Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nature ; 587(7832): 18, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33097846
2.
Front Immunol ; 11: 655, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32457735

RESUMEN

Snake envenoming is a globally neglected public health problem. Antivenoms produced using animal hyperimmune plasma remain the standard therapy for snakebites. Although effective against systemic effects, conventional antivenoms have limited efficacy against local tissue damage. In addition, potential hypersensitivity reactions, high costs for animal maintenance, and difficulties in obtaining batch-to-batch homogeneity are some of the factors that have motivated the search for innovative and improved therapeutic products against such envenoming. In this study, we have developed a set of nanobodies (recombinant single-domain antigen-binding fragments from camelid heavy chain-only antibodies) against Bothrops atrox snake venom hemorrhagic and myotoxic components. An immune library was constructed after immunizing a Lama glama with whole venom of B. atrox, from which nanobodies were selected by phage display using partially purified hemorrhagic and myotoxic proteins. Biopanning selections retrieved 18 and eight different nanobodies against the hemorrhagic and the myotoxic proteins, respectively. In vivo assays in mice showed that five nanobodies inhibited the hemorrhagic activity of the proteins; three neutralized the hemorrhagic activity of whole B. atrox venom, while four nanobodies inhibited the myotoxic protein. A mixture of the anti-hemorrhagic and anti-myotoxic nanobodies neutralized the local tissue hemorrhage and myonecrosis induced by the whole venom, although the nanobody mixture failed to prevent the venom lethality. Nevertheless, our results demonstrate the efficacy and usefulness of these nanobodies to neutralize important pathologies of the venom, highlighting their potential as innovative therapeutic agents against envenoming by B. atrox, a viperid species causing many casualties in South America.


Asunto(s)
Antivenenos/uso terapéutico , Bothrops/metabolismo , Venenos de Crotálidos/química , Venenos de Crotálidos/inmunología , Hemorragia/tratamiento farmacológico , Factores Inmunológicos/uso terapéutico , Miotoxicidad/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Mordeduras de Serpientes/tratamiento farmacológico , Animales , Camélidos del Nuevo Mundo/inmunología , Inmunización/métodos , Masculino , Ratones , Resultado del Tratamiento
3.
Toxins (Basel) ; 10(4)2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29596324

RESUMEN

Toxic effects triggered by crotalic envenoming are mainly related to crotoxin (CTX), composed of a phospholipase A2 (CB) and a subunit with no toxic activity (CA). Camelids produce immunoglobulins G devoid of light chains, in which the antigen recognition domain is called VHH. Given their unique characteristics, VHHs were selected using Phage Display against CTX from Crotalus durissus terrificus. After three rounds of biopanning, four sequence profiles for CB (KF498602, KF498603, KF498604, and KF498605) and one for CA (KF498606) were revealed. All clones presented the VHH hallmark in FR2 and a long CDR3, with the exception of KF498606. After expressing pET22b-VHHs in E. coli, approximately 2 to 6 mg of protein per liter of culture were obtained. When tested for cross-reactivity, VHHs presented specificity for the Crotalus genus and were capable of recognizing CB through Western blot. KF498602 and KF498604 showed thermostability, and displayed affinity constants for CTX in the micro or nanomolar range. They inhibited in vitro CTX PLA2 activity, and CB cytotoxicity. Furthermore, KF498604 inhibited the CTX-induced myotoxicity in mice by 78.8%. Molecular docking revealed that KF498604 interacts with the CA–CB interface of CTX, seeming to block substrate access. Selected VHHs may be alternatives for the crotalic envenoming treatment.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Crotoxina/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Crotoxina/toxicidad , Escherichia coli/genética , Masculino , Ratones , Simulación del Acoplamiento Molecular , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/tratamiento farmacológico , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/uso terapéutico , Mordeduras de Serpientes/diagnóstico , Mordeduras de Serpientes/terapia
4.
Proc Natl Acad Sci U S A ; 114(38): 10184-10189, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874561

RESUMEN

CD47 is an antiphagocytic ligand broadly expressed on normal and malignant tissues that delivers an inhibitory signal through the receptor signal regulatory protein alpha (SIRPα). Inhibitors of the CD47-SIRPα interaction improve antitumor antibody responses by enhancing antibody-dependent cellular phagocytosis (ADCP) in xenograft models. Endogenous expression of CD47 on a variety of cell types, including erythrocytes, creates a formidable antigen sink that may limit the efficacy of CD47-targeting therapies. We generated a nanobody, A4, that blocks the CD47-SIRPα interaction. A4 synergizes with anti-PD-L1, but not anti-CTLA4, therapy in the syngeneic B16F10 melanoma model. Neither increased dosing nor half-life extension by fusion of A4 to IgG2a Fc (A4Fc) overcame the issue of an antigen sink or, in the case of A4Fc, systemic toxicity. Generation of a B16F10 cell line that secretes the A4 nanobody showed that an enhanced response to several immune therapies requires near-complete blockade of CD47 in the tumor microenvironment. Thus, strategies to localize CD47 blockade to tumors may be particularly valuable for immune therapy.


Asunto(s)
Antígeno CD47/antagonistas & inhibidores , Inmunoterapia/métodos , Melanoma Experimental/terapia , Anticuerpos de Dominio Único/uso terapéutico , Anemia/inducido químicamente , Animales , Antígeno CD47/inmunología , Evaluación Preclínica de Medicamentos , Ratones Endogámicos C57BL , Fagocitosis , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/farmacología , Microambiente Tumoral
5.
Bioconjug Chem ; 26(6): 1144-55, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-25938892

RESUMEN

A novel immunoconjugate (L-DOS47) was developed and characterized as a therapeutic agent for tumors expressing CEACAM6. The single domain antibody AFAIKL2, which targets CEACAM6, was expressed in the Escherichia coli BL21 (DE3) pT7-7 system. High purity urease (HPU) was extracted and purified from Jack bean meal. AFAIKL2 was activated using N-succinimidyl [4-iodoacetyl] aminobenzoate (SIAB) as the cross-linker and then conjugated to urease. The activation and conjugation reactions were controlled by altering pH. Under these conditions, the material ratio achieved conjugation ratios of 8-11 antibodies per urease molecule, the residual free urease content was practically negligible (<2%), and high purity (>95%) L-DOS47 conjugate was produced using only ultradiafiltration to remove unreacted antibody and hydrolyzed cross-linker. L-DOS47 was characterized by a panel of analytical techniques including SEC, IEC, Western blot, ELISA, and LC-MS(E) peptide mapping. As the antibody-urease conjugate ratio increased, a higher binding signal was observed. The specificity and cytotoxicity of L-DOS47 was confirmed by screening in four cell lines (BxPC-3, A549, MCF7, and CEACAM6-transfected H23). BxPC-3, a CEACAM6-expressing cell line was found to be most susceptible to L-DOS47. L-DOS47 is being investigated as a potential therapeutic agent in human phase I clinical studies for nonsmall cell lung cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Camélidos del Nuevo Mundo , Canavalia/enzimología , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Ureasa/uso terapéutico , Secuencia de Aminoácidos , Animales , Antineoplásicos/química , Línea Celular Tumoral , Terapia Enzimática , Humanos , Inmunoconjugados/química , Masculino , Ratones Desnudos , Datos de Secuencia Molecular , Anticuerpos de Dominio Único/química , Ureasa/química
6.
Biochimie ; 111: 82-106, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25656912

RESUMEN

The deposition of misfolded peptides and proteins in the form of amyloid fibrils is the hallmark of nearly fifty medical disorders, including Alzheimer's disease, Parkinson's disease, prion diseases and type II diabetes. These disorders, referred to as amyloidoses, generally become apparent late in life. Their psycho-sociological and economic incidence in western societies will be therefore considerable in the coming decades due to the ageing of the population. Neither preventing nor curative treatments are available yet. These disorders constitute therefore a medical challenge of great importance. Thus, an extensive research is being carried out to understand, at the molecular level, (i) how amyloidogenic proteins misfold and convert from their soluble form into amyloid fibrils, and (ii) how these aggregates or some of their oligomeric precursor species are toxic. The formation of amyloid fibrils proceeds through a complex nucleation/polymerisation mechanism with the formation of various species, including small oligomers. In this review, we focus on how VHHs or nanobodies, the antigen-binding domains of camelid heavy-chain antibodies, are being increasingly used to characterise each of the species formed on the pathway of fibril formation in terms of structure, stability, kinetics of formation and toxicity. We first introduce the characteristic features of nanobodies compared to those of conventional antibody fragments. Thereafter, we discuss how nanobodies, due to their unique properties, are used as probes to dissect the molecular mechanisms of misfolding and aggregation of six proteins associated with diseases, i.e. human lysozyme, ß2-microglobulin, α-synuclein, prion, polyadenylate binding protein nuclear 1 and amyloid ß-peptide. A brief general presentation of each disease and the associated peptide/protein is also provided. In addition, we discuss how nanobodies could be used as early diagnostic tools and as novel strategies to treat diseases associated with protein misfolding and aggregation.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Cadenas Pesadas de Inmunoglobulina/uso terapéutico , Agregación Patológica de Proteínas/tratamiento farmacológico , Deficiencias en la Proteostasis/tratamiento farmacológico , Anticuerpos de Dominio Único/uso terapéutico , Animales , Camélidos del Nuevo Mundo/genética , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Agregación Patológica de Proteínas/inmunología , Deficiencias en la Proteostasis/inmunología , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/inmunología
7.
J Virol ; 88(15): 8278-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829341

RESUMEN

UNLABELLED: Influenza virus neuraminidase (NA) is an interesting target of small-molecule antiviral drugs. We isolated a set of H5N1 NA-specific single-domain antibodies (N1-VHHm) and evaluated their in vitro and in vivo antiviral potential. Two of them inhibited the NA activity and in vitro replication of clade 1 and 2 H5N1 viruses. We then generated bivalent derivatives of N1-VHHm by two methods. First, we made N1-VHHb by genetically joining two N1-VHHm moieties with a flexible linker. Second, bivalent N1-VHH-Fc proteins were obtained by genetic fusion of the N1-VHHm moiety with the crystallizable region of mouse IgG2a (Fc). The in vitro antiviral potency against H5N1 of both bivalent N1-VHHb formats was 30- to 240-fold higher than that of their monovalent counterparts, with 50% inhibitory concentrations in the low nanomolar range. Moreover, single-dose prophylactic treatment with bivalent N1-VHHb or N1-VHH-Fc protected BALB/c mice against a lethal challenge with H5N1 virus, including an oseltamivir-resistant H5N1 variant. Surprisingly, an N1-VHH-Fc fusion without in vitro NA-inhibitory or antiviral activity also protected mice against an H5N1 challenge. Virus escape selection experiments indicated that one amino acid residue close to the catalytic site is required for N1-VHHm binding. We conclude that single-domain antibodies directed against influenza virus NA protect against H5N1 virus infection, and when engineered with a conventional Fc domain, they can do so in the absence of detectable NA-inhibitory activity. IMPORTANCE: Highly pathogenic H5N1 viruses are a zoonotic threat. Outbreaks of avian influenza caused by these viruses occur in many parts of the world and are associated with tremendous economic loss, and these viruses can cause very severe disease in humans. In such cases, small-molecule inhibitors of the viral NA are among the few treatment options for patients. However, treatment with such drugs often results in the emergence of resistant viruses. Here we show that single-domain antibody fragments that are specific for NA can bind and inhibit H5N1 viruses in vitro and can protect laboratory mice against a challenge with an H5N1 virus, including an oseltamivir-resistant virus. In addition, plant-produced VHH fused to a conventional Fc domain can protect in vivo even in the absence of NA-inhibitory activity. Thus, NA of influenza virus can be effectively targeted by single-domain antibody fragments, which are amenable to further engineering.


Asunto(s)
Antivirales/uso terapéutico , Subtipo H5N1 del Virus de la Influenza A/efectos de los fármacos , Neuraminidasa/antagonistas & inhibidores , Infecciones por Orthomyxoviridae/prevención & control , Anticuerpos de Dominio Único/uso terapéutico , Animales , Antivirales/inmunología , Modelos Animales de Enfermedad , Femenino , Subtipo H5N1 del Virus de la Influenza A/inmunología , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Anticuerpos de Dominio Único/inmunología , Resultado del Tratamiento
9.
Ther Deliv ; 4(10): 1321-36, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24116915

RESUMEN

Nanobodies (Nbs) are small antibody fragments derived from camelid heavy chain antibodies through recombinant gene technology. Their exceptional physicochemical properties, possibility of humanization and unique antigen recognition properties make them excellent candidates for targeted delivery of biologically active components. Several different therapeutic approaches based on the novel camelid Nbs have been developed to treat a wide range of diseases ranging from immune, bone, blood and neurological disorders; infectious diseases and cancer. This review provides a comprehensive overview of the current state of the use of camelid-derived Nbs as novel therapeutic agents against multiple diseases.


Asunto(s)
Camélidos del Nuevo Mundo/inmunología , Camelus/inmunología , Inmunoterapia/métodos , Inmunotoxinas/uso terapéutico , Nanomedicina/métodos , Anticuerpos de Dominio Único/uso terapéutico , Animales , Especificidad de Anticuerpos , Química Farmacéutica , Portadores de Fármacos , Humanos , Inmunotoxinas/química , Inmunotoxinas/inmunología , Conformación Proteica , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/inmunología , Relación Estructura-Actividad
10.
Postepy Hig Med Dosw (Online) ; 66: 348-58, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22706121

RESUMEN

 Serum of animals belonging to the Camelidae family (camels and llamas) contains fully active antibodies that are naturally devoid of light chains. Variable domains derived from heavy chain antibodies (hcAb) called VHHs or nanobodies™ can bind antigens as effectively as full-length antibodies and are easy to clone and express. Because of their potential, VHHs are being intensively studied as potential therapeutic, diagnostic and imaging tools. The paper reviews the molecular background of heavy chain antibodies and describes methods of obtaining recombinant fragments of heavy chain antibodies as well as their therapeutic, diagnostic and other applications.


Asunto(s)
Fragmentos de Inmunoglobulinas/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/uso terapéutico , Animales , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Camélidos del Nuevo Mundo , Cromatografía de Afinidad/métodos , Enfermedades Transmisibles/tratamiento farmacológico , Peces , Enfermedades Hematológicas/tratamiento farmacológico , Humanos , Fragmentos de Inmunoglobulinas/química , Fragmentos de Inmunoglobulinas/aislamiento & purificación , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/aislamiento & purificación , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/aislamiento & purificación , Anticuerpos de Dominio Único/uso terapéutico , Especificidad de la Especie
11.
Curr Opin Investig Drugs ; 10(11): 1212-24, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19876789

RESUMEN

Evolution has been continuously honing the design of antibodies to function as specific molecular markers that are able to alert the immune system to the presence of pathogenic antigens, and to recruit complement- and Fc receptor-bearing effector cells. During the past 25 years, the versatility of antibodies has been applied to several therapeutic applications. The development of new technologies, combined with data obtained using a new generation of antibody reagents, have allowed the adaptation of the design of antibodies to better match drug development requirements. Nanobodies are therapeutic proteins derived from the heavy-chain variable (VHH) domains that occur naturally in heavy-chain-only Ig molecules in camelidae. These VHH domains are the smallest known antigen-binding antibody fragments. Nanobodies can be easily produced in prokaryotic or eukaryotic host organisms, and their unique biophysical and pharmacological characteristics render these molecules ideal candidates for drug development. This review describes the structural properties of nanobodies and focuses on their unique features, which distinguishes these molecules from other antibody formats and small-molecule drugs. Possible therapeutic applications of nanobodies are discussed and data from phase I clinical trials of the novel 'first-in-class' anti-thrombotic agent ALX-0081 (Ablynx NV) are presented.


Asunto(s)
Diseño de Fármacos , Fragmentos de Inmunoglobulinas/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/inmunología , Anticuerpos de Dominio Único/uso terapéutico , Animales , Anticuerpos/inmunología , Camélidos del Nuevo Mundo , Ensayos Clínicos Fase I como Asunto , Fibrinolíticos/inmunología , Fibrinolíticos/farmacología , Humanos , Fragmentos de Inmunoglobulinas/inmunología , Anticuerpos de Dominio Único/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA