DFO chelating with yield of 94.1 ± 3.53%. 89Zr-DFO-ZPDGFRß indicated preserved specific binding ability with PDGFRß expressing cells and effective inhibiting capability to PDGF-ß ligands ( P < 0.05) in vitro. Biodistribution indicated that tumor uptake of 89Zr-DFO-ZPDGFRß reached the peak of 6.93 ± 0.64%ID/g, and the tumor-to-blood ratio was 5.5 ± 0.6 at 2 h post-injection. LS-174T xenografts were clearly visualized by microPET/CT imaging through 1 to 4 h post-injection of 89Zr-DFO-ZPDGFRß affibody conjugate. In conclusion, the 89Zr-DFO-ZPDGFRß conjugate demonstrated specific and high binding ability with colorectal tumor, which indicated its use as a potential radiopharmaceutical for diagnostic imaging of tumor associate vasculatures with PET/CT.


Assuntos
Anticorpos Monoclonais/química , Neoplasias Colorretais/diagnóstico por imagem , Neoplasias Colorretais/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Radioisótopos/química , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Zircônio/química , Animais , Anticorpos Monoclonais/metabolismo , Células 3T3 BALB , Linhagem Celular Tumoral , Desferroxamina/análogos & derivados , Desferroxamina/química , Humanos , Camundongos , Camundongos Nus , Pericitos/metabolismo , Projetos Piloto , Radioisótopos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/imunologia , Distribuição Tecidual , Zircônio/metabolismo
8.
Biometals ; 32(3): 395-408, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30701380

RESUMO

Desferrioxamine B (DFOB) is a siderophore native to Streptomyces pilosus biosynthesised by the DesABCD enzyme cluster as a high affinity Fe(III) chelator. Although DFOB has a long clinical history for the treatment of chronic iron overload, limitations encourage the development of new analogues. This review describes a recent body of work that has used precursor-directed biosynthesis (PDB) to access new DFOB analogues. PDB exploits the native biosynthetic machinery of a producing organism in culture medium augmented with non-native substrates that compete against native substrates during metabolite assembly. The method allows access to analogues of natural products using benign methods, compared to multistep organic synthesis. The disadvantages of PDB are the production of metabolites in low yield and the need to purify complex mixtures. Streptomyces pilosus medium was supplemented with different types of non-native diamine substrates to compete against native 1,5-diaminopentane to generate DFOB analogues containing alkene bonds, fluorine atoms, ether or thioether functional groups, or a disulfide bond. All analogues retained function as Fe(III) chelators and have properties that could broaden the utility of DFOB. These PDB studies have also added knowledge to the understanding of DFOB biosynthesis.


Assuntos
Desferroxamina/metabolismo , Quelantes de Ferro/metabolismo , Streptomyces/química , Desferroxamina/análogos & derivados , Desferroxamina/química , Quelantes de Ferro/química , Estrutura Molecular , Streptomyces/metabolismo
9.
Cancer Biother Radiopharm ; 34(4): 209-217, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30676778

RESUMO

Background: The success of human epidermal growth factor receptor 2 (HER2)-targeted therapy depends on accurate characterization of HER2 expression, but current methods available have several limitations. This study aims to investigate the feasibility of [89Zr]pertuzumab imaging to monitor early response to Ado-trastuzumab emtansine (T-DM1) therapy in mice bearing xenografts of HER2-positive breast cancer (BCa). Materials and Methods: Pertuzumab was conjugated to DFO-Bz-NCS and labeled with 89Zr. Mice bearing BT-474 tumors were imaged with [89Zr]pertuzumab and [18F]FDG before and after T-DM1 therapy. Results: Pertuzumab was successfully labeled with 89Zr with a specific activity of 0.740 MBq/µg. Overall [18F]FDG images showed poor delineation of tumors. Using [18F]FDG-PET to measure tumor volume, the volume remained unchanged from 107.6 ± 20.7 mm3 before treatment to 89.87 ± 66.55 mm3 after treatment. In contrast, [89Zr]pertuzumab images showed good delineation of HER2-positive tumors, allowing accurate detection of changes in tumor volume (from 243.80 ± 40.91 mm3 before treatment to 78.4 ± 40.43 mm3 after treatment). Conclusion: [89Zr]pertuzumab may be an imaging probe for monitoring the response of HER2-positive BCa patients to T-DM1 therapy.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Neoplasias da Mama/diagnóstico por imagem , Maitansina/análogos & derivados , Compostos Radiofarmacêuticos/administração & dosagem , Receptor ErbB-2/metabolismo , Trastuzumab/uso terapêutico , Ado-Trastuzumab Emtansina , Animais , Anticorpos Monoclonais Humanizados/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Desferroxamina/análogos & derivados , Desferroxamina/química , Feminino , Humanos , Isotiocianatos/química , Maitansina/uso terapêutico , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Radioisótopos/administração & dosagem , Radioisótopos/química , Compostos Radiofarmacêuticos/química , Receptor ErbB-2/antagonistas & inibidores , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Microtomografia por Raio-X/métodos , Ensaios Antitumorais Modelo de Xenoenxerto , Zircônio/administração & dosagem , Zircônio/química
10.
J Antibiot (Tokyo) ; 71(11): 911-919, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30120394

RESUMO

Recently, a novel nitrous acid biosynthetic pathway composed of two enzymes was discovered to be involved in the biosynthesis of cremeomycin for the formation of its diazo group. In this pathway, CreE oxidizes L-aspartic acid to nitrosuccinic acid and CreD liberates nitrous acid from nitrosuccinic acid. Bioinformatic analysis showed that various actinobacteria have putative secondary metabolite biosynthesis gene clusters containing creE and creD homologs, suggesting that this pathway is widely used for the biosynthesis of various natural products. Here, we focused on creE and creD homologs (BN159_4422 and BN159_4421) in Streptomyces davawensis. In vitro analysis of recombinant BN159_4422 and BN159_4421 proteins showed that these enzymes synthesized nitrous acid from L-aspartic acid. Secondary metabolites produced by this gene cluster were investigated by comparing the metabolic profiles of the wild-type and ΔBN159_4422 strains. When these strains were co-cultured with Tsukamurella pulmonis TP-B0596, three compounds were specifically produced by the wild-type strain. These compounds were identified as novel desferrioxamine derivatives containing either of two unique five-membered heterocyclic ring structures and shown to have iron-binding properties. A putative desferrioxamine biosynthetic gene cluster was found in the S. davawensis genome, and inactivation of a desD homolog (BN159_5485) also abolished the production of these compounds. We propose that these compounds should be synthesized by the modification of desferrioxamine B and a shorter chain analog using nitrous acid produced by the CreE and CreD homologs. This study provides an important insight into the diverse usage of the secondary metabolism-specific nitrous acid biosynthetic pathway in actinomycetes.


Assuntos
Vias Biossintéticas , Desferroxamina/análogos & derivados , Desferroxamina/metabolismo , Ácido Nitroso/metabolismo , Metabolismo Secundário , Streptomyces/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Conjugação Genética , Desferroxamina/química , Escherichia coli , Regulação Bacteriana da Expressão Gênica , Estrutura Molecular , Família Multigênica , Streptomyces/classificação
11.
Chem Commun (Camb) ; 54(70): 9813-9816, 2018 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-30106398

RESUMO

An analogue of the bacterial siderophore desferrioxamine B (DFOB) containing a disulfide motif in the backbone was produced from Streptomyces pilosus cultures supplemented with cystamine. Cystamine competed against native 1,5-diaminopentane during assembly. DFOB-(SS)1[001] and its complexes with Fe(iii) or Ga(iii) were cleaved upon incubation with dithiothreitol. Compounds such as DFOB-(SS)1[001] and its thiol-containing cleavage products could expand antibiotic strategies and Au-S-based nanotechnologies.


Assuntos
Complexos de Coordenação/metabolismo , Desferroxamina/análogos & derivados , Desferroxamina/metabolismo , Dissulfetos/metabolismo , Compostos Férricos/metabolismo , Sideróforos/biossíntese , Cadaverina/metabolismo , Cistamina/metabolismo , Gálio/química , Ferro/química , Streptomyces/química
12.
ACS Chem Biol ; 13(9): 2456-2471, 2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30081629

RESUMO

The siderophore desferrioxamine B (DFOB, 1) native to Streptomyces pilosus is biosynthesized by the DesABCD enzyme cluster. DesA-mediated decarboxylation of l-lysine gives 1,5-diaminopentane (DP) for processing by DesBCD. S. pilosus culture medium was supplemented with rac-1,4-diamino-2-fluorobutane ( rac-FDB) to compete against DP to generate fluorinated analogues of DFOB, as agents of potential clinical interest. LC-MS/MS analysis identified fluorinated analogues of DFOB with one, two, or three DP units (binary notation: 0) exchanged for one (DFOA-F1[001] (2), DFOA-F1[010] (3), DFOA-F1[100] (4)), two (DFOA-F2[011] (5), DFOA-F2[110] (6), DFOA-F2[101] (7)), or three (DFOA-F3[111] (8)) rac-FDB units (binary notation: 1). The two sets of constitutional isomers 2-4 and 5-7 arose from the position of the substrates in the N-acetyl, internal, or amine-containing regions of the DFOB trimer. N-Acetylated fluorinated DFOB analogues were formed where the rac-FDB substrate was positioned in the amine region ( e.g., N-Ac-DFOA-F1[001] (2a)). Other analogues contained two hydroxamic acid groups and three amide bonds. Experiments using rac-FDB, R-FDB, or S-FDB showed a similar species profile between rac-FDB and R-FDB. These data are consistent with the following. (i) DesB can act on rac-FDB. (ii) DesC can act directly on rac-FDB. (iii) The products of DesBC or DesC catalysis of rac-FDB can undergo a second round of DesC catalysis at the free amine. (iv) DesD catalysis of these products gives N, N'-diacetylated compounds. (v) A minimum of two hydroxamic acid groups is required to form a viable DesD-substrate(s) precomplex. (vi) One or more DesBCD-catalyzed steps in DFOB biosynthesis is enantioselective. This work has provided a potential path to access fluorinated analogues of DFOB and new insight into its biosynthesis.


Assuntos
Desferroxamina/análogos & derivados , Desferroxamina/metabolismo , Streptomyces/enzimologia , Vias Biossintéticas , Halogenação , Sideróforos/química , Sideróforos/metabolismo , Estereoisomerismo , Streptomyces/química , Streptomyces/metabolismo , Especificidade por Substrato , Espectrometria de Massas em Tandem
13.
Bioelectrochemistry ; 122: 149-157, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29631207

RESUMO

A new strategy is developed for construction of the mixed molecular nanostructures from folic acid (FOA), a targeting agent, and deferrioxamoine-Ga(III), (DFO-Ga(III)), a theranostic agent, on gold-mercaptopropionic acid surface, Au-MPA. The strategy is focused to achieve a system in which all the active constituents of FOA; i.e., pteridine rings, p-aminobenzoeic acid, and the glutamic acid, having high affinity for folate receptor overexpressed on cancer cells; remain unreacted in adjacent to DFO-Ga(III), Au-MPA-[DFO-Ga(III)]‖-[FOA]. For this purpose, the NH2 groups of FOA and DFO-Ga(III) were attached covalently and separately to COOH of Au-MPA surface allowing all the active groups of FOA to be available for drug delivery purposes. The data obtained through several electrochemical and surface analysis techniques, supported successful construction of the designed mixed molecular nanostructures system. In addition, the results showed that the system is stable, and Ga(III) ion does not leave DFO-Ga(III) complex. The prepared surface was successfully tested for capturing of the breast cancer cells 4T1 as a model. The measurements showed a rapid uptake kinetics (t1/2 of ~6.0min) and efficient accessibility of the system by the cancer cells; the Rct was significantly increased in the presence of 4T1 cells compared with blank PBS (ΔRct ~420kΩ).


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/tratamento farmacológico , Desferroxamina/uso terapêutico , Ácido Fólico/uso terapêutico , Nanoestruturas/uso terapêutico , Nanomedicina Teranóstica , Animais , Antineoplásicos/química , Mama/efeitos dos fármacos , Mama/patologia , Desferroxamina/análogos & derivados , Sistemas de Liberação de Medicamentos , Feminino , Ácido Fólico/química , Gálio/química , Gálio/uso terapêutico , Ouro/química , Camundongos , Nanoestruturas/química , Nanotecnologia
14.
Biometals ; 30(5): 709-718, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28770399

RESUMO

Cell-impermeant iron chelator desferrioxamine (DFO) can have access to organelles if appended to suitable vectors. Mitochondria are important targets for the treatment of iron overload-related neurodegenerative diseases. Triphenylphosphonium (TPP) is a delocalized lipophilic cation used to ferry molecules to mitochondria. Here we report the synthesis and characterization of the conjugate TPP-DFO as a mitochondrial iron chelator. TPP-DFO maintained both a high affinity for iron and the antioxidant activity when compared to parent DFO. TPP-DFO was less toxic than TPP alone to A2780 cells (IC50 = 135.60 ± 1.08 and 4.34 ± 1.06 µmol L-1, respectively) and its native fluorescence was used to assess its mitochondrial localization (Rr = +0.56). These results suggest that TPP-DFO could be an interesting alternative for the treatment of mitochondrial iron overload e.g. in Friedreich's ataxia.


Assuntos
Desferroxamina/farmacologia , Quelantes de Ferro/farmacologia , Mitocôndrias/efeitos dos fármacos , Imagem Óptica/métodos , Compostos Organofosforados/química , Ligação Competitiva , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desferroxamina/análogos & derivados , Desferroxamina/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Fluoresceínas/metabolismo , Humanos , Quelantes de Ferro/síntese química , Quelantes de Ferro/metabolismo , Cinética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura
15.
Mol Pharm ; 14(8): 2831-2842, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28665620

RESUMO

New bifunctional hexa- and octadentate analogues of the hydroxamate-containing siderophore desferrichrome (DFC) have been synthesized and evaluated as 89Zr-chelating agents for immunoPET applications. The in vitro and in vivo inertness of these new ligands, Orn3-hx (hexadentate) and Orn-4hx derivatives (octadentate), was compared to the gold standard hexadentate, hydroxamate-containing chelator for 89Zr desferrioxamine (DFO). Density functional theory was employed to model the geometries of the resulting Zr(IV) complexes and to predict their relative stabilities as follows: Zr(Orn4-hx) > Zr(DFC) > Zr(Orn3-hx). Transchelation challenge experiments of the corresponding radiochemical complexes with excess ethylenediaminetetraacetate (EDTA) indicated complex stability in accordance with DFT calculations. Radiolabeling of these ligands with 89Zr was quantitative (0.25 µmol of ligand, pH 7.4, room temperature, 20 min). For antibody conjugation, the isothiocyanate (NCS) functional group was introduced to the N terminus of Orn3-hx and Orn-4hx. An additional trifunctional derivative that bears a silicon-rhodamine fluorophore on the C-terminus and NCS on the N terminus was also furnished. As proof of concept, all NCS derivatives were conjugated to the HER2-targeting antibody, trastuzumab. Radiolabeling of immunoconjugates with 89Zr was accomplished with radiochemical yields of 16 ± 2% to 95 ± 2%. These constructs were administered to naive mice (male, C57BL/6J, n = 4) to assess in vivo inertness, which is inversely correlated with uptake of 89Zr in bone, after 96 h circulation time. We found bone uptake to range from 7.0 ± 2.2 to 10.7 ± 1.3% ID/g, values that compare well to the corresponding DFO conjugate (7.1 ± 0.8% ID/g). In conclusion, we have rationally designed linear, bifunctional and trifunctional desferrichrome analogues suitable for the mild and inert radiolabeling of antibodies with the radionuclide 89Zr.


Assuntos
Quelantes/química , Desferroxamina/análogos & derivados , Radioisótopos/química , Compostos Radiofarmacêuticos/química , Trastuzumab/química , Zircônio/química , Animais , Ácido Edético/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Temperatura
16.
Metallomics ; 9(7): 852-864, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28466891

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterised by the death of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the brain and formation of α-synuclein-containing intracellular inclusions. Excess intraneuronal iron in the SNpc increases reactive oxygen species (ROS), which identifies removing iron as a possible therapeutic strategy. Desferrioxamine B (DFOB, 1) is an iron chelator produced by bacteria. Its high Fe(iii) affinity, water solubility and low chronic toxicity is useful in removing iron accumulated in plasma from patients with transfusion-dependent blood disorders. Here, lipophilic analogues of DFOB with increased potential to cross the blood-brain barrier (BBB) have been prepared by conjugating ancillary compounds onto the amine terminus. The ancillary compounds included the antioxidants rac-6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (rac-trolox, rac-TLX (a truncated vitamin E variant)), R-TLX, S-TLX, methylated derivatives of 3-(6-hydroxy-2-methylchroman-2-yl)propionic acid (α-CEHC, γ-CEHC, δ-CEHC), or 4-(5-hydroxy-3-methyl-1H-pyrazol-1-yl)benzoic acid (carboxylic acid derivative of edaravone, EDA). Compounds 2-8 could have dual function in attenuating ROS by chelating Fe(iii) and via the antioxidant ancillary group. A conjugate between DFOB and an ancillary unit without antioxidant properties (3,5-dimethyladamantane-1-carboxylic acid (AdAdMe)) was included (9). Compounds 2-9 were more lipophilic (log P -0.05 to 3.39) than DFOB (log P -2.62) and showed an average plasma protein binding 6 times greater than DFOB. The ABTS˙+ radical assay indicated 2-8 had antioxidant activity ascribable to the ancillary fragment. Administration of 2 and 9 in the mouse model of PD using the neurotoxin prodrug 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which recapitulates elevated iron of human PD, resulted in significant neuronal protection (p < 0.05; up to 89% of that in non-lesioned control animals), demonstrating the neuroprotective potential of these compounds for PD.


Assuntos
Desferroxamina/análogos & derivados , Desferroxamina/uso terapêutico , Ferro/efeitos adversos , Degeneração Neural/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Benzotiazóis/metabolismo , Proteínas Sanguíneas/metabolismo , Desferroxamina/química , Desferroxamina/farmacologia , Modelos Animais de Doenças , Quelantes de Ferro/química , Camundongos , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ácidos Sulfônicos/metabolismo
17.
ACS Infect Dis ; 3(7): 542-553, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28505405

RESUMO

Staphylococcus aureus produces a cocktail of metallophores (staphylopine, staphyloferrin A, and staphyloferrin B) to scavenge transition metals during infection of a host. In addition, S. aureus displays the extracellular surface lipoproteins FhuD1 and FhuD2 along with the ABC transporter complex FhuCBG to facilitate the use of hydroxamate xenosiderophores such as desferrioxamine B (DFOB) for iron acquisition. DFOB is used as a chelation therapy to treat human iron overload diseases and has been linked to an increased risk of S. aureus infections. We used a panel of synthetic DFOB analogs and a FhuD2-selective trihydroxamate sideromycin to probe xenosiderophore utilization in S. aureus and establish structure-activity relationships for Fe(III) binding, FhuD2 binding, S. aureus growth promotion, and competition for S. aureus cell entry. Fe(III) binding assays and FhuD2 intrinsic fluorescence quenching experiments revealed that diverse chemical modifications of the terminal ends of linear ferrioxamine siderophores influences Fe(III) affinity but not FhuD2 binding. Siderophore-sideromycin competition assays and xenosiderophore growth promotion assays revealed that S. aureus SG511 and ATCC 11632 can distinguish between competing siderophores based exclusively on net charge of the siderophore-Fe(III) complex. Our work provides a roadmap for tuning hydroxamate xenosiderophore scaffolds to suppress (net negative charge) or enhance (net positive or neutral charge) uptake by S. aureus for applications in metal chelation therapy and siderophore-mediated antibiotic delivery, respectively.


Assuntos
Desferroxamina/metabolismo , Ferro/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Staphylococcus aureus Resistente à Meticilina/metabolismo , Proteínas Periplásmicas de Ligação/metabolismo , Sideróforos/metabolismo , Ligação Competitiva , Cátions , Desferroxamina/análogos & derivados , Desferroxamina/farmacologia , Expressão Gênica , Cinética , Proteínas de Membrana Transportadoras/genética , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/genética , Modelos Moleculares , Proteínas Periplásmicas de Ligação/genética , Ligação Proteica , Sideróforos/síntese química , Sideróforos/farmacologia , Eletricidade Estática , Relação Estrutura-Atividade
18.
Bioorg Med Chem Lett ; 27(8): 1698-1704, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28285915

RESUMO

The death of dopaminergic neurons is a major pathological hallmark of Parkinson's disease (PD). Elevated iron within the substantia nigra of the PD brain is thought to catalyze this neuronal death through hydroxyl radical-derived oxidative damage. Removing this excess iron presents a potential therapeutic strategy for PD. Seventeen derivatives of the non-toxic iron chelator desferrioxamine B (DFOB) were prepared by the conjugation of adamantyl- (1-4, 8-12), deconstructed adamantyl units (5-7), norborna(e)ne- (13-16) or bicyclo[2.2.2]octane-based (17) ancillary fragments to the terminal amine group. The range of experimental logP values of 1-17 (logP=0.15-2.82) was greater than water soluble DFOB (logP -2.29), with the increased hydrophobicity designed to improve cell membrane carriage to facilitate intracellular iron sequestration. The first activity screen showed compounds with methyl-substituted adamantyl (1-3), noradamantyl (5), or 1-pentylbicyclo[2.2.2]octane (17) ancillary groups significantly rescued iron-mediated oxidative stress in confluent PD-relevant SK-N-BE2-M17 neuroblastoma cells (M17 cells) exposed to 1,1'-dimethyl-4,4'-bipyridinium (paraquat, PQ) or H2O2. The second dose-dependence screen ranked 1-3 and 17 as the top candidates (EC50 ∼10µM) in the rescue of PQ-treated M17 cells. The ancillary fragments of 1-3 and 17 clustered in a region defined by a close-to-zero dipole moment, logP values of 2-2.8 and a surface area:volume ratio of 0.60-0.61. Results of iron leaching studies indicate that the compounds may be operating via mechanisms beyond solely removing intracellular iron. The DFOB conjugates with methyl-substituted adamantyl ancillary groups (1-3) were the top and most consistent performers in this class of compound designed for PD.


Assuntos
Desferroxamina/análogos & derivados , Desferroxamina/farmacologia , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Ferro/metabolismo , Doença de Parkinson/tratamento farmacológico , Astrócitos/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Doença de Parkinson/metabolismo , Compostos Policíclicos/química , Compostos Policíclicos/farmacologia
19.
Molecules ; 21(9)2016 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-27618884

RESUMO

Activation of silent biosynthetic gene clusters in Streptomyces bacteria via overexpression of cluster-specific regulatory genes is a promising strategy for the discovery of novel bioactive secondary metabolites. This approach was used in an attempt to activate a cryptic gene cluster in a marine sponge-derived Streptomyces albus PVA94-07 presumably governing the biosynthesis of peptide-based secondary metabolites. While no new peptide-based metabolites were detected in the recombinant strain, it was shown to produce at least four new analogues of deferoxamine with additional acyl and sugar moieties, for which chemical structures were fully elucidated. Biological activity tests of two of the new deferoxamine analogues revealed weak activity against Escherichia coli. The gene knockout experiment in the gene cluster targeted for activation, as well as overexpression of certain genes from this cluster did not have an effect on the production of these compounds by the strain overexpressing the regulator. It seems plausible that the production of such compounds is a response to stress imposed by the production of an as-yet unidentified metabolite specified by the cryptic cluster.


Assuntos
Antibacterianos , Organismos Aquáticos/microbiologia , Desferroxamina , Escherichia coli/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica/fisiologia , Poríferos/microbiologia , Streptomyces/metabolismo , Animais , Antibacterianos/biossíntese , Antibacterianos/farmacologia , Desferroxamina/análogos & derivados , Desferroxamina/metabolismo , Desferroxamina/farmacologia
20.
J Nucl Med ; 57(6): 974-80, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26940768

RESUMO

UNLABELLED: Subtype A2 of the erythropoietin-producing hepatocellular tyrosine kinase (EphA2) cell surface receptor is expressed in a range of epithelial cancers. This study evaluated the molecular imaging of EphA2 expression in vivo in mouse tumor models using SPECT/MR and PET/MR and a humanized anti-EphA2 antibody, DS-8895a. METHODS: DS-8895a was labeled with (111)In, (125)I, and (89)Zr and assessed for radiochemical purity, immunoreactivity (Lindmo analysis), antigen-binding affinity (Scatchard analysis), and serum stability in vitro. In vivo biodistribution, imaging, and pharmacokinetic studies were performed with SPECT/MR and PET/MR. A dose-escalation study was also performed to determine EphA2 receptor saturability through tissue and imaging quantitative analysis. RESULTS: All conjugates demonstrated good serum stability and specific binding to EphA2-expressing cells in vitro. In vivo biodistribution studies showed high uptake of (111)In-CHX-A″-DTPA-DS-8895a and (89)Zr-Df-Bz-NCS-DS-8895a in EphA2-expressing xenograft models, with no specific uptake in normal tissues. In comparison, retention of (125)I-DS-8895a in tumors was lower because of internalization of the radioconjugate and dehalogenation. These results were confirmed by SPECT/MR and PET/MR. EphA2 receptor saturation was observed at the 30 mg/kg dose. CONCLUSION: Molecular imaging of tumor uptake of DS-8895a allows noninvasive measurement of EphA2 expression in tumors in vivo and determination of receptor saturation. (89)Zr-Df-Bz-NCS-DS-8895a is suited for human bioimaging trials on the basis of superior imaging characteristics and will inform DS-8895a dose assessment and patient response evaluation in clinical trials.


Assuntos
Anticorpos Monoclonais Humanizados/química , Transformação Celular Neoplásica , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos , Receptor EphA2/metabolismo , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Zircônio/química , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacocinética , Linhagem Celular Tumoral , Desferroxamina/análogos & derivados , Desferroxamina/química , Feminino , Humanos , Isotiocianatos/química , Camundongos , Ácido Pentético/química , Controle de Qualidade , Receptor EphA2/imunologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA