Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Int J Mol Sci ; 23(15)2022 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-35955604

RESUMEN

Monoclonal antibodies (mAbs) are one of the most successful and versatile protein-based pharmaceutical products used to treat multiple pathological conditions. The remarkable specificity of mAbs and their affinity for biological targets has led to the implementation of mAbs in the therapeutic regime of oncogenic, chronic inflammatory, cardiovascular, and infectious diseases. Thus, the discovery of novel mAbs with defined functional activities is of crucial importance to expand our ability to address current and future clinical challenges. In vitro, antigen-driven affinity selection employing phage display biopanning is a commonly used technique to isolate mAbs. The success of biopanning is dependent on the quality and the presentation format of the antigen, which is critical when isolating mAbs against membrane protein targets. Here, we provide a comprehensive investigation of two established panning strategies, surface-tethering of a recombinant extracellular domain and cell-based biopanning, to examine the impact of antigen presentation on selection outcomes with regards to the isolation of positive mAbs with functional potential against a proof-of-concept type I cell surface receptor. Based on the higher sequence diversity of the resulting antibody repertoire, presentation of a type I membrane protein in soluble form was more advantageous over presentation in cell-based format. Our results will contribute to inform and guide future antibody discovery campaigns against cell surface proteins.


Asunto(s)
Bacteriófagos , Biblioteca de Péptidos , Anticuerpos Monoclonales , Bacteriófagos/genética , Bioprospección , Técnicas de Visualización de Superficie Celular/métodos , Proteínas de la Membrana
2.
Biomaterials ; 283: 121416, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35217483

RESUMEN

Personalised nanomedicine is an advancing field which has developed significant improvements for targeting therapeutics to aggressive cancer and with fewer side effects. The treatment of gliomas such as glioblastoma (or other brain tumours), with nanomedicine is complicated by a commonly poor accumulation of drugs in tumour tissue owing to the partially intact blood-brain barrier (BBB). Nonetheless, the BBB becomes compromised following surgical intervention, and gradually with disease progression. Increased vasculature permeability generated by a tumour, combined with decreased BBB integrity, offers a mechanism to enhance therapeutic outcomes. We monitored a spontaneous glioma tumour model in immunocompetent mice with ongoing T2-weighted and contrast-enhanced T1-weighted magnetic resonance imaging gradient echo and spin echo sequences to predict an optimal "leakiness" stage for nanomedicine injections. To ascertain the effectiveness of targeted nanomedicines in treating brain tumours, subsequent systemic administration of targeted hyperbranched polymers was then utislised, to deliver the therapeutic payload when both the tumour and brain vascularity had become sufficiently susceptible to allow drug accumulation. Treatment with either doxorubicin-loaded hyperbranched polymer, or the same nanomedicine targeted to an ephrin receptor (EphA2) using a bispecific antibody, resulted in uptake of chemotherapeutic doxorubicin in the tumour and in reduced tumour growth. Compared to vehicle and doxorubicin only, nanoparticle delivered doxorubicin resulted in increased tumour apoptosis, while averting cardiotoxicity. This suggests that polyethylene based (PEGylated)-nanoparticle delivered doxorubicin could provide a more efficient treatment in tumours with a disrupted BBB, and that treatment should commence immediately following detection of gadolinium permeability, with early detection and ongoing 'leakiness' monitoring in susceptible patients being a key factor.


Asunto(s)
Neoplasias Encefálicas , Nanomedicina , Animales , Barrera Hematoencefálica , Encéfalo , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina , Sistemas de Liberación de Medicamentos/métodos , Humanos , Ratones , Nanomedicina/métodos
3.
Commun Biol ; 4(1): 390, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33758337

RESUMEN

Coagulation factor IX (FIX) is a complex post-translationally modified human serum glycoprotein and high-value biopharmaceutical. The quality of recombinant FIX (rFIX), especially complete γ-carboxylation, is critical for rFIX clinical efficacy. Bioreactor operating conditions can impact rFIX production and post-translational modifications (PTMs). With the goal of optimizing rFIX production, we developed a suite of Data Independent Acquisition Mass Spectrometry (DIA-MS) proteomics methods and used these to investigate rFIX yield, γ-carboxylation, other PTMs, and host cell proteins during bioreactor culture and after purification. We detail the dynamics of site-specific PTM occupancy and structure on rFIX during production, which correlated with the efficiency of purification and the quality of the purified product. We identified new PTMs in rFIX near the GLA domain which could impact rFIX GLA-dependent purification and function. Our workflows are applicable to other biologics and expression systems, and should aid in the optimization and quality control of upstream and downstream bioprocesses.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Coagulantes/aislamiento & purificación , Medios de Cultivo/metabolismo , Factor IX/aislamiento & purificación , Células Cultivadas , Cromatografía de Fase Inversa , Humanos , Conformación Proteica , Procesamiento Proteico-Postraduccional , Proteómica , Control de Calidad , Proteínas Recombinantes/aislamiento & purificación , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem , Carga de Trabajo
4.
ACS Sens ; 6(3): 764-776, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33481587

RESUMEN

Engineering antibodies to improve target specificity, reduce detection limits, or introduce novel functionality is an important research area for biosensor development. While various affinity biosensors have been developed to generate an output signal upon varying analyte concentrations, reversible and continuous protein monitoring in complex biological samples remains challenging. Herein, we explore the concept of directed evolution to modulate dissociation kinetics of a high affinity anti-epidermal growth factor receptor (EGFR) single-chain variable antibody fragment (scFv) to enable continuous protein sensing in a label-free binding assay. A mutant scFv library was generated from the wild type (WT) fragment via targeted permutation of four residues in the antibody-antigen-binding interface. A single round of phage display biopanning complemented with high-throughput screening methods then permitted isolation of a specific binder with fast reaction kinetics. We were able to obtain ∼30 times faster dissociation rates when compared to the WT without appreciably affecting overall affinity and specificity by targeting a single paratope that is known to contribute to the binding interaction. Suitability of a resulting mutant fragment to sense varying antigen concentrations in continuous mode was demonstrated in a modified label-free binding assay, achieving low nanomolar detection limits (KD = 8.39 nM). We also confirmed these results using an independent detection mechanism developed previously by our group, incorporating a polarity-dependent fluorescent dye into the scFv and reading out EGFR binding based on fluorescence wavelength shifts. In future, this generic approach could be employed to generate improved or novel binders for proteins of interest, ready for deployment in a broad range of assay platforms.


Asunto(s)
Técnicas Biosensibles , Anticuerpos de Cadena Única , Proteínas Recombinantes , Anticuerpos de Cadena Única/genética
5.
Blood Transfus ; 19(1): 64-72, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33085594

RESUMEN

BACKGROUND: RhD-immunoglobulin (RhIg) prevents anti-D alloimmunisation in D-negative pregnant women when the fetus is D-positive, reducing the incidence of haemolytic disease of the fetus and newborn. Manufacturing RhIg is reliant on the limited supply of plasma donations with anti-D antibodies. Monoclonal antibody (mAb) development platforms such as phage display, require blood samples to be collected from anti-D donors, which may be a complicated process. The blood filter chamber (BFC) discarded after an anti-D donor's donation might provide a source of Ig-encoding RNA. This study aims to evaluate whether used BFCs are a suitable source of Ig-encoding RNA for phage display. MATERIAL AND METHODS: Haemonetics PCS2 BFCs were obtained from 10 anti-D donors for total RNA extraction, cDNA synthesis and amplification of VH and VL IgG sequences for assembly of single-chain variable fragments (scFvs). A scFv-phage display library was constructed and 3 rounds of biopanning were performed using D-positive and D-negative red blood cells (RBCs). Positive phage clones were isolated, Sanger sequenced and, where possible, reformatted into full-length human IgGs to define specificity. The BFC aggregates from 2 anti-D donors underwent a Wright-Giemsa stain and hematological cell count. RESULTS: Of 10 BFCs, a sufficient yield of total RNA for library construction was obtained from BFCs containing cellular aggregates (n=5). Aggregate analysis showed lymphocytes were the cellular source of Ig-encoding RNA. From the 5 samples with aggregates, scFvs were assembled from amplified IgG variable regions. The library constructed from 1 of these samples resulted in the isolation of clones binding to D-positive RBCs with IGHV3 gene usage. Of the 4 reformatted IgG, 3 were anti-D and 1 had undefined specificity. DISCUSSION: BFC aggregates are a new and convenient source of Ig-encoding RNA which can be used to construct Ig gene libraries for mAb isolation and discovery via antibody phage display.


Asunto(s)
Anticuerpos Monoclonales/análisis , Plasma/química , Globulina Inmune rho(D)/análisis , Animales , Donantes de Sangre , Células CHO , Cricetulus , Filtración , Biblioteca de Genes , Humanos , Biblioteca de Péptidos , ARN/análisis
6.
BMC Cancer ; 20(1): 1214, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33302918

RESUMEN

BACKGROUND: Glypican-1 is a heparan sulfate proteoglycan that is overexpressed in prostate cancer (PCa), and a variety of solid tumors. Importantly, expression is restricted in normal tissue, making it an ideal tumor targeting antigen. Since there is clinical and preclinical evidence of the efficacy of Bispecific T cell Engager (BiTE) therapy in PCa, we sought to produce and test the efficacy of a GPC-1 targeted BiTE construct based on the Miltuximab® sequence. Miltuximab® is a clinical stage anti-GPC-1 antibody that has proven safe in first in human trials. METHODS: The single chain variable fragment (scFv) of Miltuximab® and the CD3 binding sequence of Blinatumomab were combined in a standard BiTE format. Binding of the construct to immobilised recombinant CD3 and GPC-1 antigens was assessed by ELISA and BiaCore, and binding to cell surface-expressed antigens was measured by flow cytometry. The ability of MIL-38-CD3 to activate T cells was assessed using in vitro co-culture assays with tumour cell lines of varying GPC-1 expression by measurement of CD69 and CD25 expression, before cytolytic activity was assessed in a similar co-culture. The release of inflammatory cytokines from T cells was measured by ELISA and expression of PD-1 on the T cell surface was measured by flow cytometry. RESULTS: Binding activity of MIL-38-CD3 to both cell surface-expressed and immobilised recombinant GPC-1 and CD3 was retained. MIL-38-CD3 was able to mediate the activation of peripheral blood T cells from healthy individuals, resulting in the release of inflammatory cytokines TNF and IFN-g. Activation was reliant on GPC-1 expression as MIL-38-CD3 mediated only low level T cell activation in the presence of C3 cells (constitutively low GPC-1 expression). Activated T cells were redirected to lyse PCa cell lines PC3 and DU-145 (GPC-1 moderate or high expression, respectively) but could not kill GPC-1 negative Raji cells. The expression of PD-1 was up-regulated on the surface of MIL-38-CD3 activated T cells, suggesting potential for synergy with checkpoint inhibition. CONCLUSIONS: This study reports preclinical findings into the efficacy of targeting GPC-1 in PCa with BiTE construct MIL-38-CD3. We show the specificity and efficacy of the construct, supporting its further preclinical development.


Asunto(s)
Adenocarcinoma/patología , Anticuerpos Biespecíficos/farmacología , Glipicanos/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/patología , Anticuerpos de Cadena Única/farmacología , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Citotóxicos/inmunología , Adenocarcinoma/inmunología , Anticuerpos Biespecíficos/inmunología , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Complejo CD3/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/metabolismo , Citotoxicidad Inmunológica , Glipicanos/antagonistas & inhibidores , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Subunidad alfa del Receptor de Interleucina-2/análisis , Lectinas Tipo C/análisis , Activación de Linfocitos , Masculino , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Próstata/inmunología , Proteínas Recombinantes/inmunología , Anticuerpos de Cadena Única/inmunología , Linfocitos T Citotóxicos/metabolismo
7.
Front Immunol ; 11: 1986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983137

RESUMEN

Monoclonal antibodies (mAbs) have become one of the most important classes of biopharmaceutical products, and they continue to dominate the universe of biopharmaceutical markets in terms of approval and sales. They are the most profitable single product class, where they represent six of the top ten selling drugs. At the beginning of the 1990s, an in vitro antibody selection technology known as antibody phage display was developed by John McCafferty and Sir. Gregory Winter that enabled the discovery of human antibodies for diverse applications, particularly antibody-based drugs. They created combinatorial antibody libraries on filamentous phage to be utilized for generating antigen specific antibodies in a matter of weeks. Since then, more than 70 phage-derived antibodies entered clinical studies and 14 of them have been approved. These antibodies are indicated for cancer, and non-cancer medical conditions, such as inflammatory, optical, infectious, or immunological diseases. This review will illustrate the utility of phage display as a powerful platform for therapeutic antibodies discovery and describe in detail all the approved mAbs derived from phage display.


Asunto(s)
Anticuerpos Monoclonales , Técnicas de Visualización de Superficie Celular , Desarrollo de Medicamentos/métodos , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos/genética , Especificidad de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Ingeniería Genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Terapia Molecular Dirigida , Investigación Biomédica Traslacional , Resultado del Tratamiento
8.
ACS Nano ; 14(10): 13739-13753, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32936613

RESUMEN

Integrating nanomaterials with biological entities has led to the development of diagnostic tools and biotechnology-derived therapeutic products. However, to optimize the design of these hybrid bionanomaterials, it is essential to understand how controlling the biological interactions will influence desired outcomes. Ultimately, this knowledge will allow more rapid translation from the bench to the clinic. In this paper, we developed a micellar system that was assembled using modular antibody-polymer amphiphilic materials. The amphiphilic nature was established using either poly(ethylene glycol) (PEG) or a single-chain variable fragment (scFv) from an antibody as the hydrophile and a thermoresponsive polymer (poly(oligoethylene glycol) methyl ether methacrylate) as the hydrophobe. By varying the ratios of these components, a series of nanoparticles with different antibody content was self-assembled, where the surface presentation of targeting ligand was carefully controlled. In vitro and in vivo analysis of these systems identified a mismatch between the optimal targeting ligand density to achieve maximum cell association in vitro compared to tumor accumulation in vivo. For this system, we determined an optimum antibody density for both longer circulation and enhanced targeting to tumors that balanced stealthiness of the particle (to evade immune recognition as determined in both mouse models and in whole human blood) with enhanced accumulation achieved through receptor binding on tumor cells in solid tumors. This approach provides fundamental insights into how different antibody densities affect the interaction of designed nanoparticles with both target cells and immune cells, thereby offering a method to probe the intricate interplay between increased targeting efficiency and the subsequent immune response to nanoparticles.


Asunto(s)
Micelas , Nanopartículas , Ligandos , Polietilenglicoles , Polímeros
9.
Expert Opin Drug Deliv ; 17(9): 1189-1211, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32524851

RESUMEN

INTRODUCTION: Monoclonal antibodies have been utilized in clinical and basic research for the treatment of various malignancies. Whilst all therapeutically approved monoclonal antibodies or fragments thereof are directed against cell-surface receptors or proteins of the human secretome, intracellular antigen targeting strategies still await translation into the clinic. This contradicts the notion of antibodies being the magic bullet concept as many cancer targets are out of reach. AREAS COVERED: This review provides a summary of intracellular translocation strategies that were successfully employed for antibody delivery in preclinical studies. Examples encompass a variety of different approaches such as polymeric and lipid-based nanoparticles (NP), biomimetics, bispecific antibody constructs, the use of cell-penetrating peptides, as well as various sophisticated combinations thereof. We will further discuss endosomal escape as the major bottleneck in functional intracellular transport and provide suggestions on how to overcome current challenges. EXPERT OPINION: Despite significant advances in protein delivery technologies, reports of highly efficient transport vehicles are sparse when systemically applied in vivo. Consequently, more detailed mechanistic studies are needed to identify and optimize the molecular 'Achilles heel' of individual methodologies. Ultimately, to target intracellular proteins that have been undruggable in the past, a combination of strategies may be required.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Sistemas de Liberación de Medicamentos , Transporte Biológico , Péptidos de Penetración Celular/metabolismo , Endosomas/metabolismo , Humanos , Nanopartículas/química , Polímeros/química
10.
ACS Cent Sci ; 6(5): 727-738, 2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32490189

RESUMEN

Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.

11.
Lancet Infect Dis ; 20(4): 445-454, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32027842

RESUMEN

BACKGROUND: The monoclonal antibody m102.4 is a potent, fully human antibody that neutralises Hendra and Nipah viruses in vitro and in vivo. We aimed to investigate the safety, tolerability, pharmacokinetics, and immunogenicity of m102.4 in healthy adults. METHODS: In this double-blind, placebo-controlled, single-centre, dose-escalation, phase 1 trial of m102.4, we randomly assigned healthy adults aged 18-50 years with a body-mass index of 18·0-35·0 kg/m2 to one of five cohorts. A sentinel pair for each cohort was randomly assigned to either m102.4 or placebo. The remaining participants in each cohort were randomly assigned (5:1) to receive m102.4 or placebo. Cohorts 1-4 received a single intravenous infusion of m102.4 at doses of 1 mg/kg (cohort 1), 3 mg/kg (cohort 2), 10 mg/kg (cohort 3), and 20 mg/kg (cohort 4), and were monitored for 113 days. Cohort 5 received two infusions of 20 mg/kg 72 h apart and were monitored for 123 days. The primary outcomes were safety and tolerability. Secondary outcomes were pharmacokinetics and immunogenicity. Analyses were completed according to protocol. The study was registered on the Australian New Zealand Clinical Trials Registry, ACTRN12615000395538. FINDINGS: Between March 27, 2015, and June 16, 2016, 40 (52%) of 77 healthy screened adults were enrolled in the study. Eight participants were assigned to each cohort (six received m102.4 and two received placebo). 86 treatment-emergent adverse events were reported, with similar rates between placebo and treatment groups. The most common treatment-related event was headache (12 [40%] of 30 participants in the combined m102.4 group, and three [30%] of ten participants in the pooled placebo group). No deaths or severe adverse events leading to study discontinuation occurred. Pharmacokinetics based on those receiving m102.4 (n=30) were linear, with a median half-life of 663·3 h (range 474·3-735·1) for cohort 1, 466·3 h (382·8-522·3) for cohort 2, 397·0 h (333·9-491·8) for cohort 3, and 466·7 h (351·0-889·6) for cohort 4. The elimination kinetics of those receiving repeated dosing (cohort 5) were similar to those of single-dose recipients (median elimination half-time 472·0 [385·6-592·0]). Anti-m102.4 antibodies were not detected at any time-point during the study. INTERPRETATION: Single and repeated dosing of m102.4 were well tolerated and safe, displayed linear pharmacokinetics, and showed no evidence of an immunogenic response. This study will inform future dosing regimens for m102.4 to achieve prolonged exposure for systemic efficacy to prevent and treat henipavirus infections. FUNDING: Queensland Department of Health, the National Health and Medical Research Council, and the National Hendra Virus Research Program.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Glicoproteínas/inmunología , Voluntarios Sanos , Henipavirus/inmunología , Inmunogenicidad Vacunal , Seguridad , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Australia , Método Doble Ciego , Femenino , Cefalea/etiología , Humanos , Infusiones Intravenosas , Masculino
12.
Chem Sci ; 11(12): 3268-3280, 2020 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-34122834

RESUMEN

There remain several key challenges to existing therapeutic systems for cancer therapy, such as quantitatively determining the true, tissue-specific drug release profile in vivo, as well as reducing side-effects for an increased standard of care. Hence, it is crucial to engineer new materials that allow for a better understanding of the in vivo pharmacokinetic/pharmacodynamic behaviours of therapeutics. We have expanded on recent "click-to-release" bioorthogonal pro-drug activation of antibody-drug conjugates (ADCs) to develop a modular and controlled theranostic system for quantitatively assessing site-specific drug activation and deposition from a nanocarrier molecule, by employing defined chemistries. The exploitation of quantitative imaging using positron emission tomography (PET) together with pre-targeted bioorthogonal chemistries in our system provided an effective means to assess in real-time the exact amount of active drug administered at precise sites in the animal; our methodology introduces flexibility in both the targeting and therapeutic components that is specific to nanomedicines and offers unique advantages over other technologies. In this approach, the in vivo click reaction facilitates pro-drug activation as well as provides a quantitative means to investigate the dynamic behaviour of the therapeutic agent.

13.
Lab Chip ; 19(24): 4083-4092, 2019 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-31712799

RESUMEN

Phage display methodologies offer a versatile platform for the isolation of single-chain Fv (scFv) molecules which may be rebuilt into monoclonal antibodies. Herein, we report on a complete workflow termed PhageXpress, for rapid selection of single-chain Fv sequences by leveraging electrohydrodynamic-manipulation of a solution containing phage library particles to enhance target binding whilst minimizing non-specific interactions. Our PhageXpress technique is combined with Oxford Nanopore Technologies' MinION sequencer and custom bioinformatics to achieve high-throughput screening of phage libraries. We performed 4 rounds of biopanning against Dengue virus (DENV) non-structural protein 1 (NS1) using traditional methods (4 week turnaround), which resulted in the isolation of 19 unique scFv clones. We validated the feasibility and efficiency of the PhageXpress method utilizing the same phage library and antigen target. Notably, we successfully mapped 14 of the 19 anti-NS1 scFv sequences (∼74%) with our new method, despite using ∼30-fold less particles during screening and conducting only a single round of biopanning. We believe this approach supersedes traditional methods for the discovery of bio-recognition molecules such as antibodies by speeding up the process for the development of therapeutic and diagnostic biologics.


Asunto(s)
Anticuerpos Antivirales , Secuenciación de Nanoporos , Biblioteca de Péptidos , Anticuerpos de Cadena Única , Anticuerpos Antivirales/química , Anticuerpos Antivirales/genética , Virus del Dengue/química , Humanos , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Proteínas no Estructurales Virales/química
14.
Antibodies (Basel) ; 8(1)2019 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-31544821

RESUMEN

CD117 (c-Kit) is a tyrosine kinase receptor that is overexpressed in multiple dog tumors. There is 100% homology between the juxtamembrane domain of human and canine CD117, and many cancer-causing mutations occur in this region in both species. Thus, CD117 is an important target for cancer treatment in dogs and for comparative oncology studies. Currently, there is no monoclonal antibody (mAb) specifically designed to target the exposed region of canine CD117, although there exist some with species cross-reactivity. We panned a naïve phage display library to isolate antibodies against recombinant CD117 on whole cells. Several mAbs were isolated and were shown to bind recombinant canine CD117 at low- to sub-nanomolar affinity. Additionally, binding to native canine CD117 was confirmed by immunohistochemistry and by flow cytometry. Competitive binding assays also identified mAbs that competed with the CD117 receptor-specific ligand, the stem cell factor (SCF). These results show the ability of our cell-based biopanning strategy to isolate a panel of antibodies that have varied characteristics when used in different binding assays. These in vitro/ex vivo assessments suggest that some of the isolated mAbs might be promising candidates for targeting overexpressed CD117 in canine cancers for different useful applications.

15.
Biomaterials ; 216: 119232, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31195300

RESUMEN

Combined and targeted therapy have been extensively employed to achieve more effective elimination of tumor tissues. In this study, biocompatible multifunctional lipid-coated calcium phosphate nanoparticles (LCP NPs) were designed and constructed as an efficient targeted delivery system for combined gene/photothermal therapy to inhibit growth of the triple negative breast tumor (MDA-MB-468) in vitro and in vivo. LCP NPs were functionalized with a bispecific antibody (BsAb) via non-covalent bond specific for methoxy group of PEG (mPEG) on the particle surface. This BsAb is also able to target epidermal growth factor receptor (EGFR) expressed on MDA-MB-468 cells. Such LCP-BsAb NPs loaded with Cell Death (CD)-siRNA and indocyanine green (ICG) were efficiently taken up by MDA-MB-468 cells, significantly inducing cell apoptosis and synergistically suppressing cell proliferation upon irradiation of 808 nm near-infrared laser. These targeted multifunctional LCP NPs more efficiently accumulated in the tumor tissue. The combined RNAi (CD-siRNA) and photothermal (ICG) therapy using the targeted LCP NPs nearly eliminated both small tumors (∼100 mm3) and large tumors (∼500 mm3) in the mouse model. Thus, the well-devised multifunctional LCP NPs are one of the most promising delivery systems for combined and targeted cancer therapy.


Asunto(s)
Fosfatos de Calcio/química , Colorantes/administración & dosificación , Portadores de Fármacos/química , Verde de Indocianina/administración & dosificación , Lípidos/química , Neoplasias de la Mama Triple Negativas/terapia , Animales , Línea Celular Tumoral , Colorantes/uso terapéutico , Sistemas de Liberación de Medicamentos , Femenino , Terapia Genética , Humanos , Hipertermia Inducida , Verde de Indocianina/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Tratamiento con ARN de Interferencia , Neoplasias de la Mama Triple Negativas/genética
16.
Adv Healthc Mater ; 8(9): e1801607, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30868751

RESUMEN

Low-fouling or "stealth" particles composed of poly(ethylene glycol) (PEG) display a striking ability to evade phagocytic cell uptake. However, functionalizing them for specific targeting is challenging. To address this challenge, stealth PEG particles prepared by a mesoporous silica templating method are functionalized with bispecific antibodies (BsAbs) to obtain PEG-BsAb particles via a one-step binding strategy for cell and tumor targeting. The dual specificity of the BsAbs-one arm binds to the PEG particles while the other targets a cell antigen (epidermal growth factor receptor, EGFR)-is exploited to modulate the number of targeting ligands per particle. Increasing the BsAb incubation concentration increases the amount of BsAb tethered to the PEG particles and enhances targeting and internalization into breast cancer cells overexpressing EGFR. The degree of BsAb functionalization does not significantly reduce the stealth properties of the PEG particles ex vivo, as assessed by their interactions with primary human blood granulocytes and monocytes. Although increasing the BsAb amount on PEG particles does not lead to the expected improvement in tumor accumulation in vivo, BsAb functionalization facilitates tumor cell uptake of PEG particles. This work highlights strategies to balance evading nonspecific clearance pathways, while improving tumor targeting and accumulation.


Asunto(s)
Anticuerpos Biespecíficos/química , Sistemas de Liberación de Medicamentos/métodos , Polietilenglicoles/química , Animales , Células CHO , Línea Celular Tumoral , Cricetulus , Receptores ErbB/química , Humanos
17.
Methods Mol Biol ; 1827: 179-195, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196498

RESUMEN

Cell membrane proteins serve as attractive targets for biopharmaceutical development in addition to gauging their fundamental process in a biological system. Approximately 38% of the entire genome codes for plasma membrane proteins; however the discovery and development of antibody binders to such targets are a technical challenge. Methods to raise binders against such targets by cloning and expressing soluble extracellular regions have been met with limited success due to the loss of critical epitopes, with the resulting antibodies failing to bind to their target in its native conformation. This chapter outlines a "cell based biopanning" method in order to isolate antibodies against membrane proteins in their native conformation using transiently expressed, GFP-tagged target proteins. This method overcomes the limitations of non-specific binding of phage to the cells, abundance of irrelevant antigens on the cell surface, while retaining the native structure of the antigen on the cell surface.


Asunto(s)
Anticuerpos/aislamiento & purificación , Técnicas de Visualización de Superficie Celular/métodos , Proteínas de la Membrana/inmunología , Animales , Bacteriófagos/metabolismo , Células CHO , Precipitación Química , Células Clonales , Cricetinae , Cricetulus , Escherichia coli/metabolismo , Citometría de Flujo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Transfección
18.
Nanoscale ; 10(9): 4258-4266, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29436549

RESUMEN

The conjugation of ligands to nanoparticle platforms for the target delivery of therapeutic agents to the tumor tissue is one of the promising anti-cancer strategies. However, conventional nanoparticle platforms are not so effective in terms of the selectivity and transfection efficiency. In this study, we designed and developed a dual-target drug/gene delivery system based on lipid-coated calcium phosphate (LCP) nanoparticles (NPs) for significantly enhanced siRNA cellular uptake and transfection efficiency. LCP NPs loaded with therapeutic siRNA were conjugated with a controlled number of folic acid and/or EGFR-specific single chain fragment antibody (ABX-EGF scFv). The uptake of ABX-EGF scFv-modified (LCP-scFv) and folic acid-modified LCP NPs (LCP-FA) by human breast tumor cells (MDA-MB-468) was significantly higher with an optimal ligand density on each NP surface (LCP-125scFv and LCP-100FA). Co-conjugation with sub-optimal dual ligands (50 FA and 75 ABX-EGF scFv) per LCP NP (LCP-50FA-75scFv) further enhanced the cellular uptake. More significantly, much more NPs were delivered to the MDA-MB-468 tumor tissue in the nude mouse model when LCP-50FA-75scFv NPs were used. Therefore, the new dual-ligand LCP NPs may be a valuable targeting system for human breast cancer diagnosis and therapy.


Asunto(s)
Fosfatos de Calcio/química , Sistemas de Liberación de Medicamentos , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Humanos , Ligandos , Lípidos , Ratones , Ratones Desnudos , Anticuerpos de Cadena Única
19.
Adv Healthc Mater ; 7(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28961378

RESUMEN

Targeted nanomedicines have significantly changed the way new therapeutics are designed to treat disease. Central to successful therapeutics is the ability to control the dynamics of protein-nanomaterial interactions to enhance the therapeutic effect of the nanomedicine. The aim of this review is to illustrate the diversity and versatility of the conjugation approaches involved in the synthesis of antibody-nanoparticle conjugates, and highlight significant new advances in the field of bioconjugation. Such nanomedicines have found utility as both advanced therapeutic agents, as well as more complex imaging contrast agents that can provide both anatomical and functional information of diseased tissue. While such conjugates show significant promise as next generation targeted nanomedicines, it is recognized that there are in fact no clinically approved targeted therapeutics on the market. This fact is reflected upon within this review, and attempts are made to draw some reasoning from the complexities associated with the bioconjugation chemistry approaches that are typically utilized. Present trends, as well as future directions of next generation targeted nanomedicines are also discussed.


Asunto(s)
Anticuerpos/química , Nanopartículas/química , Nanoestructuras/química , Animales , Humanos , Nanomedicina/métodos
20.
PLoS One ; 12(10): e0186137, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29059207

RESUMEN

Human malignant mesothelioma is a chemoresistant tumour that develops from mesothelial cells, commonly associated with asbestos exposure. Malignant mesothelioma incidence rates in European countries are still rising and Australia has one of the highest burdens of malignant mesothelioma on a population basis in the world. Therapy using systemic delivery of free cytotoxic agents is associated with many undesirable side effects due to non-selectivity, and is thus dose-limited which limits its therapeutic potential. Therefore, increasing the selectivity of anti-cancer agents has the potential to dramatically enhance drug efficacy and reduce toxicity. EnGeneIC Dream Vectors (EDV) are antibody-targeted nanocells which can be loaded with cytotoxic drugs and delivered to specific cancer cells via bispecific antibodies (BsAbs) which target the EDV and a cancer cell-specific receptor, simultaneously. BsAbs were designed to target doxorubicin-loaded EDVs to cancer cells via cell surface mesothelin (MSLN). Flow cytometry was used to investigate cell binding and induction of apoptosis, and confocal microscopy to visualize internalization. Mouse xenograft models were used to assess anti-tumour effects in vivo, followed by immunohistochemistry for ex vivo evaluation of proliferation and necrosis. BsAb-targeted, doxorubicin-loaded EDVs were able to bind to and internalize within mesothelioma cells in vitro via MSLN receptors and induce apoptosis. In mice xenografts, the BsAb-targeted, doxorubicin-loaded EDVs suppressed the tumour growth and also decreased cell proliferation. Thus, the use of MSLN-specific antibodies to deliver encapsulated doxorubicin can provide a novel and alternative modality for treatment of mesothelioma.


Asunto(s)
Proliferación Celular , Mesotelioma/patología , Receptores de Superficie Celular/metabolismo , Animales , Humanos , Mesotelina , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...