Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Blood Adv ; 7(19): 5877-5889, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37428863

RESUMO

The importance of the stromal microenvironment in chronic lymphocytic leukemia (CLL) pathogenesis and drug resistance is well established. Despite recent advances in CLL therapy, identifying novel ways to disrupt interactions between CLL and its microenvironment may identify new combination partners for the drugs currently in use. To understand the role of microenvironmental factors on primary CLL cells, we took advantage of an observation that conditioned media (CM) collected from stroma was protective of CLL cells from spontaneous cell death ex vivo. The cytokine in the CM-dependent cells that most supports CLL survival in short-term ex vivo culture was CCL2. Pretreatment of CLL cells with anti-CCL2 antibody enhanced venetoclax-mediated killing. Surprisingly, we found a group of CLL samples (9/23 cases) that are less likely to undergo cell death in the absence of CM support. Functional studies revealed that CM-independent (CMI) CLL cells are less sensitive to apoptosis than conventional stroma-dependent CLL. In addition, a majority of the CMI CLL samples (80%) harbored unmutated immunoglobulin heavy-chain variable (IGHV) region. Bulk-RNA sequence analysis revealed upregulation of the focal adhesion and RAS signaling pathways in this group, along with expression of fms-like tyrosine kinase 3 (FLT3) and CD135. Treatment with FLT3 inhibitors caused a significant reduction in cell viability among CMI samples. In summary, we were able to discriminate and target 2 biologically distinct subgroups of CLL based on CM dependence with distinct microenvironmental vulnerabilities.


Assuntos
Leucemia Linfocítica Crônica de Células B , Humanos , Leucemia Linfocítica Crônica de Células B/patologia , Meios de Cultivo Condicionados/farmacologia , Tirosina Quinase 3 Semelhante a fms/uso terapêutico , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/uso terapêutico , Transdução de Sinais , Microambiente Tumoral
2.
Front Immunol ; 12: 728694, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646268

RESUMO

Monoclonal antibodies (mAbs) are an important class of therapeutics used to treat cancer, inflammation, and infectious diseases. Identifying highly developable mAb sequences in silico could greatly reduce the time and cost required for therapeutic mAb development. Here, we present position-specific scoring matrices (PSSMs) for antibody framework mutations developed using baseline human antibody repertoire sequences. Our analysis shows that human antibody repertoire-based PSSMs are consistent across individuals and demonstrate high correlations between related germlines. We show that mutations in existing therapeutic antibodies can be accurately predicted solely from baseline human antibody sequence data. We find that mAbs developed using humanized mice had more human-like FR mutations than mAbs originally developed by hybridoma technology. A quantitative assessment of entire framework regions of therapeutic antibodies revealed that there may be potential for improving the properties of existing therapeutic antibodies by incorporating additional mutations of high frequency in baseline human antibody repertoires. In addition, high frequency mutations in baseline human antibody repertoires were predicted in silico to reduce immunogenicity in therapeutic mAbs due to the removal of T cell epitopes. Several therapeutic mAbs were identified to have common, universally high-scoring framework mutations, and molecular dynamics simulations revealed the mechanistic basis for the evolutionary selection of these mutations. Our results suggest that baseline human antibody repertoires may be useful as predictive tools to guide mAb development in the future.


Assuntos
Anticorpos Monoclonais/genética , Desenvolvimento de Medicamentos , Epitopos de Linfócito T/genética , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/genética , Mutação , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Análise Mutacional de DNA , Bases de Dados Genéticas , Aprovação de Drogas , Estabilidade de Medicamentos , Epitopos de Linfócito T/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/uso terapêutico , Modelos Genéticos , Simulação de Dinâmica Molecular , Estabilidade Proteica , Estados Unidos , United States Food and Drug Administration
3.
Biomolecules ; 10(10)2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-33003418

RESUMO

Multiple myeloma (MM) is a B-cell malignancy that is incurable for a majority of patients. B-cell maturation antigen (BCMA) is a lineage-restricted differentiation protein highly expressed in multiple myeloma cells but not in other normal tissues except normal plasma B cells. Due to the restricted expression and being a cell surface membrane protein, BCMA is an ideal target for immunotherapy approaches in MM. Recombinant immunotoxins (RITs) are a novel class of protein therapeutics that are composed of the Fv or Fab portion of an antibody fused to a cytotoxic agent. RITs were produced by expressing plasmids encoding the components of the anti-BCMA RITs in E. coli followed by inclusion body preparation, solubilization, renaturation, and purification by column chromatography. The cytotoxic activity of RITs was tested in vitro by WST-8 assays using BCMA expressing cell lines and on cells isolated from MM patients. The in vivo efficacy of RITs was tested in a xenograft mouse model using BCMA expressing multiple myeloma cell lines. Anti-BCMA recombinant immunotoxins are very effective in killing myeloma cell lines and cells isolated from myeloma patients expressing BCMA. Two mouse models of myeloma showed that the anti-BCMA immunotoxins can produce a long-term complete response and warrant further preclinical development.


Assuntos
Antígeno de Maturação de Linfócitos B/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Imunotoxinas/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Animais , Antígeno de Maturação de Linfócitos B/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/uso terapêutico , Imunotoxinas/imunologia , Camundongos , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Biomolecules ; 10(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32121592

RESUMO

The constant region of immunoglobulin (Ig) G antibodies is responsible for their effector immune mechanism and prolongs serum half-life, while the fragment variable (Fv) region is responsible for cellular or tissue targeting. Therefore, antibody engineering for cancer therapeutics focuses on both functional efficacy of the constant region and tissue- or cell-specificity of the Fv region. In the functional aspect of therapeutic purposes, antibody engineers in both academia and industry have capitalized on the constant region of different IgG subclasses and engineered the constant region to enhance therapeutic efficacy against cancer, leading to a number of successes for cancer patients in clinical settings. In this article, we review IgG subclasses for cancer therapeutics, including i) IgG1, ii) IgG2, 3, and 4, iii) recent findings on Fc receptor functions, and iv) future directions of reprogramming the constant region of IgG to maximize the efficacy of antibody drug molecules in cancer patients.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoconjugados , Regiões Constantes de Imunoglobulina , Imunoglobulina G , Neoplasias/tratamento farmacológico , Engenharia de Proteínas , Imunoconjugados/genética , Imunoconjugados/uso terapêutico , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/uso terapêutico , Imunoglobulina G/genética , Imunoglobulina G/uso terapêutico , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/uso terapêutico , Neoplasias/metabolismo
5.
Methods Enzymol ; 629: 383-399, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31727250

RESUMO

The programmed death 1 (PD-1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) are negative regulators of T-cell immune function. Removal of these "brakes" in T cells results in increased activation of the immune system and controlling and eradicating tumor. The development of immune checkpoint inhibitors (ICIs) is a revolutionary milestone in tumor immunotherapy. Obtaining the atomic structure of the human immune checkpoint receptor/ICI therapeutic antibody complex is essential for understanding its inhibition mechanism and the rational design of improved biotherapeutics. In this chapter, we describe the methods for efficient production of extracellular domain of human immune checkpoint receptors and Fv fragments of ICI therapeutic antibodies in milligram quantities sufficient for structural studies, taking examples of the PD-1/pembrolizumab Fv and CTLA-4-ipilimumab Fv complexes.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Região Variável de Imunoglobulina/farmacologia , Neoplasias/tratamento farmacológico , Difração de Raios X/métodos , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/uso terapêutico , Medicamentos Biossimilares/química , Medicamentos Biossimilares/farmacologia , Medicamentos Biossimilares/uso terapêutico , Antígeno CTLA-4/antagonistas & inibidores , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Antígeno CTLA-4/isolamento & purificação , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/uso terapêutico , Ipilimumab/química , Ipilimumab/farmacologia , Ipilimumab/uso terapêutico , Modelos Moleculares , Neoplasias/imunologia , Neoplasias/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/isolamento & purificação , Domínios Proteicos/genética , Domínios Proteicos/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
6.
J Allergy Clin Immunol ; 144(2): 514-523.e5, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30995457

RESUMO

BACKGROUND: Variable heavy chain (VH) family frameworks (FWRs) have been reported to affect antibody receptor and superantigen binding; however, such effects in IgE remain largely unknown. Given that VH family biases have been previously reported in IgE of certain allergies, there is a need to investigate this phenomenon for biotechnological and therapeutic purposes. OBJECTIVE: We sought to investigate the effects of VH families on IgE interaction with FcεRIα, anti-IgE omalizumab, antigen, and superantigen protein A (spA) by using the pertuzumab and trastuzumab IgE models. METHODS: Pertuzumab VH1-VH7 family variants of IgE with the same complementarity-determining regions were investigated with regard to their binding interactions to FcεRIα, Her2, omalizumab, and spA. Notable FcεRIα-IgE observations were cross-checked against appropriate trastuzumab IgE VH variants. Computational structural modeling and simulations were also performed for insight into the mechanism of interactions with various VH FWRs. RESULTS: The pertuzumab VH5 IgE variant, but not the trastuzumab VH5 IgE, was found to interact with FcεRIα significantly longer than the respective VH family variants within each model antibody. No significant differences in interaction were found between IgE and omalizumab for the pertuzumab VH variants. Although trastuzumab VH3 interacted with spA, none of our pertuzumab VH variants, including VH3, associated with spA. CONCLUSION: We found unexpected varying allosteric communications caused by the VH family FWRs to the FcεRIα-, Her2-, and spA-binding regions of pertuzumab IgE, with implications for use of IgE/anti-IgE therapeutics to treat allergy and IgE therapeutics in allergo-oncology.


Assuntos
Antígenos de Bactérias/química , Imunoglobulina E/química , Cadeias Pesadas de Imunoglobulinas/química , Região Variável de Imunoglobulina/imunologia , Receptores de IgE/química , Superantígenos/química , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Antígenos de Bactérias/imunologia , Humanos , Hipersensibilidade/imunologia , Hipersensibilidade/terapia , Imunoglobulina E/imunologia , Imunoglobulina E/uso terapêutico , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/uso terapêutico , Imunoterapia , Omalizumab/química , Omalizumab/imunologia , Receptores de IgE/imunologia , Superantígenos/imunologia , Trastuzumab/química , Trastuzumab/imunologia
8.
Int Rev Immunol ; 37(1): 69-76, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29182399

RESUMO

Camelid heavy-chain variable domains (VHHs) are the smallest, intact, antigen-binding units to occur in nature. VHHs possess high degrees of solubility and robustness enabling generation of multivalent constructs with increased avidity - characteristics that mark their superiority to other antibody fragments and monoclonal antibodies. Capable of effectively binding to molecular targets inaccessible to classical immunotherapeutic agents and easily produced in microbial culture, VHHs are considered promising tools for pharmaceutical biotechnology. With the aim to demonstrate the perspective and potential of VHHs for the development of prophylactic and therapeutic drugs to target diseases caused by bacterial and viral infections, this review article will initially describe the structural features that underlie the unique properties of VHHs and explain the methods currently used for the selection and recombinant production of pathogen-specific VHHs, and then thoroughly summarize the experimental findings of five distinct studies that employed VHHs as inhibitors of host-pathogen interactions or neutralizers of infectious agents. Past and recent studies suggest the potential of camelid heavy-chain variable domains as a novel modality of immunotherapeutic drugs and a promising alternative to monoclonal antibodies. VHHs demonstrate the ability to interfere with bacterial pathogenesis by preventing adhesion to host tissue and sequestering disease-causing bacterial toxins. To protect from viral infections, VHHs may be employed as inhibitors of viral entry by binding to viral coat proteins or blocking interactions with cell-surface receptors. The implementation of VHHs as immunotherapeutic agents for infectious diseases is of considerable potential and set to contribute to public health in the near future.


Assuntos
Infecções Bacterianas/prevenção & controle , Camelidae/imunologia , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Viroses/prevenção & controle , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/terapia , Camelidae/genética , Cárie Dentária/microbiologia , Cárie Dentária/terapia , Diarreia/microbiologia , Diarreia/prevenção & controle , Infecções por Escherichia coli/prevenção & controle , Humanos , Imunização Passiva , Cadeias Pesadas de Imunoglobulinas/biossíntese , Cadeias Pesadas de Imunoglobulinas/genética , Região Variável de Imunoglobulina/biossíntese , Região Variável de Imunoglobulina/genética , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/toxicidade , Poliomielite/prevenção & controle , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapêutico , Infecções por Vírus Respiratório Sincicial/terapia , Streptococcus mutans/imunologia , Viroses/imunologia , Viroses/terapia
9.
PLoS One ; 11(9): e0162351, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27603013

RESUMO

Species A Rotaviruses (RVA) remain a leading cause of mortality in children under 5 years of age. Current treatment options are limited. We assessed the efficacy of two VP6-specific llama-derived heavy chain antibody fragments (VHH) -2KD1 and 3B2- as an oral prophylactic and therapeutic treatment against RVA-induced diarrhea in a neonatal mouse model inoculated with virulent murine RVA (ECw, G16P[16]I7). Joint therapeutic administration of 2KD1+3B2 (200 µg/dose) successfully reduced diarrhea duration, RVA infection severity and virus shedding in feces. While the same dose of 2KD1 or 3B2 (200 µg) significantly reduced duration of RVA-induced diarrhea, 2KD1 was more effective in diminishing the severity of intestinal infection and RVA shedding in feces, perhaps because 2KD1 presented higher binding affinity for RVA particles than 3B2. Neither prophylactic nor therapeutic administration of the VHH interfered with the host's humoral immune response against RVA. When 2KD1 (200 µg) was administered after diarrhea development, it also significantly reduced RVA intestinal infection and fecal shedding. Host antibody responses against the oral VHH treatment were not detected, nor did viral escape mutants. Our findings show that oral administration of anti-VP6 VHH constitute, not only an effective prophylactic treatment against RVA-associated diarrhea, but also a safe therapeutic tool against RVA infection, even once diarrhea is present. Anti-VP6 VHH could be used complementary to ongoing vaccination, especially in populations that have shown lower immunization efficacy. These VHH could also be scaled-up to develop pediatric medication or functional food like infant milk formulas that might help treat RVA diarrhea.


Assuntos
Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Infecções por Rotavirus/tratamento farmacológico , Infecções por Rotavirus/virologia , Rotavirus/fisiologia , Animais , Animais Recém-Nascidos , Camelídeos Americanos , Diarreia/tratamento farmacológico , Diarreia/virologia , Fezes/virologia , Concentração de Íons de Hidrogênio , Imunidade Humoral/imunologia , Cadeias Pesadas de Imunoglobulinas/administração & dosagem , Região Variável de Imunoglobulina/administração & dosagem , Intestinos/patologia , Intestinos/virologia , Camundongos Endogâmicos BALB C , Mutação/genética , Filogenia , Proteólise , Infecções por Rotavirus/imunologia , Vírion/metabolismo , Eliminação de Partículas Virais
10.
Curr Drug Discov Technol ; 11(1): 28-40, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23863097

RESUMO

The smallest independently folded antibody fragments, the domains, are emerging as promising scaffolds for candidate therapeutics and diagnostics that bind specifically targets of interest. The discovery of such binders is based on several technologies including structure-based design and generation of libraries of mutants displayed on phage or yeast, next-generation sequencing for diversity analysis, panning and screening of the libraries, affinity maturation of selected binders, and their expression, purification, and characterization for specific binding, function, and aggregation propensity. In this review, we describe these technologies as applied for the generation of engineered antibody domains (eAds), especially those derived from the human immunoglobulin heavy chain variable region (VH) and the second domain of IgG1 heavy chain constant region (CH2) as potential candidate therapeutics and diagnostics, and discuss examples of eAds against HIV-1 and cancer-related proteins.


Assuntos
Anticorpos , Região Variável de Imunoglobulina , Animais , Anticorpos/química , Anticorpos/uso terapêutico , Descoberta de Drogas , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/uso terapêutico , Engenharia de Proteínas , Estrutura Terciária de Proteína
12.
Antiviral Res ; 92(3): 389-407, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21939690

RESUMO

In 1989, a new type of antibody was identified, first in the sera of dromedaries and later also in all other species of the Camelidae family. These antibodies do not contain a light chain and also lack the first constant heavy domain. Today it is still unclear what the evolutionary advantage of such heavy chain-only antibodies could be. In sharp contrast, the broad applicability of the isolated variable antigen-binding domains (VHH) was rapidly recognized, especially for the development of therapeutic proteins, called Nanobodies(®). Here we summarize first some of the unique characteristics and features of VHHs. These will next be described in the context of different experimental therapeutic applications of Nanobodies against different viruses: HIV, Hepatitis B virus, influenza virus, Respiratory Syncytial virus, Rabies virus, FMDV, Poliovirus, Rotavirus, and PERVs. Next, the diagnostic application of VHHs (Vaccinia virus, Marburg virus and plant Tulip virus X), as well as an industrial application (lytic lactococcal 936 phage) will be described. In addition, the described data show that monovalent Nanobodies can possess unique characteristics not observed with conventional antibodies. The straightforward formatting into bivalent, multivalent, and/or multispecific Nanobodies allowed tailoring molecules for potency and cross-reactivity against viral targets with high sequence diversity.


Assuntos
Anticorpos Antivirais/uso terapêutico , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/uso terapêutico , Viroses/diagnóstico , Viroses/tratamento farmacológico , Vírus/imunologia , Animais , Anticorpos Antivirais/imunologia , Camelídeos Americanos/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Viroses/imunologia
13.
mBio ; 2(1): e00345-10, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21304166

RESUMO

The structural and functional significance of somatic insertions and deletions in antibody chains is unclear. Here, we demonstrate that a naturally occurring three-amino-acid insertion within the influenza virus-specific human monoclonal antibody 2D1 heavy-chain variable region reconfigures the antibody-combining site and contributes to its high potency against the 1918 and 2009 pandemic H1N1 influenza viruses. The insertion arose through a series of events, including a somatic point mutation in a predicted hot-spot motif, introduction of a new hot-spot motif, a molecular duplication due to polymerase slippage, a deletion due to misalignment, and additional somatic point mutations. Atomic resolution structures of the wild-type antibody and a variant in which the insertion was removed revealed that the three-amino-acid insertion near the base of heavy-chain complementarity-determining region (CDR) H2 resulted in a bulge in that loop. This enlarged CDR H2 loop impinges on adjacent regions, causing distortion of the CDR H1 architecture and its displacement away from the antigen-combining site. Removal of the insertion restores the canonical structure of CDR H1 and CDR H2, but binding, neutralization activity, and in vivo activity were reduced markedly because of steric conflict of CDR H1 with the hemagglutinin antigen.


Assuntos
Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/genética , Anticorpos Antivirais/imunologia , Sítios de Ligação de Anticorpos , Mutagênese Insercional , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/uso terapêutico , Sequência de Bases , Linhagem Celular , Feminino , Humanos , Cadeias Pesadas de Imunoglobulinas/química , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Pesadas de Imunoglobulinas/uso terapêutico , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/uso terapêutico , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/tratamento farmacológico , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica
14.
J Biol Chem ; 286(5): 3707-16, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-20980262

RESUMO

Interfering intracellular antibodies are valuable for biological studies as drug surrogates and as potential macromolecular drugs per se. Their application is still limited because of the difficulty of acquisition of functional intracellular antibodies. We describe the use of the new intracellular antibody capture procedure (IAC(3)) to facilitate direct isolation of functional single domain antibody fragments using four independent target molecules (LMO2, TP53, CRAF1, and Hoxa9) from a set of diverse libraries. Initially, these have variability in only one of the three antigen-binding CDR regions of VH or VL and first round single domains are affinity matured by iterative randomization of the two other CDRs and reselection. We highlight the approach using a single domain binding to LMO2 protein. Our results show that interfering with LMO2 protein function demonstrates a role specifically in erythroid differentiation, confirm a necessary and sufficient function for LMO2 as a cancer therapy target in T-cell neoplasia and allowed for the first time production of soluble recombinant LMO2 protein by co-expression with intracellular domain antibodies. Co-crystallization of LMO2 and the anti-LMO2 VH protein was successful. These results demonstrate that this third generation IAC(3) offers a robust toolbox for various biomedical applications and consolidates functional features of the LMO2 protein complex, which includes the importance of Lmo2-Ldb1 protein interaction.


Assuntos
Proteínas de Ligação a DNA/imunologia , Região Variável de Imunoglobulina/farmacologia , Metaloproteínas/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Anticorpos , Antineoplásicos/imunologia , Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Células Eritroides , Cadeias Pesadas de Imunoglobulinas , Região Variável de Imunoglobulina/uso terapêutico , Proteínas com Domínio LIM , Metaloproteínas/metabolismo , Metaloproteínas/fisiologia , Camundongos , Biblioteca de Peptídeos , Ligação Proteica/efeitos dos fármacos
15.
Zhonghua Yi Xue Za Zhi ; 89(20): 1411-5, 2009 May 26.
Artigo em Chinês | MEDLINE | ID: mdl-19671337

RESUMO

OBJECTIVE: To study the effect of anti-type IV collagenase single-chain antibody scFv (3G11) on bleomycin (BLM)-induced pulmonary fibrosis. METHODS: C57BL/6 mice were divided into six groups (blank, saline control, BLM, low-dose treatment, intermediate-dose treatment, and high-dose treatment), the animal model of pulmonary fibrosis was induced with 400 ng/g intratracheal BLM. The day that treatment groups were injected with BLM was marked Day 0. Then at Days 1 - 7, scFv (3G11) was injected once intraperitoneally each day with three dosage [low-dose treatment (15 microg/g), intermediate-dose treatment (30 microg/g) and high-dose treatment (45 microg/g)]. At Day 21, 6 mice of each group (saline control, BLM, intermediate-dose treatment) were sacrificed and pathomorphological changes of left lungs evaluated with HE stained sections. Meanwhile, 6 mice of each group (blank, saline control, BLM, low-dose treatment, intermediate-dose treatment, and high-dose treatment) were sacrificed, the content of hydroxyproline was detected to measure the degree of collagen deposition. Macrophages were extracted from murine abdominal cavity and primarily cultured. And the inhibition of matrix metalloproteinase (MMP)-2 and MMP-9 excretion by different concentrations of scFv (3G11) (0, 15, 30, 45, and 60 micromol/L) was determined by gelatin zymography. RESULTS: By image analysis, the degree of fibrosis in the lungs of treated group was lighter than that of the BLM group. The percentages of the area of interalveolar septum (45.3% +/- 3.2%) and the number of nucleated cells (451 +/- 47) of treated group were remarkably reduced as compared with the BLM group (59.0% +/- 3.0%, 599 +/- 42, both P < 0.01). The content of hydroxyproline of the intermediate-dose treatment group [(0.82 +/- 0.05) microg/mg] and high-dose treatment group [(0.80 +/- 0.03) microg/mg] was lower than that of the BLM group [(0.92 +/- 0.07) microg/mg, P < 0.05, P < 0.01]. The result of gelatin zymography demonstrated that the excretion of MMP-2 and MMP-9 by macrophage was inhibited by scFv (3G11) at the concentration of 15 micromol/L. CONCLUSION: Single-chain antibody directed against type IV collagenase might inhibit the development of bleomycin-induced pulmonary fibrosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Pulmão/patologia , Fibrose Pulmonar/patologia , Fibrose Pulmonar/prevenção & controle , Animais , Bleomicina/efeitos adversos , Região Variável de Imunoglobulina/uso terapêutico , Pulmão/metabolismo , Masculino , Metaloproteinase 2 da Matriz/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/imunologia , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente
16.
Mol Interv ; 9(3): 119-24, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19592672

RESUMO

Immunotherapy for treating illicit drug abuse is a rapidly advancing field. There are currently two major approaches to developing drug-specific immunotherapies: active and passive. Active immunotherapy involves conjugating a drug-like hapten to a carrier protein and using traditional immunization approaches to generate a drug-specific immune response in the patient. In contrast, passive immunotherapy utilizes preformed monoclonal antibodies. Whether generated by active immunization or delivered passively, antibodies act as pharmacokinetic antagonists by binding the drug in the blood-stream and reducing the amount and rate of drug delivery to receptors in the brain. A newly emerging technology in anti-drug immunotherapy is the use of antibody fragments, or scFvs, rather than intact immunoglobulin G (IgG). These scFvs can retain the same binding properties as the original mAbs, and are onethird the molecular weight, providing a scaffold for creating antibody treatments with more customizable properties. Another nascent area of research utilizing the scFv scaffold is in creating drug-specific scFv-nanoparticle conjugates. These conjugates could improve upon current drug-specific antibody paradigms by increasing multivalency and allowing pharmacokinetic customization, while avoiding interactions with endogenous antibody receptor pathways. These parallel approaches to immunotherapy are moving rapidly toward the clinic and may soon provide new therapies for treating drug abuse.


Assuntos
Desenho de Fármacos , Região Variável de Imunoglobulina/uso terapêutico , Imunoterapia , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Transtornos Relacionados ao Uso de Substâncias/imunologia , Animais , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia , Imunoterapia/economia , Transtornos Relacionados ao Uso de Substâncias/economia
17.
Vaccine ; 27(39): 5383-92, 2009 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-19596413

RESUMO

Interleukin-12 (IL-12), despite exerting antitumor activity, has limited therapeutic uses due to its systemic toxicity. Since HER2 (also known as ErbB-2, neu, and HER2/neu) is frequently overexpressed on cancer cells, HER2-targeted delivery of IL-12 to tumors may be a promising strategy for enhancing antitumor immunity. Here we showed that intramuscular electrogene transfer of an expression vector encoding a fusion protein antiHER2scFv-IL12, which consists of antiHER2 single-chain variable fragment (scFv) and single-chain IL-12, significantly retarded tumor growth and prolonged the survival in a syngeneic bladder tumor model. Elevated IL-12 and interferon-gamma (IFN-gamma) levels, increased infiltration of CD4(+) and CD8(+) T cells, and reduced vascular endothelial growth factor (VEGF) expression in the tumors, as well as enhanced cytolytic activity of splenocytes were noted in the treated mice. Our results suggest that this approach may be effective for the treatment of HER2-overexpressing tumors.


Assuntos
Terapia Genética/métodos , Região Variável de Imunoglobulina/uso terapêutico , Interleucina-12/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Receptor ErbB-2/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Humanos , Região Variável de Imunoglobulina/imunologia , Interferon gama/imunologia , Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos C3H , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Baço/citologia , Baço/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Cancer Biol Ther ; 8(8): 707-13, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19417562

RESUMO

T-cell costimulatory molecules such as 4-1BB may provide a distinct and important signal for promoting positive immune regulation. 4-1BB is thought to have potential use as a cancer immunotherapeutic drug. In our previous study, a nonreplicative adenovirus (Ad.4-1BB scFv) carrying single-chain Fv fragments (scFv) specific for the 4-1BB gene (anti-4-1BB scFv) possessed remarkable in vivo anti-hepatoma efficacy. However, monotherapy achieved by triggering 4-1BB signaling was not sufficient to induce eradicative antitumor activities in low immunogenic tumors. It is of great interest to explore any possible synergistic antitumor effect of 4-1BB signaling combined with low dose cyclophosphamide (CTX), which is well documented to inhibit the suppressive capability of regulatory T-cells in mice and humans. In the present study, recombinant nonreplicative adenoviruses carrying an anti-4-1BB scFv gene were generated, characterized and explored for their stimulation of antilung tumor (TC-1) immunity in immunocompetent C57BL/6 mice. Compared to adenovirus and cyclophosphamide alone, adenovirus-mediated anti-4-1BB scFv in combination with low dose CTX treatment could obviously augment the antitumor activity, in which some established TC-1 tumors were eradicated and the survival of mice was significantly extended. This synergistic antitumor effect could be largely attributed to the depletion of T regulatory cells induced by low dose CTX. These findings may provide a new and promising strategy for immunogene therapy against cancer.


Assuntos
Ligante 4-1BB/genética , Antineoplásicos Alquilantes/uso terapêutico , Ciclofosfamida/uso terapêutico , Terapia Genética , Região Variável de Imunoglobulina/uso terapêutico , Neoplasias Pulmonares/terapia , Adenoviridae/genética , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Terapia Combinada , Sinergismo Farmacológico , Citometria de Fluxo , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia
19.
Biochem J ; 418(3): 507-15, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19000036

RESUMO

Prions are infectious proteins responsible for a group of fatal neurodegenerative diseases called TSEs (transmissible spongiform encephalopathies) or prion diseases. In mammals, prions reproduce themselves by recruiting the normal cellular protein PrP(C) and inducing its conversion into the disease-causing isoform denominated PrP(Sc). Recently, anti-prion antibodies have been shown to permanently cure prion-infected cells. However, the inability of full-length antibodies and proteins to cross the BBB (blood-brain barrier) hampers their use in the therapy of TSEs in vivo. Alternatively, brain delivery of prion-specific scFv (single-chain variable fragment) by AAV (adeno-associated virus) transfer delays the onset of the disease in infected mice, although protection is not complete. We investigated the anti-prion effects of a recombinant anti-PrP (D18) scFv by direct addition to scrapie-infected cell cultures or by infection with both lentivirus and AAV-transducing vectors. We show that recombinant anti-PrP scFv is able to reduce proteinase K-resistant PrP content in infected cells. In addition, we demonstrate that lentiviruses are more efficient than AAV in gene transfer of the anti-PrP scFv gene and in reducing PrP(Sc) content in infected neuronal cell lines. Finally, we have used a bioinformatic approach to construct a structural model of the D18scFv-PrP(C) complex. Interestingly, according to the docking results, Arg(PrP)(151) (Arg(151) from prion protein) is the key residue for the interactions with D18scFv, anchoring the PrP(C) to the cavity of the antibody. Taken together, these results indicate that combined passive and active immunotherapy targeting PrP might be promising strategies for therapeutic intervention in prion diseases.


Assuntos
Anticorpos/imunologia , Região Variável de Imunoglobulina/imunologia , Região Variável de Imunoglobulina/uso terapêutico , Imunoterapia/métodos , Proteínas PrPSc/imunologia , Doenças Priônicas/terapia , Animais , Células Cultivadas , Dependovirus/genética , Vetores Genéticos , Lentivirus/genética , Camundongos , Proteínas PrPC/imunologia , Príons , Scrapie/terapia , Proteínas Virais de Fusão/imunologia
20.
Immunol Lett ; 121(2): 110-5, 2008 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-19007817

RESUMO

Enterohaemorrhagic Escherichia coli (EHEC) O157:H7, a primary enteric pathogen, has been implicated in a wide spectrum of food/water-borne infectious diseases such as hemorrhagic colitis (HC) and hemolyticuremic syndrome (HUS). Effective treatments for EHEC O157:H7 induced disease are not available yet. Shiga-toxin 2 (Stx2) has been related to clinical manifestations of HUS, suggesting its critical role in pathology following infection with EHEC O157:H7. Here we report the development of four anti-Stx2-Monoclonal antibodies (McAbs) (5F3and 5C11 for Stx2A, and 1A4 and 1A5 for Stx2B), all of which have strong immunogenicity and neutralization activities in vitro and in vivo. The full-length cDNA coding for anti-Stx2A McAb, 5F3, was cloned and an engineered antibody was developed whose therapeutic effects were evaluated. Our data indicate that the engineered scFv together with two new McAbs may be applicable for the prevention and therapy of EHEC induced pathology.


Assuntos
Anticorpos Monoclonais/imunologia , Linfócitos B/metabolismo , Infecções por Escherichia coli/imunologia , Escherichia coli O157/imunologia , Região Variável de Imunoglobulina/imunologia , Proteínas Recombinantes de Fusão/imunologia , Toxina Shiga II/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Linfócitos B/imunologia , Linfócitos B/patologia , Infecções por Escherichia coli/complicações , Infecções por Escherichia coli/terapia , Escherichia coli O157/patogenicidade , Engenharia Genética , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/prevenção & controle , Humanos , Hibridomas , Imunização Passiva/tendências , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Região Variável de Imunoglobulina/uso terapêutico , Camundongos , Conformação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Toxina Shiga II/genética , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA