Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2410: 229-263, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34914050

RESUMO

Vaccines are one of mankind's greatest medical advances, and their use has drastically reduced and in some cases eliminated (e.g., smallpox) disease and death caused by infectious agents. Traditional vaccine modalities including live-attenuated pathogen vaccines, wholly inactivated pathogen vaccines, and protein-based pathogen subunit vaccines have successfully been used to create efficacious vaccines against measles, mumps, rubella, polio, and yellow fever. These traditional vaccine modalities, however, take many months to years to develop and have thus proven less effective for use in creating vaccines to emerging or reemerging infectious diseases (EIDs) including influenza, Human immunodeficiency virus (HIV), dengue virus (DENV), chikungunya virus (CHIKV), West Nile virus (WNV), Middle East respiratory syndrome (MERS), and the severe acute respiratory syndrome coronaviruses 1 and 2 (SARS-CoV and SARS-CoV-2). As factors such as climate change and increased globalization continue to increase the pace of EID development, newer vaccine modalities are required to develop vaccines that can prevent or attenuate EID outbreaks throughout the world. One such modality, DNA vaccines, has been studied for over 30 years and has numerous qualities that make them ideal for meeting the challenge of EIDs including; (1) DNA vaccine candidates can be designed within hours of publishing of a pathogens genetic sequence; (2) they can be manufactured cheaply and rapidly in large quantities; (3) they are thermostable and have reduced requirement for a cold-chain during distribution, and (4) they have a remarkable safety record in the clinic. Optimizations made in plasmid design as well as in DNA vaccine delivery have greatly improved the immunogenicity of these vaccines. Here we describe the process of making a DNA vaccine to an EID pathogen and describe methods used for assessing the immunogenicity and protective efficacy of DNA vaccines in small animal models.


Assuntos
Doenças Transmissíveis Emergentes , Vacinas de DNA , Vacinas Virais , Animais , COVID-19 , Doenças Transmissíveis Emergentes/prevenção & controle , Humanos , Imunidade , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave , SARS-CoV-2 , Vacinas Atenuadas/imunologia , Vacinas de DNA/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
2.
Genes Cancer ; 12: 51-64, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33884106

RESUMO

Prostate cancer is a prevalent cancer in men and consists of both indolent and aggressive phenotypes. While active surveillance is recommended for the former, current treatments for the latter include surgery, radiation, chemo and hormonal therapy. It has been observed that the recurrence in the treated patients is high and results in castration resistant prostate cancer for which treatment options are limited. This scenario has prompted us to consider immunotherapy with synthetic DNA vaccines, as this approach can generate antigen-specific tumor-killing immune cells. Given the multifocal and heterogeneous nature of prostate cancer, we hypothesized that synthetic DNA vaccines targeting different prostate specific antigens are likely to induce broader and improved immunity who are at high risk as well as advanced clinical stage of prostate cancer, compared to a single antigen approach. Utilizing a bioinformatics approach, synthetic enhanced DNA vaccine (SEV) constructs were generated against STEAP1, PAP, PARM1, PSCA, PCTA and PSP94. Synthetic enhanced vaccines for prostate cancer antigens were shown to elicit antigen-specific immune responses in mice and the anti-tumor activity was evident in a prostate tumor challenge mouse model. These studies support further evaluation of the DNA tools for immunotherapy of prostate cancer and perhaps other cancers.

3.
Nucleic Acid Ther ; 30(6): 379-391, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32907467

RESUMO

Hyperammonemia is a dangerous life-threatening metabolic complication characterized by markedly elevated ammonia levels that can lead to irreversible brain damage if not carefully monitored. Current pharmacological treatment strategies available for hyperammonemia patients are suboptimal and associated with major side effects. In this study, we focus on developing and evaluating the in vivo delivery of novel DNA-encoded glutamine synthetase (GS) enzymes for the treatment of hyperammonemia. Direct in vivo delivered DNA-encoded GS enzyme was evaluated in ammonium acetate-induced hyperammonemia and thioacetamide-induced acute liver injury (ALI) models in C57BL/6 mice. In ammonium acetate-induced hyperammonemia model, we achieved a 30.5% decrease in blood ammonia levels 15 min postadministration of ammonium acetate, with DNA-encoded GS-treated group. Significant increase in survival was observed in ALI model with the treated mice. A comparison of the secreted versus intracellular DNA-encoded GS enzyme demonstrated similar increases in survival in the ALI model, with 40% mortality in the secreted enzymes and 30% mortality in the intracellular enzymes, as compared with 90% mortality in the control group. Direct in vivo delivery of DNA-encoded GS demonstrated important ammonia-lowering potential. These results provide the initial steps toward development of delivered DNA as a potential new approach to ammonia-lowering therapeutics.


Assuntos
DNA/farmacologia , Glutamato-Amônia Ligase/genética , Hiperamonemia/tratamento farmacológico , Fígado/efeitos dos fármacos , Amônia/metabolismo , Animais , Modelos Animais de Doenças , Glutamato-Amônia Ligase/farmacologia , Glutamina/metabolismo , Humanos , Hiperamonemia/metabolismo , Fígado/metabolismo , Camundongos
4.
Mol Ther Methods Clin Dev ; 18: 652-663, 2020 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-32802913

RESUMO

Arginase is a complex and unique enzyme that plays diverse roles in health and disease. By metabolizing arginine, it can shape the outcome of innate and adaptive immune responses. The immunomodulatory capabilities of arginase could potentially be applied for local immunosuppression or induction of immune tolerance. With the use of an enhanced DNA delivery approach, we designed and studied a DNA-encoded secretable arginase enzyme as a tool for immune modulation and evaluated its immunomodulatory function in vivo. Strong immunosuppression of cluster of differentiation 4 (CD4) and CD8 T cells, as well as macrophages and dendritic cells, was observed in vitro in the presence of an arginase-rich supernatant. To further evaluate the efficacy of DNA-encoded arginase on in vivo immunosuppression against an antigen, a cancer antigen vaccine model was used in the presence or absence of DNA-encoded arginase. Significant in vivo immunosuppression was observed in the presence of DNA-encoded arginase. The efficacy of this DNA-encoded arginase delivery was examined in a local, imiquimod-induced, psoriasis-like, skin-inflammation model. Pretreatment of animals with the synthetic DNA-encoded arginase led to significant decreases in skin acanthosis, proinflammatory cytokines, and costimulatory molecules in extracted macrophages and dendritic cells. These results draw attention to the potential of direct in vivo-delivered arginase to function as an immunomodulatory agent for treatment of local inflammation or autoimmune diseases.

5.
Nat Commun ; 11(1): 2601, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32433465

RESUMO

The coronavirus family member, SARS-CoV-2 has been identified as the causal agent for the pandemic viral pneumonia disease, COVID-19. At this time, no vaccine is available to control further dissemination of the disease. We have previously engineered a synthetic DNA vaccine targeting the MERS coronavirus Spike (S) protein, the major surface antigen of coronaviruses, which is currently in clinical study. Here we build on this prior experience to generate a synthetic DNA-based vaccine candidate targeting SARS-CoV-2 S protein. The engineered construct, INO-4800, results in robust expression of the S protein in vitro. Following immunization of mice and guinea pigs with INO-4800 we measure antigen-specific T cell responses, functional antibodies which neutralize the SARS-CoV-2 infection and block Spike protein binding to the ACE2 receptor, and biodistribution of SARS-CoV-2 targeting antibodies to the lungs. This preliminary dataset identifies INO-4800 as a potential COVID-19 vaccine candidate, supporting further translational study.


Assuntos
Antígenos Virais/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas de DNA/imunologia , Vacinas Virais/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Anticorpos Neutralizantes/imunologia , Antígenos Virais/química , Vacinas contra COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Mapeamento de Epitopos , Cobaias , Imunidade Humoral , Imunoglobulina G/imunologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Coronavírus da Síndrome Respiratória do Oriente Médio , Modelos Animais , Peptidil Dipeptidase A/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Vacinas Virais/química
6.
Hum Vaccin Immunother ; 16(9): 2156-2164, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32463327

RESUMO

Hepatitis B virus (HBV) causes a potentially life-threatening liver infection that frequently results in life-long chronic infection. HBV is responsible for 887,000 deaths each year, most resulting from chronic liver diseases and hepatocellular carcinoma. Presently, there are 250 million chronic HBV carriers worldwide who are at a high risk for developing cirrhosis and hepatocellular carcinoma (HCC). HCC is the most common type of liver cancer with a strong association with HBV infection. HBV transmission through blood transfusions and perinatal transfer from infected mother to child have been common routes of infection. In the present study, we describe the development of a synthetic DNA plasmid encoding an anti-HBV human monoclonal antibody specific for the common "a determinant region" of HBsAg of hepatitis B virus and demonstrate the ability of this platform at directing in vivo antibody expression. In vivo delivery of this DNA encoded monoclonal antibody (DMAb) plasmid in mice resulted in expression of human IgG over a period of one month following a single injection. Serum antibody was found to recognize the relevant conformational epitope from plasma purified native HBsAg as well as bound HBV in HepG2.2.15 cells. The serum DMAb efficiently neutralized HBV and prevented infection of HepaRG cells in vitro. Additional study of these HBV-DMAb as a possible therapy or immunoprophylaxis for HBV infection is warranted.


Assuntos
Carcinoma Hepatocelular , Hepatite B , Neoplasias Hepáticas , Animais , Anticorpos Monoclonais , DNA Viral/genética , Feminino , Hepatite B/prevenção & controle , Antígenos de Superfície da Hepatite B , Vírus da Hepatite B/genética , Transmissão Vertical de Doenças Infecciosas , Camundongos
7.
Proc Natl Acad Sci U S A ; 117(7): 3405-3414, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32005712

RESUMO

Drug targeting to inflammatory brain pathologies such as stroke and traumatic brain injury remains an elusive goal. Using a mouse model of acute brain inflammation induced by local tumor necrosis factor alpha (TNFα), we found that uptake of intravenously injected antibody to vascular cell adhesion molecule 1 (anti-VCAM) in the inflamed brain is >10-fold greater than antibodies to transferrin receptor-1 and intercellular adhesion molecule 1 (TfR-1 and ICAM-1). Furthermore, uptake of anti-VCAM/liposomes exceeded that of anti-TfR and anti-ICAM counterparts by ∼27- and ∼8-fold, respectively, achieving brain/blood ratio >300-fold higher than that of immunoglobulin G/liposomes. Single-photon emission computed tomography imaging affirmed specific anti-VCAM/liposome targeting to inflamed brain in mice. Intravital microscopy via cranial window and flow cytometry showed that in the inflamed brain anti-VCAM/liposomes bind to endothelium, not to leukocytes. Anti-VCAM/LNP selectively accumulated in the inflamed brain, providing de novo expression of proteins encoded by cargo messenger RNA (mRNA). Anti-VCAM/LNP-mRNA mediated expression of thrombomodulin (a natural endothelial inhibitor of thrombosis, inflammation, and vascular leakage) and alleviated TNFα-induced brain edema. Thus VCAM-directed nanocarriers provide a platform for cerebrovascular targeting to inflamed brain, with the goal of normalizing the integrity of the blood-brain barrier, thus benefiting numerous brain pathologies.


Assuntos
Anticorpos/administração & dosagem , Barreira Hematoencefálica/efeitos dos fármacos , Encefalite/tratamento farmacológico , Endotélio Vascular/efeitos dos fármacos , Nanomedicina/métodos , Animais , Barreira Hematoencefálica/imunologia , Encefalite/genética , Encefalite/imunologia , Endotélio Vascular/imunologia , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Camundongos , Receptores da Transferrina/genética , Receptores da Transferrina/imunologia , Trombomodulina/genética , Trombomodulina/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
8.
Methods Mol Biol ; 2033: 81-93, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31332749

RESUMO

Covalent conjugation of chemical moieties to antibodies has numerous applications, including antibody-drug conjugates, antibody conjugation for diagnostics, and more. Most nonspecific chemical conjugation methods ligate onto any of a number of sites on the antibody, leading to multiple conjugated species, many of which perturb antibody function. To solve these problems, we used CRISPR/Cas9-edited hybridomas to introduce a Sortase tag (LPXTG) and a Flag tag at the 3' end of the CH3 heavy chain region of a mouse monoclonal antibody. The Flag tag allows easy purification of the antibody, while the LPXTG is then acted on by the bacterial transpeptidase Sortase to site-specifically add on any of a number of chemical moieties that possess a triglycine repeat. This technique thus allows rapid production of an antibody onto which a wide array of chemical moieties can be site-specifically conjugated.


Assuntos
Anticorpos Monoclonais/genética , Edição de Genes/métodos , Engenharia Genética/métodos , Imunoconjugados/genética , Animais , Anticorpos Monoclonais/imunologia , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/imunologia , Humanos , Hibridomas/imunologia , Imunoconjugados/imunologia , Camundongos , Oligopeptídeos/genética , Oligopeptídeos/imunologia
9.
Mol Ther ; 27(1): 188-199, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30449662

RESUMO

Elevated low-density lipoprotein cholesterol (LDL-C) is one of the major contributors to cardiovascular heart disease (CHD), the leading cause of death worldwide. Due to severe side effects of statins, alternative treatment strategies are required for statin-intolerant patients. Monoclonal antibodies (mAbs) targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) have shown great efficacy in LDL-C reduction. Limitations for this approach include the need for multiple injections as well as increased costs associated with patient management. Here, we engineered a DNA-encoded mAb (DMAb) targeting PCSK9 (daPCSK9), as an alternative approach to protein-based lipid-lowering therapeutics, and we characterized its expression and activity. A single intramuscular administration of mouse daPCSK9 generated expression in vivo for over 42 days that corresponded with a substantial decrease of 28.6% in non-high-density lipoprotein cholesterol (non-HDL-C) and 10.3% in total cholesterol by day 7 in wild-type mice. Repeated administrations of the DMAb plasmid led to increasing expression, with DMAb levels of 7.5 µg/mL at day 62. daPCSK9 therapeutics may provide a novel, simple, less frequent, cost-effective approach to reducing LDL-C, either as a stand-alone therapy or in combination with other LDL-lowering therapeutics for synergistic effect.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Pró-Proteína Convertase 9/imunologia , Animais , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/terapia , LDL-Colesterol/sangue , Terapia Genética/métodos , Células HEK293 , Humanos , Camundongos , Plasmídeos/genética
10.
Biomaterials ; 185: 348-359, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273834

RESUMO

One of the goals of nanomedicine is targeted delivery of therapeutic enzymes to the sub-cellular compartments where their action is needed. Endothelial caveolae-derived endosomes represent an important yet challenging destination for targeting, in part due to smaller size of the entry aperture of caveolae (ca. 30-50 nm). Here, we designed modular, multi-molecular, ferritin-based nanocarriers with uniform size (20 nm diameter) for easy drug-loading and targeted delivery of enzymatic cargo to these specific vesicles. These nanocarriers targeted to caveolar Plasmalemmal Vesicle-Associated Protein (Plvap) deliver superoxide dismutase (SOD) into endosomes in endothelial cells, the specific site of influx of superoxide mediating by such pro-inflammatory signaling as some cytokines and lipopolysaccharide (LPS). Cell studies showed efficient internalization of Plvap-targeted SOD-loaded nanocarriers followed by dissociation from caveolin-containing vesicles and intracellular transport to endosomes. The nanocarriers had a profound protective anti-inflammatory effect in an animal model of LPS-induced inflammation, in agreement with the characteristics of their endothelial uptake and intracellular transport, indicating that these novel, targeted nanocarriers provide an advantageous platform for caveolae-dependent delivery of biotherapeutics.


Assuntos
Cavéolas/metabolismo , Portadores de Fármacos/metabolismo , Ferritinas/metabolismo , Nanopartículas/metabolismo , Superóxido Dismutase/administração & dosagem , Animais , Proteínas Arqueais/metabolismo , Archaeoglobus fulgidus/metabolismo , Linhagem Celular , Sistemas de Liberação de Medicamentos , Imunoconjugados/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Superóxido Dismutase/farmacocinética
11.
J Control Release ; 291: 106-115, 2018 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-30336167

RESUMO

Systemic administration of lipid nanoparticle (LNP)-encapsulated messenger RNA (mRNA) leads predominantly to hepatic uptake and expression. Here, we conjugated nucleoside-modified mRNA-LNPs with antibodies (Abs) specific to vascular cell adhesion molecule, PECAM-1. Systemic (intravenous) administration of Ab/LNP-mRNAs resulted in profound inhibition of hepatic uptake concomitantly with ~200-fold and 25-fold elevation of mRNA delivery and protein expression in the lungs compared to non-targeted counterparts. Unlike hepatic delivery of LNP-mRNA, Ab/LNP-mRNA is independent of apolipoprotein E. Vascular re-targeting of mRNA represents a promising, powerful, and unique approach for novel experimental and clinical interventions in organs of interest other than liver.


Assuntos
Apolipoproteínas E/metabolismo , Sistemas de Liberação de Medicamentos , Endotélio Vascular/metabolismo , Nanopartículas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/administração & dosagem , Administração Intravenosa , Animais , Linhagem Celular , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoconjugados/metabolismo , Camundongos Endogâmicos C57BL , RNA Mensageiro/farmacocinética , Distribuição Tecidual
12.
Adv Drug Deliv Rev ; 130: 90-112, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-30149885

RESUMO

Humoral and cellular host defense mechanisms including diverse phagocytes, leukocytes, and immune cells have evolved over millions of years to protect the body from microbes and other external and internal threats. These policing forces recognize engineered sub-micron drug delivery systems (DDS) as such a threat, and react accordingly. This leads to impediment of the therapeutic action, extensively studied and discussed in the literature. Here, we focus on side effects of DDS interactions with host defenses. We argue that for nanomedicine to reach its clinical potential, the field must redouble its efforts in understanding the interaction between drug delivery systems and the host defenses, so that we can engineer safer interventions with the greatest potential for clinical success.


Assuntos
Portadores de Fármacos/efeitos adversos , Portadores de Fármacos/química , Animais , Humanos , Nanomedicina , Nanopartículas/efeitos adversos , Nanopartículas/química , Tamanho da Partícula , Propriedades de Superfície
13.
Nat Commun ; 9(1): 2684, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29992966

RESUMO

Drug delivery by nanocarriers (NCs) has long been stymied by dominant liver uptake and limited target organ deposition, even when NCs are targeted using affinity moieties. Here we report a universal solution: red blood cell (RBC)-hitchhiking (RH), in which NCs adsorbed onto the RBCs transfer from RBCs to the first organ downstream of the intravascular injection. RH improves delivery for a wide range of NCs and even viral vectors. For example, RH injected intravenously increases liposome uptake in the first downstream organ, lungs, by ~40-fold compared with free NCs. Intra-carotid artery injection of RH NCs delivers >10% of the injected NC dose to the brain, ~10× higher than that achieved with affinity moieties. Further, RH works in mice, pigs, and ex vivo human lungs without causing RBC or end-organ toxicities. Thus, RH is a clinically translatable platform technology poised to augment drug delivery in acute lung disease, stroke, and several other diseases.


Assuntos
Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Eritrócitos/química , Nanopartículas/química , Adsorção , Animais , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Humanos , Pulmão/metabolismo , Pneumopatias/metabolismo , Pneumopatias/terapia , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Ratos , Suínos
14.
J Control Release ; 282: 13-24, 2018 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-29522833

RESUMO

Ferritin subunits of heavy and light polypeptide chains self-assemble into a spherical nanocage that serves as a natural transport vehicle for metals but can include diverse cargoes. Ferritin nanoparticles are characterized by remarkable stability, small and uniform size. Chemical modifications and molecular re-engineering of ferritin yield a versatile platform of nanocarriers capable of delivering a broad range of therapeutic and imaging agents. Targeting moieties conjugated to the ferritin external surface provide multivalent anchoring of biological targets. Here, we highlight some of the current work on ferritin as well as examine potential strategies that could be used to functionalize ferritin via chemical and genetic means to enable its utility in vascular drug delivery.


Assuntos
Vasos Sanguíneos , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Ferritinas/química , Nanopartículas/química , Animais , Antineoplásicos/administração & dosagem , Vasos Sanguíneos/diagnóstico por imagem , Vasos Sanguíneos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Humanos , Pulmão/irrigação sanguínea , Pulmão/diagnóstico por imagem , Pulmão/metabolismo , Modelos Moleculares , Neoplasias/irrigação sanguínea , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Vacinas/administração & dosagem
15.
Bioconjug Chem ; 29(4): 1209-1218, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29429330

RESUMO

Genetic incorporation of biologically orthogonal functional groups into macromolecules has the potential to yield efficient, controlled, reproducible, site-specific conjugation of affinity ligands, contrast agents, or therapeutic cargoes. Here, we applied this approach to ferritin, a ubiquitous iron-storage protein that self-assembles into multimeric nanocages with remarkable stability, size uniformity (12 nm), and endogenous capacity for loading and transport of a variety of inorganic and organic cargoes. The unnatural amino acid, 4-azidophenylalanine (4-AzF), was incorporated at different sites in the human ferritin light chain (hFTL) to allow site-specific conjugation of alkyne-containing small molecules or affinity ligands to the exterior surface of the nanocage. The optimal positioning of the 4-AzF residue was evaluated by screening a library of variants for the efficiency of copper-free click conjugation. One of the engineered ferritins, hFTL-5X, was found to accommodate ∼14 small-molecule fluorophores (AlexaFluor 488) and 3-4 IgG molecules per nanocage. Intravascular injection in mice of radiolabeled hFTL-5X carrying antibody to cell adhesion molecule ICAM-1, but not control IgG, enabled specific targeting to the lung due to high basal expression of ICAM-1 (43.3 ± 6.99 vs 3.48 ± 0.14%ID/g for Ab vs IgG). Treatment of mice with endotoxin known to stimulate inflammatory ICAM-1 overexpression resulted in 2-fold enhancement of pulmonary targeting (84.4 ± 12.89 vs 43.3 ± 6.99%ID/g). Likewise, injection of fluorescent, ICAM-targeted hFTL-5X nanocages revealed the effect of endotoxin by enhancement of near-infrared signal, indicating potential utility of this approach for both vascular targeting and imaging.


Assuntos
Azidas/química , Ferritinas/química , Corantes Fluorescentes/química , Imunoconjugados/química , Molécula 1 de Adesão Intercelular/análise , Imagem Óptica/métodos , Fenilalanina/análogos & derivados , Alcinos/síntese química , Alcinos/química , Animais , Azidas/síntese química , Química Click/métodos , Ferritinas/síntese química , Corantes Fluorescentes/síntese química , Humanos , Inflamação/diagnóstico por imagem , Pulmão/diagnóstico por imagem , Camundongos , Nanoestruturas/química , Fenilalanina/síntese química , Fenilalanina/química
16.
Sci Rep ; 8(1): 1760, 2018 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-29379029

RESUMO

Site-specific modification of antibodies has become a critical aspect in the development of next-generation immunoconjugates meeting criteria of clinically acceptable homogeneity, reproducibility, efficacy, ease of manufacturability, and cost-effectiveness. Using CRISPR/Cas9 genomic editing, we developed a simple and novel approach to produce site-specifically modified antibodies. A sortase tag was genetically incorporated into the C-terminal end of the third immunoglobulin heavy chain constant region (CH3) within a hybridoma cell line to manufacture antibodies capable of site-specific conjugation. This enabled an effective enzymatic site-controlled conjugation of fluorescent and radioactive cargoes to a genetically tagged mAb without impairment of antigen binding activity. After injection in mice, these immunoconjugates showed almost doubled specific targeting in the lung vs. chemically conjugated maternal mAb, and concomitant reduction in uptake in the liver and spleen. The approach outlined in this work provides a facile method for the development of more homogeneous, reproducible, effective, and scalable antibody conjugates for use as therapeutic and diagnostic tools.


Assuntos
Anticorpos Monoclonais/genética , Sistemas CRISPR-Cas/genética , Imunoconjugados/genética , Animais , Antígenos/genética , Linhagem Celular Tumoral , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Cisteína/genética , Humanos , Camundongos , Ratos , Reprodutibilidade dos Testes
17.
Bioconjug Chem ; 29(1): 56-66, 2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29200285

RESUMO

The conjugation of antibodies to drugs and drug carriers improves delivery to target tissues. Widespread implementation and effective translation of this pharmacologic strategy awaits the development of affinity ligands capable of a defined degree of modification and highly efficient bioconjugation without loss of affinity. To date, such ligands are lacking for the targeting of therapeutics to vascular endothelial cells. To enable site-specific, click-chemistry conjugation to therapeutic cargo, we used the bacterial transpeptidase, sortase A, to attach short azidolysine containing peptides to three endothelial-specific single chain antibody fragments (scFv). While direct fusion of a recognition motif (sortag) to the scFv C-terminus generally resulted in low levels of sortase-mediated modification, improved reaction efficiency was observed for one protein, in which two amino acids had been introduced during cloning. This prompted insertion of a short, semi-rigid linker between scFv and sortag. The linker significantly enhanced modification of all three proteins, to the extent that unmodified scFv could no longer be detected. As proof of principle, purified, azide-modified scFv was conjugated to the antioxidant enzyme, catalase, resulting in robust endothelial targeting of functional cargo in vitro and in vivo.


Assuntos
Química Click/métodos , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/farmacocinética , Sequência de Aminoácidos , Aminoaciltransferases/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Catalase/metabolismo , Linhagem Celular , Cisteína Endopeptidases/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/administração & dosagem , Molécula 1 de Adesão Intercelular/química , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Molécula-1 de Adesão Celular Endotelial a Plaquetas/administração & dosagem , Molécula-1 de Adesão Celular Endotelial a Plaquetas/química , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/farmacocinética , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/metabolismo , Distribuição Tecidual
18.
J Control Release ; 234: 115-23, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27210108

RESUMO

Controlled endothelial delivery of SOD may alleviate abnormal local surplus of superoxide involved in ischemia-reperfusion, inflammation and other disease conditions. Targeting SOD to endothelial surface vs. intracellular compartments is desirable to prevent pathological effects of external vs. endogenous superoxide, respectively. Thus, SOD conjugated with antibodies to cell adhesion molecule PECAM (Ab/SOD) inhibits pro-inflammatory signaling mediated by endogenous superoxide produced in the endothelial endosomes in response to cytokines. Here we defined control of surface vs. endosomal delivery and effect of Ab/SOD, focusing on conjugate size and targeting to PECAM vs. ICAM. Ab/SOD enlargement from about 100 to 300nm enhanced amount of cell-bound SOD and protection against extracellular superoxide. In contrast, enlargement inhibited endocytosis of Ab/SOD and diminished mitigation of inflammatory signaling of endothelial superoxide. In addition to size, shape is important: endocytosis of antibody-coated spheres was more effective than that of polymorphous antibody conjugates. Further, targeting to ICAM provides higher endocytic efficacy than targeting to PECAM. ICAM-targeted Ab/SOD more effectively mitigated inflammatory signaling by intracellular superoxide in vitro and in animal models, although total uptake was inferior to that of PECAM-targeted Ab/SOD. Therefore, both geometry and targeting features of Ab/SOD conjugates control delivery to cell surface vs. endosomes for optimal protection against extracellular vs. endosomal oxidative stress, respectively.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Endotélio Vascular/efeitos dos fármacos , Imunoconjugados/administração & dosagem , Molécula 1 de Adesão Intercelular/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Superóxido Dismutase/administração & dosagem , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/farmacologia , Endocitose/efeitos dos fármacos , Endossomos/metabolismo , Endotélio Vascular/metabolismo , Endotoxemia/tratamento farmacológico , Células Endoteliais da Veia Umbilical Humana , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Camundongos Endogâmicos C57BL , Nanopartículas/química , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/farmacocinética , Superóxido Dismutase/farmacologia , Superóxidos/metabolismo
19.
J Control Release ; 226: 229-37, 2016 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-26855052

RESUMO

Endothelial cells (EC) represent an important target for pharmacologic intervention, given their central role in a wide variety of human pathophysiologic processes. Studies in lab animal species have established that conjugation of drugs and carriers with antibodies directed to surface targets like the Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1, a highly expressed endothelial transmembrane protein) help to achieve specific therapeutic interventions in ECs. To translate such "vascular immunotargeting" to clinical practice, it is necessary to replace antibodies by advanced ligands that are more amenable to use in humans. We report the molecular design of a single chain variable antibody fragment (scFv) that binds with high affinity to human PECAM-1 and cross-reacts with its counterpart in rats and other animal species, allowing parallel testing in vivo and in human endothelial cells in microfluidic model. Site-specific modification of the scFv allows conjugation of protein cargo and liposomes, enabling their endothelial targeting in these models. This study provides a template for molecular engineering of ligands, enabling studies of drug targeting in animal species and subsequent use in humans.


Assuntos
Sistemas de Liberação de Medicamentos , Endotélio/imunologia , Lipossomos/imunologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Anticorpos de Cadeia Única/imunologia , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Linhagem Celular , Células Endoteliais/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/farmacocinética , Ratos , Anticorpos de Cadeia Única/administração & dosagem , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/farmacocinética
20.
Bioconjug Chem ; 27(3): 628-37, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26718023

RESUMO

Targeting nanocarriers to the endothelium, using affinity ligands to cell adhesion molecules such as ICAM-1 and PECAM-1, holds promise to improve the pharmacotherapy of many disease conditions. This approach capitalizes on the observation that antibody-targeted carriers of 100 nm and above accumulate in the pulmonary vasculature more effectively than free antibodies. Targeting of prospective nanocarriers in the 10-50 nm range, however, has not been studied. To address this intriguing issue, we conjugated monoclonal antibodies (Ab) to ICAM-1 and PECAM-1 or their single chain antigen-binding fragments (scFv) to ferritin nanoparticles (FNPs, size 12 nm), thereby producing Ab/FNPs and scFv/FNPs. Targeted FNPs retained their typical symmetric core-shell structure with sizes of 20-25 nm and ∼4-5 Ab (or ∼7-9 scFv) per particle. Ab/FNPs and scFv/FNPs, but not control IgG/FNPs, bound specifically to cells expressing target molecules and accumulated in the lungs after intravenous injection, with pulmonary targeting an order of magnitude higher than free Ab. Most intriguing, the targeting of Ab/FNPs to ICAM-1, but not PECAM-1, surpassed that of larger Ab/carriers targeted by the same ligand. These results indicate that (i) FNPs may provide a platform for targeting endothelial adhesion molecules with carriers in the 20 nm size range, which has not been previously reported; and (ii) ICAM-1 and PECAM-1 (known to localize in different domains of endothelial plasmalemma) differ in their accessibility to circulating objects of this size, common for blood components and nanocarriers.


Assuntos
Endotélio Vascular/metabolismo , Ferritinas/química , Nanopartículas , Animais , Microscopia Eletrônica de Transmissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...