Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Langmuir ; 39(18): 6539-6547, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37127842

RESUMEN

Attaching polymers, especially polyethylene glycol (PEG), to protein drugs has emerged as a successful strategy to prolong circulation time in the bloodstream. The hypothesis is that the flexible chain wobbles on the protein's surface, thus resisting potential nonspecific adsorption. Such a theoretical framework may be challenged when a helical polyglutamate is used to conjugate with target proteins. In this study, we investigated the structure-activity relationships of polyglutamate-interferon conjugates P(EG3Glu)-IFN using molecular simulations. Our results show that the local crowding effect induced by oligoethylene glycols (i.e., EG3) is the primary driving force for helix formation in P(EG3Glu), and its helicity can be effectively increased by reducing the free volume of the two termini. Furthermore, it was found that the steric hindrance induced by IFN is not conductive to the helicity of P(EG3Glu) but contributes to its dominant orientation relative to interferon. The orientation of IFN relative to the helical P(EG3Glu) can help to protect the protein drug from neutralizing antibodies while maintaining its bioactivity. These findings suggest that the helical structure and its orientation are critical factors to consider when updating the theoretical framework for protein-polymer conjugates.


Asunto(s)
Interferones , Ácido Poliglutámico , Interferones/química , Polietilenglicoles/química , Polímeros/química , Proteínas
2.
Nucleic Acids Res ; 49(19): 11211-11223, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34614173

RESUMEN

Binding and unbinding of transcription factors to DNA are kinetically controlled to regulate the transcriptional outcome. Control of the release of the transcription factor NF-κB from DNA is achieved through accelerated dissociation by the inhibitor protein IκBα. Using single-molecule FRET, we observed a continuum of conformations of NF-κB in free and DNA-bound states interconverting on the subseconds to minutes timescale, comparable to in vivo binding on the seconds timescale, suggesting that structural dynamics directly control binding kinetics. Much of the DNA-bound NF-κB is partially bound, allowing IκBα invasion to facilitate DNA dissociation. IκBα induces a locked conformation where the DNA-binding domains of NF-κB are too far apart to bind DNA, whereas a loss-of-function IκBα mutant retains the NF-κB conformational ensemble. Overall, our results suggest a novel mechanism with a continuum of binding modes for controlling association and dissociation of transcription factors.


Asunto(s)
ADN/genética , Interferones/genética , Inhibidor NF-kappaB alfa/genética , Factor de Transcripción ReIA/genética , Transcripción Genética , Animales , Avidina/química , Sitios de Unión , Biotina/química , ADN/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Interferones/química , Interferones/metabolismo , Secuencias Invertidas Repetidas , Ratones , Simulación de Dinámica Molecular , Inhibidor NF-kappaB alfa/química , Inhibidor NF-kappaB alfa/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Imagen Individual de Molécula/métodos , Factor de Transcripción ReIA/química , Factor de Transcripción ReIA/metabolismo
3.
ACS Chem Biol ; 16(5): 844-856, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33887136

RESUMEN

Interferon-induced transmembrane proteins (IFITMs) are S-palmitoylated proteins in vertebrates that restrict a diverse range of viruses. S-palmitoylated IFITM3 in particular engages incoming virus particles, prevents their cytoplasmic entry, and accelerates their lysosomal clearance by host cells. However, how S-palmitoylation modulates the structure and biophysical characteristics of IFITM3 to promote its antiviral activity remains unclear. To investigate how site-specific S-palmitoylation controls IFITM3 antiviral activity, we employed computational, chemical, and biophysical approaches to demonstrate that site-specific lipidation of cysteine 72 enhances the antiviral activity of IFITM3 by modulating its conformation and interaction with lipid membranes. Collectively, our results demonstrate that site-specific S-palmitoylation of IFITM3 directly alters its biophysical properties and activity in cells to prevent virus infection.


Asunto(s)
Antivirales/química , Membrana Celular/metabolismo , Interferones/química , Lípidos/química , Proteínas de la Membrana/metabolismo , Proteínas de Unión al ARN/metabolismo , Secuencia de Aminoácidos , Antivirales/farmacología , Sitios de Unión , Membrana Celular/ultraestructura , Biología Computacional , Diseño de Fármacos , Humanos , Interferones/farmacología , Lipoilación , Lisosomas/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Transducción de Señal
4.
Front Immunol ; 11: 606489, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281831

RESUMEN

Interferons (IFNs) are a family of cytokines with the unique ability to induce cell intrinsic programs that enhance resistance to viral infection. Induction of an antiviral state at the cell, tissue, organ, and organismal level is performed by three distinct IFN families, designated as Type-I, Type-II, and Type-III IFNs. Overall, there are 21 human IFNs, (16 type-I, 12 IFNαs, IFNß, IFNϵ, IFNκ, and IFNω; 1 type-II, IFNγ; and 4 type-III, IFNλ1, IFNλ2, IFNλ3, and IFNλ4), that induce pleotropic cellular activities essential for innate and adaptive immune responses against virus and other pathogens. IFN signaling is initiated by binding to distinct heterodimeric receptor complexes. The three-dimensional structures of the type-I (IFNα/IFNAR1/IFNAR2), type-II (IFNγ/IFNGR1/IFNGR2), and type-III (IFNλ3/IFNλR1/IL10R2) signaling complexes have been determined. Here, we highlight similar and unique features of the IFNs, their cell surface complexes and discuss their role in inducing downstream IFN signaling responses.


Asunto(s)
Interferones/metabolismo , Receptores de Interferón/metabolismo , Transducción de Señal , Animales , Humanos , Interferones/química , Ligandos , Ratones , Modelos Moleculares , Conformación Proteica , Receptores de Interferón/química , Especificidad de la Especie , Relación Estructura-Actividad
5.
Mar Drugs ; 18(6)2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32549252

RESUMEN

Chitosan nanoparticles (CS-NPs) are under increasing investigation for the delivery of therapeutic proteins, such as vaccines, interferons, and biologics. A large number of studies have been taken on the characteristics of CS-NPs, and very few of these studies have focused on the microstructure of protein-loaded NPs. In this study, we prepared the CS-NPs by an ionic gelation method, and bovine serum albumin (BSA) was used as a model protein. Dynamic high pressure microfluidization (DHPM) was utilized to post-treat the nanoparticles so as to improve the uniformity, repeatability and controllability. The BSA-loaded NPs were then characterized for particle size, Zeta potential, morphology, encapsulation efficiency (EE), loading capacity (LC), and subsequent release kinetics. To improve the long-term stability of NPs, trehalose, glucose, sucrose, and mannitol were selected respectively to investigate the performance as a cryoprotectant. Furthermore, trehalose was used to obtain re-dispersible lyophilized NPs that can significantly reduce the dosage of cryoprotectants. Multiple spectroscopic techniques were used to characterize BSA-loaded NPs, in order to explain the release process of the NPs in vitro. The experimental results indicated that CS and Tripolyphosphate pentasodium (TPP) spontaneously formed the basic skeleton of the NPs through electrostatic interactions. BSA was incorporated in the basic skeleton, adsorbed on the surface of the NPs (some of which were inlaid on the NPs), without any change in structure and function. The release profiles of the NPs showed high consistency with the multispectral results.


Asunto(s)
Quitosano/química , Crioprotectores/química , Portadores de Fármacos/química , Nanopartículas/química , Productos Biológicos/administración & dosificación , Productos Biológicos/química , Frío/efectos adversos , Composición de Medicamentos/métodos , Liberación de Fármacos , Estabilidad de Medicamentos , Almacenaje de Medicamentos/métodos , Interferones/administración & dosificación , Interferones/química , Tamaño de la Partícula , Estabilidad Proteica , Albúmina Sérica Bovina/administración & dosificación , Albúmina Sérica Bovina/química , Albúmina Sérica Bovina/farmacocinética , Vacunas/administración & dosificación , Vacunas/química
6.
Chembiochem ; 21(18): 2595-2598, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32346955

RESUMEN

Cyclic dinucleotides (CDNs) trigger the innate immune response in eukaryotic cells through the stimulator of interferon genes (STING) signaling pathway. To decipher this complex cellular process, a better correlation between structure and downstream function is required. Herein, we report the design and immunostimulatory effect of a novel group of c-di-GMP analogues. By employing an "atomic mutagenesis" strategy, changing one atom at a time, a class of gradually modified CDNs was prepared. These c-di-GMP analogues induce type-I interferon (IFN) production, with some being more potent than c-di-GMP, their native archetype. This study demonstrates that CDN analogues bearing modified nucleobases are able to tune the innate immune response in eukaryotic cells.


Asunto(s)
GMP Cíclico/inmunología , Interferones/inmunología , Nucleótidos Cíclicos/inmunología , GMP Cíclico/análogos & derivados , GMP Cíclico/química , Inmunidad Innata , Interferones/química , Interferones/genética , Nucleótidos Cíclicos/química , Transducción de Señal/genética , Transducción de Señal/inmunología
7.
Bioorg Med Chem Lett ; 30(2): 126819, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31780305

RESUMEN

Twenty-nine nucleoside analogues have been synthesized and evaluated in a cell based assay for their ability to activate the human Stimulator of Interferon Genes (hSTING), a key protein of the innate immune defense. Some 6-O-alkyl nucleoside analogues activate hSTING without associated cytotoxicity. SAR and combination studies were performed to decipher possible activation mechanism. The described nucleoside hSTING activators represent first-in-class modulators of the innate immune defense; a highly relevant target for antiviral, antibacterial, anticancer or Alzheimer's disease treatments and may present advantages over other types of hSTING activators.


Asunto(s)
Interferones/química , Purinas/química , Humanos , Estructura Molecular , Relación Estructura-Actividad
8.
Viruses ; 11(11)2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31717991

RESUMEN

Hepatitis E virus (HEV) causes predominantly acute and self-limiting hepatitis. However, in HEV-infected pregnant women, the case fatality rate because of fulminant hepatitis can be up to 30%. HEV infection is zoonotic for some genotypes. The HEV genome contains three open reading frames: ORF1 encodes the non-structural polyprotein involved in viral RNA replication; ORF2 encodes the capsid protein; ORF3 encodes a small multifunctional protein. Interferons (IFNs) play a significant role in the early stage of the host antiviral response. In this study, we discovered that the capsid protein antagonizes IFN induction. Mechanistically, the capsid protein blocked the phosphorylation of IFN regulatory factor 3 (IRF3) via interaction with the multiprotein complex consisting of mitochondrial antiviral-signaling protein (MAVS), TANK-binding kinase 1 (TBK1), and IRF3. The N-terminal domain of the capsid protein was found to be responsible for the inhibition of IRF3 activation. Further study showed that the arginine-rich-motif in the N-terminal domain is indispensable for the inhibition as mutations of any of the arginine residues abolished the blockage of IRF3 phosphorylation. These results provide further insight into HEV interference with the host innate immunity.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus de la Hepatitis E/fisiología , Hepatitis E/metabolismo , Hepatitis E/virología , Interacciones Huésped-Patógeno , Interferones/biosíntesis , Dominios y Motivos de Interacción de Proteínas , Proteínas de la Cápside/genética , Genotipo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferones/química , Modelos Biológicos , Factores de Crecimiento Nervioso , Fosforilación , Poli I-C/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo
9.
PLoS One ; 14(9): e0213880, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31490936

RESUMEN

The current study highlights, for the first time, cloning, overexpression and purification of the novel interferon epsilon (IFNƐ), from the Arabian camel Camelus dromedaries. The study then assesses the cytotoxicity of IFNε against two human breast cancer cell lines MDA-MB-231 and MCF-7. Full-length cDNA encoding interferon epsilon (IFNε) was isolated and cloned from the liver of the Arabian camel, C. dromedarius using reverse transcription-polymerase chain reaction. The sequence analysis of the camel IFNε cDNA showed a 582-bp open reading frame encoding a protein of 193 amino acids with an estimated molecular weight of 21.230 kDa. A BLAST search analysis revealed that the C. dromedarius IFNε shared high sequence identity with the IFN genes of other species, such as Camelus ferus, Vicugna pacos, and Homo sapiens. Expression of the camel IFNε cDNA in Escherichia coli gave a fusion protein band of 24.97 kDa after induction with either isopropyl ß-D-1-thiogalactopyranoside or lactose for 5 h. Recombinant IFNε protein was overexpressed in the form of inclusion bodies that were easily solubilized and refolded using SDS and KCl. The solubilized inclusion bodies were purified to apparent homogeneity using nickel affinity chromatography. We examined the effect of IFNε on two breast cancer cell lines MDA-MB-231 and MCF-7. In both cell lines, IFNε inhibited cell survival in a dose dependent manner as observed by MTT assay, morphological changes and apoptosis assay. Caspase-3 expression level was found to be increased in MDA-MB-231 treated cells as compared to untreated cells.


Asunto(s)
Camelus/genética , Interferones/genética , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Clonación Molecular , Humanos , Interferones/química , Interferones/metabolismo , Interferones/farmacología , Células MCF-7 , Masculino , Pliegue de Proteína , Homología de Secuencia
10.
Biomacromolecules ; 20(8): 3000-3008, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31310511

RESUMEN

Protein-polymer conjugation is a clinically validated approach to enhanced pharmacokinetic properties. However, the permanent attachment of polymers often leads to irreversibly reduced protein bioactivity and poor tissue penetration. As such, the use of protein-polymer conjugates for solid tumors remains elusive. Herein, we report a simple strategy using enzyme-activatable and size-shrinkable protein-polypeptide conjugates to overcome this clinical challenge. Briefly, a matrix metalloproteinase (MMP)-responsive peptide sequence is introduced between a therapeutic protein interferon (IFN) and a synthetic polypeptide P(EG3Glu)20. The resulting site-specific MMP-responsive conjugate, denoted as PEP20-M-IFN, can, therefore, release the attached P(EG3Glu)20 to achieve both protein activation and deep penetration into the tumor microenvironment (TME). Compared to a similarly produced nonresponsive analogue conjugate PEP20-IFN, our results find PEP20-M-IFN to show higher bioactivity in vitro, improved tumor retention, and deeper penetration in a MMP2-dependent manner. Moreover, systemic administration of PEP20-M-IFN shows outstanding antitumor efficacy in both OVCAR3 and SKOV3 ovarian tumor models in mice. This work highlights the releasable PEPylation strategy for protein drug potentiation at the TME and opens up new opportunities in clinics for the treatment of malignant solid tumors.


Asunto(s)
Activadores de Enzimas/farmacología , Interferones/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Nanopartículas/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Polímeros/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Animales , Antivirales/química , Antivirales/metabolismo , Apoptosis , Proliferación Celular , Activadores de Enzimas/química , Femenino , Humanos , Interferones/química , Metaloproteinasa 2 de la Matriz/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Polímeros/química , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 6): 455-460, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-31204693

RESUMEN

Interferon-inducible protein 204 (p204) binds to microbial DNA to elicit inflammatory responses and induce interferon production. p204 also modulates cell proliferation and differentiation by regulating various transcription factors. The C-terminal HIN domains in p204 are believed to be responsible for DNA binding, but the binding mode is not fully understood. The DNA-binding affinity of the p204 HIN1 domain has been characterized and its crystal structure has been determined, providing insight into its interaction with DNA. Surface-charge distribution together with sequence alignment suggests that the p204 HIN domain uses its L12 and L45 loops for DNA binding.


Asunto(s)
Interferones/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Interferones/química , Ratones , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
12.
Proc Natl Acad Sci U S A ; 116(6): 2103-2111, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30655338

RESUMEN

Cells of all mammals recognize double-stranded RNA (dsRNA) as a foreign material. In response, they release interferons (IFNs) and activate a ubiquitously expressed pseudokinase/endoribonuclease RNase L. RNase L executes regulated RNA decay and halts global translation. Here, we developed a biosensor for 2',5'-oligoadenylate (2-5A), the natural activator of RNase L. Using this biosensor, we found that 2-5A was acutely synthesized by cells in response to dsRNA sensing, which immediately triggered cellular RNA cleavage by RNase L and arrested host protein synthesis. However, translation-arrested cells still transcribed IFN-stimulated genes and secreted IFNs of types I and III (IFN-ß and IFN-λ). Our data suggest that IFNs escape from the action of RNase L on translation. We propose that the 2-5A/RNase L pathway serves to rapidly and accurately suppress basal protein synthesis, preserving privileged production of defense proteins of the innate immune system.


Asunto(s)
Técnicas Biosensibles , Endorribonucleasas/química , Interferón beta/química , Interferones/química , Biosíntesis de Proteínas , Línea Celular , Endorribonucleasas/metabolismo , Humanos , Interferón beta/metabolismo , Interferones/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Relación Estructura-Actividad
13.
Cancer Res ; 78(21): 6308-6319, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30224374

RESUMEN

Surgeons have unique in situ access to tumors enabling them to apply immunotherapies to resection margins as a means to prevent local recurrence. Here, we developed a surgical approach to deliver stimulator of interferon genes (STING) ligands to the site of a purposeful partial tumor resection using a gel-based biomaterial. In a range of head and neck squamous cell carcinoma (HNSCC) murine tumor models, we demonstrate that although control-treated tumors recur locally, tumors treated with STING-loaded biomaterials are cured. The mechanism of tumor control required activation of STING and induction of type I IFN in host cells, not cancer cells, and resulted in CD8 T-cell-mediated cure of residual cancer cells. In addition, we used a novel tumor explant assay to screen individual murine and human HNSCC tumor responses to therapies ex vivo We then utilized this information to personalize the biomaterial and immunotherapy applied to previously unresponsive tumors in mice. These data demonstrate that explant assays identify the diversity of tumor-specific responses to STING ligands and establish the utility of the explant assay to personalize immunotherapies according to the local response.Significance: Delivery of immunotherapy directly to resection sites via a gel-based biomaterial prevents locoregional recurrence of head and neck squamous cell carcinoma. Cancer Res; 78(21); 6308-19. ©2018 AACR.


Asunto(s)
Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia/métodos , Interferones/química , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Animales , Materiales Biocompatibles/química , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/cirugía , Humanos , Ligandos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Recurrencia Local de Neoplasia , Trasplante de Neoplasias , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/cirugía , Cicatrización de Heridas
14.
Org Biomol Chem ; 16(28): 5097-5101, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29972388

RESUMEN

A counterintuitive freezing-induced peptide ligation was discovered during the total synthesis of human interferon-ε (hIFN-ε) which blocks HIV infection through unique mechanisms. The successful synthesis of hIFN-ε (187 amino acids) in this research laid the foundation for related anti-AIDS drug development. Moreover, alanine mutation based on sequence alignment to solve the maldistribution of the ligation site and freezing-induced dominant conformation that facilitates peptide ligation are expected to be helpful for the synthesis of macrobiomolecules.


Asunto(s)
Descubrimiento de Drogas , Congelación , Interferones/síntesis química , Interferones/metabolismo , Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Interferones/química , Modelos Moleculares , Conformación Proteica
15.
Bioconjug Chem ; 29(7): 2232-2238, 2018 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-29863329

RESUMEN

The performance of many therapeutic proteins, including human interferon-α2b (IFN), is often impeded by their intrinsic instability to protease, poor pharmacokinetics, and strong immunity. Although PEGylation has been an effective approach to improve the pharmacokinetics of many proteins, a few noticeable limitations have aroused vast research efforts in seeking alternatives to PEG for bioconjugation. Herein, we report our investigation on the use of polysarcosine (PSar), a nonionic and hydrophilic polypeptoid, for IFN modification. The site-specific conjugate PSar-IFN, generated by native chemical ligation in high yield, is systematically compared with a similarly produced PEG-interferon conjugate (PEG-IFN) to evaluate the in vitro and in vivo behaviors. PSar is found to show comparable ability in stabilizing IFN from protease digestion in vitro and prolonging the circulation half-life in vivo. Interestingly, PSar-IFN retains more activity in vitro and accumulates more in the tumor sites upon systemic administration than PEG-IFN. Most importantly, PSar-IFN is significantly more potent in inhibiting tumor growth and elicits considerably less anti-IFN antibodies in mouse than PEG-IFN. Together, our results demonstrate for the first time that PSar is an outstanding candidate for therapeutic protein conjugation. Considering the low toxicity, biodegradability, and excellent stealth effect of PSar, this study suggests that such polypeptoids hold enormous potential for many biomedical applications including protein delivery, colloidal stabilization, and nanomedicine.


Asunto(s)
Péptidos/química , Proteínas/química , Sarcosina/análogos & derivados , Animales , Formación de Anticuerpos , Semivida , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interferones/química , Interferones/inmunología , Interferones/uso terapéutico , Ratones , Neoplasias/tratamiento farmacológico , Polietilenglicoles , Proteínas/farmacocinética , Proteínas/uso terapéutico , Sarcosina/química
16.
J Control Release ; 272: 17-28, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29305921

RESUMEN

Therapeutic proteins can facilitate the targeting and treatment of lymphatic diseases (such as cancer metastases, infections and inflammatory diseases) since they are cleared via the lymphatics following interstitial (SC or IM) administration. However, therapeutic proteins are often administered intravenously (IV). Recently therapeutic proteins have been found to access the thoracic lymph in surprisingly high quantities after IV administration. The aim of this study was to determine, for the first time, the major sites of thoracic lymph access of therapeutic proteins, and the protein properties that enhance lymph access, after IV administration. In order to achieve this, novel methods were developed or optimized to collect hepatic, mesenteric or thoracic lymph from male SD rats. Four different sized PEGylated or non-PEGylated therapeutic proteins (native interferon α2b (IFN, 19kDa), PEGylated interferon α2b (IFN-PEG12, 31kDa), PEGylated interferon α2a (IFN-PEG40, 60kDa) or trastuzumab (150kDa)) were then administered via short IV infusion, and plasma and lymph concentrations of the proteins determined via ELISA. The recovery of the therapeutic proteins in the thoracic lymph duct, which collects lymph from most of the body, was significantly greater for trastuzumab, IFN-PEG40 and IFN-PEG12 (all >3% dose over 8h) when compared to native IFN (0.9% dose). Conversely, the thoracic lymph/plasma (L/P) concentration ratio and thus efficiency of extravasation and transport through the interstitium to lymph was highest for the smaller proteins IFN and IFN-PEG12 (at 90-100% vs 15-30% for trastuzumab and IFN-PEG40). The lower total recovery of IFN and IFN-PEG12 in thoracic lymph reflected more rapid systemic clearance and thus lower systemic exposure. For all therapeutic proteins, the majority (>80%) of lymph access occurred via the hepatic and mesenteric lymphatics. This lymphatic distribution pattern was supported by quantitative imaging of the lymph node distribution of IV administered Cy5 labelled trastuzumab. Optimizing the properties of IV administered therapeutic proteins represents a viable approach to better target and treat pathological states involving the lymphatics, particularly in the liver and mesentery. This includes cancer metastases, infections and inflammatory diseases. Successful development of the novel technique to collect hepatic lymph will also enable future work to evaluate tissue-specific lymph transport in health and disease.


Asunto(s)
Interferones/administración & dosificación , Linfa/metabolismo , Polietilenglicoles/administración & dosificación , Trastuzumab/administración & dosificación , Administración Intravenosa , Animales , Interferones/química , Interferones/farmacocinética , Hígado , Masculino , Mesenterio , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Ratas Sprague-Dawley , Tórax , Trastuzumab/farmacocinética
17.
J Am Chem Soc ; 140(3): 1170-1178, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29262256

RESUMEN

Cyclization and polymer conjugation are two commonly used approaches for enhancing the pharmacological properties of protein drugs. However, cyclization of parental proteins often only affords a modest improvement in biochemical or cell-based in vitro assays. Moreover, very few studies have included a systematic pharmacological evaluation of cyclized protein-based therapeutics in live animals. On the other hand, polymer-conjugated proteins have longer circulation half-lives but usually show poor tumor penetration and suboptimal pharmacodynamics due to increased steric hindrance. We herein report the generation of a head-to-tail interferon-poly(α-amino acid) macrocycle conjugate circ-P(EG3Glu)20-IFN by combining the aforementioned two approaches. We then compared the antitumor pharmacological activity of this macrocycle conjugate against its linear counterparts, N-P(EG3Glu)20-IFN, C-IFN-P(EG3Glu)20, and C-IFN-PEG. Our results found circ-P(EG3Glu)20-IFN to show considerably greater stability, binding affinity, and in vitro antiproliferative activity toward OVCAR3 cells than the three linear conjugates. More importantly, circ-P(EG3Glu)20-IFN exhibited longer circulation half-life, remarkably higher tumor retention, and deeper tumor penetration in vivo. As a result, administration of the macrocyclic conjugate could effectively inhibit tumor progression and extend survival in mice bearing established xenograft human OVCAR3 or SKOV3 tumors without causing severe paraneoplastic syndromes. Taken together, our study provided until now the most relevant experimental evidence in strong support of the in vivo benefit of macrocyclization of protein-polymer conjugates and for its application in next-generation therapeutics.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Interferones/química , Interferones/farmacología , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Interferones/farmacocinética , Interferones/uso terapéutico , Compuestos Macrocíclicos/farmacocinética , Compuestos Macrocíclicos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Ácido Poliglutámico/química , Ácido Poliglutámico/farmacocinética , Ácido Poliglutámico/farmacología , Ácido Poliglutámico/uso terapéutico , Ratas Sprague-Dawley
19.
Forensic Sci Int ; 278: 313-325, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28800550

RESUMEN

Protein-based medicines, or large molecule medicines, are innovative products used to treat various diseases like hepatitis or cancer. This new generation of molecules are usually expensive, and thus represents an attractive target for the counterfeiters. Due to the complexity of their chemical structure, their analysis for counterfeit detection is more difficult than small molecule medicines. The aim of the article is to demonstrate that Raman spectroscopy and microscopy can be used for the fast analysis of counterfeits of protein-based medicines. Twelve types of medicines, under liquid or lyophilized form, have been analyzed by a Raman spectrometer through their glass packaging and ten of them also by a Raman microscope with drop deposition on a gold plate. The optimization of the acquisition parameters has first been described. Then the identification of the studied products has been presented with the attribution of the protein bands observed on the spectra. Finally the methods were successfully applied to seven counterfeits of these products and their chemical composition identified by spectral analysis. Counterfeits can indeed be detected if the excipient profile differs, if no protein is present, or if the genuine sample has been strongly diluted by the counterfeiters. Raman spectroscopy and microscopy have thus proved efficient for the fast analysis of counterfeits of protein-based medicines.


Asunto(s)
Medicamentos Falsificados , Preparaciones Farmacéuticas/química , Espectrometría Raman , Anticuerpos Monoclonales/química , Medicamentos Falsificados/análisis , Medicamentos Falsificados/química , Humanos , Interferones/química , Proteínas/química
20.
J Clin Pharmacol ; 57(3): 287-296, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27654843

RESUMEN

This review paper summarizes the epidemiology of hepatitis C virus (HCV) and chronic HCV infection, including HCV virology and treatment regimens. Specifically, we focus on the evolution of past, current, and future HCV treatment options, the reasons for treatment failure, and the impact of resistance-associated variants on treatment success.


Asunto(s)
Antivirales/uso terapéutico , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/epidemiología , Antivirales/administración & dosificación , Antivirales/efectos adversos , Quimioterapia Combinada , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatitis C Crónica/complicaciones , Humanos , Interferones/química , Interferones/uso terapéutico , Fosfoproteínas/antagonistas & inhibidores , Inhibidores de Proteasas/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Ribavirina/uso terapéutico , Serina Proteasas , Respuesta Virológica Sostenida , Proteínas no Estructurales Virales/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...