Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncoimmunology ; 11(1): 2113697, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36016696

RESUMO

The use of T-cell engagers (TCEs) to treat solid tumors is challenging, and several have been limited by narrow therapeutic windows due to substantial on-target, off-tumor toxicities due to the expression of low levels of target antigens on healthy tissues. Here, we describe TNB-928B, a fully human TCE that has a bivalent binding arm for folate receptor alpha (FRα) to selectively target FRα overexpressing tumor cells while avoiding the lysis of cells with low levels of FRα expression. The bivalent design of the FRα binding arm confers tumor selectivity due to low-affinity but high-avidity binding to high FRα antigen density cells. TNB-928B induces preferential effector T-cell activation, proliferation, and selective cytotoxic activity on high FRα expressing cells while sparing low FRα expressing cells. In addition, TNB-928B induces minimal cytokine release compared to a positive control TCE containing OKT3. Moreover, TNB-928B exhibits substantial ex vivo tumor cell lysis using endogenous T-cells and robust tumor clearance in vivo, promoting T-cell infiltration and antitumor activity in mouse models of ovarian cancer. TNB-928B exhibits pharmacokinetics similar to conventional antibodies, which are projected to enable favorable administration in humans. TNB-928B is a novel TCE with enhanced safety and specificity for the treatment of ovarian cancer.


Assuntos
Anticorpos Biespecíficos , Neoplasias Ovarianas , Animais , Anticorpos Biespecíficos/uso terapêutico , Carcinoma Epitelial do Ovário , Feminino , Receptor 1 de Folato/metabolismo , Receptor 1 de Folato/uso terapêutico , Humanos , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Linfócitos T
2.
MAbs ; 14(1): 2095949, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35867844

RESUMO

Cluster of differentiation 38 (CD38) is an ecto-enzyme expressed primarily on immune cells that metabolize nicotinamide adenine dinucleotide (NAD+) to adenosine diphosphate ribose or cyclic ADP-ribose and nicotinamide. Other substrates of CD38 include nicotinamide adenine dinucleotide phosphate and nicotinamide mononucleotide, a critical NAD+ precursor in the salvage pathway. NAD+ is an important coenzyme involved in several metabolic pathways and is a required cofactor for the function of sirtuins (SIRTs) and poly (adenosine diphosphate-ribose) polymerases. Declines in NAD+ levels are associated with metabolic and inflammatory diseases, aging, and neurodegenerative disorders. To inhibit CD38 enzyme activity and boost NAD+ levels, we developed TNB-738, an anti-CD38 biparatopic antibody that pairs two non-competing heavy chain-only antibodies in a bispecific format. By simultaneously binding two distinct epitopes on CD38, TNB-738 potently inhibited its enzymatic activity, which in turn boosted intracellular NAD+ levels and SIRT activities. Due to its silenced IgG4 Fc, TNB-738 did not deplete CD38-expressing cells, in contrast to the clinically available anti-CD38 antibodies, daratumumab, and isatuximab. TNB-738 offers numerous advantages compared to other NAD-boosting therapeutics, including small molecules, and supplements, due to its long half-life, specificity, safety profile, and activity. Overall, TNB-738 represents a novel treatment with broad therapeutic potential for metabolic and inflammatory diseases associated with NAD+ deficiencies.Abbreviations: 7-AAD: 7-aminoactinomycin D; ADCC: antibody dependent cell-mediated cytotoxicity; ADCP: antibody dependent cell-mediated phagocytosis; ADPR: adenosine diphosphate ribose; APC: allophycocyanin; cADPR: cyclic ADP-ribose; cDNA: complementary DNA; BSA: bovine serum albumin; CD38: cluster of differentiation 38; CDC: complement dependent cytotoxicity; CFA: Freund's complete adjuvant; CHO: Chinese hamster ovary; CCP4: collaborative computational project, number 4; COOT: crystallographic object-oriented toolkit; DAPI: 4',6-diamidino-2-phenylindole; DNA: deoxyribonucleic acid; DSC: differential scanning calorimetry; 3D: three dimensional; εNAD+: nicotinamide 1,N6-ethenoadenine dinucleotide; ECD: extracellular domain; EGF: epidermal growth factor; FACS: fluorescence activated cell sorting; FcγR: Fc gamma receptors; FITC: fluorescein isothiocyanate; HEK: human embryonic kidney; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IgG: immunoglobulin; IFA: incomplete Freund's adjuvant; IFNγ: Interferon gamma; KB: kinetic buffer; kDa: kilodalton; KEGG: kyoto encyclopedia of genes and genomes; LDH: lactate dehydrogenase; M: molar; mM: millimolar; MFI: mean fluorescent intensity; NA: nicotinic acid; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; NAM: nicotinamide; NGS: next-generation sequencing; NHS/EDC: N-Hydroxysuccinimide/ ethyl (dimethylamino propyl) carbodiimide; Ni-NTA: nickel-nitrilotriacetic acid; nL: nanoliter; NK: natural killer; NMN: nicotinamide mononucleotide; OD: optical density; PARP: poly (adenosine diphosphate-ribose) polymerase; PBS: phosphate-buffered saline; PBMC: peripheral blood mononuclear cell; PDB: protein data bank; PE: phycoerythrin; PISA: protein interfaces, surfaces, and assemblies: PK: pharmacokinetics; mol: picomolar; RNA: ribonucleic acid; RLU: relative luminescence units; rpm: rotations per minute; RU: resonance unit; SEC: size exclusion chromatography; SEM: standard error of the mean; SIRT: sirtuins; SPR: surface plasmon resonance; µg: microgram; µM: micromolar; µL: microliter.


Assuntos
NAD , Sirtuínas , Adenosina Difosfato Ribose/química , Adenosina Difosfato Ribose/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , ADP-Ribose Cíclica , Humanos , Imunoglobulina G , Leucócitos Mononucleares/metabolismo , NAD/química , NAD/metabolismo , NADP , Niacinamida , Mononucleotídeo de Nicotinamida , Ribose
3.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34088740

RESUMO

BACKGROUND: Therapeutic options currently available for metastatic castration-resistant prostate cancer (mCRPC) do not extend median overall survival >6 months. Therefore, the development of novel and effective therapies for mCRPC represents an urgent medical need. T cell engagers (TCEs) have emerged as a promising approach for the treatment of mCRPC due to their targeted mechanism of action. However, challenges remain in the clinic due to the limited efficacy of TCEs observed thus far in solid tumors as well as the toxicities associated with cytokine release syndrome (CRS) due to the usage of high-affinity anti-CD3 moieties such as OKT3. METHODS: Using genetically engineered transgenic rats (UniRat and OmniFlic) that express fully human IgG antibodies together with an NGS-based antibody discovery pipeline, we developed TNB-585, an anti-CD3xPSMA TCE for the treatment of mCRPC. TNB-585 pairs a tumor-targeting anti-PSMA arm together with a unique, low-affinity anti-CD3 arm in bispecific format. We tested TNB-585 in T cell-redirected cytotoxicity assays against PSMA+ tumor cells in both two-dimensional (2D) cultures and three-dimensional (3D) spheroids as well as against patient-derived prostate tumor cells. Cytokines were measured in culture supernatants to assess the ability of TNB-585 to induce tumor killing with low cytokine release. TNB-585-mediated T cell activation, proliferation, and cytotoxic granule formation were measured to investigate the mechanism of action. Additionally, TNB-585 efficacy was evaluated in vivo against C4-2 tumor-bearing NCG mice. RESULTS: In vitro, TNB-585 induced activation and proliferation of human T cells resulting in the killing of PSMA+ prostate tumor cells in both 2D cultures and 3D spheroids with minimal cytokine release and reduced regulatory T cell activation compared with a positive control antibody that contains the same anti-PSMA arm but a higher affinity anti-CD3 arm (comparable with OKT3). In addition, TNB-585 demonstrated potent efficacy against patient-derived prostate tumors ex vivo and induced immune cell infiltration and dose-dependent tumor regression in vivo. CONCLUSIONS: Our data suggest that TNB-585, with its low-affinity anti-CD3, may be efficacious while inducing a lower incidence and severity of CRS in patients with prostate cancer compared with TCEs that incorporate high-affinity anti-CD3 domains.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Antígenos de Superfície/imunologia , Complexo CD3/imunologia , Glutamato Carboxipeptidase II/imunologia , Imunoglobulina G/genética , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Macaca fascicularis , Masculino , Camundongos , Células PC-3 , Neoplasias de Próstata Resistentes à Castração/imunologia , Ratos , Ratos Transgênicos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Sci Rep ; 11(1): 10592, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34011961

RESUMO

The use of recombinant interleukin-2 (IL-2) as a therapeutic protein has been limited by significant toxicities despite its demonstrated ability to induce durable tumor-regression in cancer patients. The adverse events and limited efficacy of IL-2 treatment are due to the preferential binding of IL-2 to cells that express the high-affinity, trimeric receptor, IL-2Rαßγ such as endothelial cells and T-regulatory cells, respectively. Here, we describe a novel bispecific heavy-chain only antibody which binds to and activates signaling through the heterodimeric IL-2Rßγ receptor complex that is expressed on resting T-cells and NK cells. By avoiding binding to IL-2Rα, this molecule circumvents the preferential T-reg activation of native IL-2, while maintaining the robust stimulatory effects on T-cells and NK-cells in vitro. In vivo studies in both mice and cynomolgus monkeys confirm the molecule's in vivo biological activity, extended pharmacodynamics due to the Fc portion of the molecule, and enhanced safety profile. Together, these results demonstrate that the bispecific antibody is a safe and effective IL-2R agonist that harnesses the benefits of the IL-2 signaling pathway as a potential anti-cancer therapy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Subunidade gama Comum de Receptores de Interleucina/agonistas , Subunidade beta de Receptor de Interleucina-2/agonistas , Linfócitos/efeitos dos fármacos , Animais , Células CHO , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Subunidade gama Comum de Receptores de Interleucina/imunologia , Subunidade beta de Receptor de Interleucina-2/imunologia , Macaca fascicularis , Masculino , Camundongos Endogâmicos BALB C
5.
MAbs ; 13(1): 1890411, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33818299

RESUMO

The therapeutic potential of targeting CD19 in B cell malignancies has garnered attention in the past decade, resulting in the introduction of novel immunotherapy agents. Encouraging clinical data have been reported for T cell-based targeting agents, such as anti-CD19/CD3 bispecific T-cell engager blinatumomab and chimeric antigen receptor (CAR)-T therapies, for acute lymphoblastic leukemia and B cell non-Hodgkin lymphoma (B-NHL). However, clinical use of both blinatumomab and CAR-T therapies has been limited due to unfavorable pharmacokinetics (PK), significant toxicity associated with cytokine release syndrome and neurotoxicity, and manufacturing challenges. We present here a fully human CD19xCD3 bispecific antibody (TNB-486) for the treatment of B-NHL that could address the limitations of the current approved treatments. In the presence of CD19+ target cells and T cells, TNB-486 induces tumor cell lysis with minimal cytokine release, when compared to a positive control. In vivo, TNB-486 clears CD19+ tumor cells in immunocompromised mice in the presence of human peripheral blood mononuclear cells in multiple models. Additionally, the PK of TNB-486 in mice or cynomolgus monkeys is similar to conventional antibodies. This new T cell engaging bispecific antibody targeting CD19 represents a novel therapeutic that induces potent T cell-mediated tumor-cell cytotoxicity uncoupled from high levels of cytokine release, making it an attractive candidate for B-NHL therapy.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/efeitos dos fármacos , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Linfoma não Hodgkin/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacocinética , Anticorpos Monoclonais Humanizados/farmacocinética , Antígenos CD19/imunologia , Antineoplásicos Imunológicos/farmacocinética , Complexo CD3/antagonistas & inibidores , Complexo CD3/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Técnicas de Cocultura , Humanos , Células K562 , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/metabolismo , Macaca fascicularis , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
6.
MAbs ; 12(1): 1846900, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33228444

RESUMO

Transgenic animals incorporating human antibody genes are extremely attractive for drug development because they obviate subsequent antibody humanization procedures required for therapeutic translation. Transgenic platforms have previously been established using mice, but also more recently rats, chickens, and cows and are now in abundant use for drug development. However, rabbit-based antibody generation, with a strong track record for specificity and affinity, is able to include gene conversion mediated sequence diversification, thereby enhancing binder maturation and improving the variance/selection of output antibodies in a different way than in rodents. Since it additionally frequently permits good binder generation against antigens that are only weakly immunogenic in other organisms, it is a highly interesting species for therapeutic antibody generation. We report here on the generation, utilization, and analysis of the first transgenic rabbit strain for human antibody production. Through the knockout of endogenous IgM genes and the introduction of human immunoglobulin sequences, this rabbit strain has been engineered to generate a highly diverse human IgG antibody repertoire. We further incorporated human CD79a/b and Bcl2 (B-cell lymphoma 2) genes, which enhance B-cell receptor expression and B-cell survival. Following immunization against the angiogenic factor BMP9 (Bone Morphogenetic Proteins 9), we were able to isolate a set of exquisitely affine and specific neutralizing antibodies from these rabbits. Sequence analysis of these binders revealed that both somatic hypermutation and gene conversion are fully operational in this strain, without compromising the very high degree of humanness. This powerful new transgenic strategy will allow further expansion of the use of endogenous immune mechanisms in drug development.


Assuntos
Animais Geneticamente Modificados , Afinidade de Anticorpos/imunologia , Especificidade de Anticorpos/imunologia , Linfócitos B/imunologia , Imunoglobulina G/imunologia , Animais , Humanos , Imunoglobulina G/genética , Coelhos
7.
MAbs ; 11(4): 639-652, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30698484

RESUMO

T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.


Assuntos
Anticorpos Biespecíficos/metabolismo , Anticorpos Monoclonais/metabolismo , Complexo CD3/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Animais , Animais Endogâmicos , Antígenos de Neoplasias/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Feminino , Humanos , Células Jurkat , Ativação Linfocitária , Camundongos , Neoplasias/imunologia , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L710-L721, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28235950

RESUMO

Over past years, a critical role for the immune system and, in particular, for mast cells in the pathogenesis of pulmonary hypertension (PH) has emerged. However, the way in which mast cells promote PH is still poorly understood. Here, we investigated the mechanisms by which mast cells may contribute to PH, specifically focusing on the interaction between the innate and adaptive immune response and the role of B cells and autoimmunity. Experiments were performed in Sprague-Dawley rats and B cell-deficient JH-KO rats in the monocrotaline, Sugen/hypoxia, and the aortic banding model of PH. Hemodynamics, cell infiltration, IL-6 expression, and vascular remodeling were analyzed. Gene array analyses revealed constituents of immunoglobulins as most prominently regulated mast cell-dependent genes in the lung in experimental PH. IL-6 was shown to link mast cells to B cells, as 1) IL-6 was upregulated and colocalized with mast cells and was reduced by mast-cell stabilizers and 2) IL-6 or mast cell blockade reduced B cells in lungs of monocrotaline-treated rats. A functional role for B cells in PH was demonstrated in that either blocking B cells by an anti-CD20 antibody or B-cell deficiency in JH-KO rats attenuated right ventricular systolic pressure and vascular remodeling in experimental PH. We here identify a mast cell-B cell axis driven by IL-6 as a critical immune pathway in the pathophysiology of PH. Our results provide novel insights into the role of the immune system in PH, which may be therapeutically exploited by targeted immunotherapy.


Assuntos
Linfócitos B/metabolismo , Hipertensão Pulmonar/fisiopatologia , Pulmão/irrigação sanguínea , Pulmão/fisiopatologia , Mastócitos/metabolismo , Remodelação Vascular , Animais , Autoanticorpos/metabolismo , Pressão Sanguínea , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Interleucina-6/metabolismo , Masculino , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Ratos Sprague-Dawley , Sístole , Fatores de Tempo
9.
Transplantation ; 101(8): 1770-1776, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28230642

RESUMO

Techniques to obtain large quantities of antigen-specific monoclonal antibodies (mAbs) were first established in the 1970s when Georges Köhler and César Milstein immortalized antibody-producing mouse B-lymphocytes by fusion with myeloma cells (http://www.whatisbiotechnology.org/exhibitions/milstein). Combined with the expression of human antibodies in transgenic animals, this technique allowed upon immunization the generation of highly specific fully human mAbs for therapeutic applications. Apart from being extremely beneficial, mAbs are a huge success commercially. However, despite cell fusion generating many useful mAbs questions have been asked about which types of cells are prone to fuse and whether other methods may identify a wider range of binders. The discovery that expression libraries, using Escherichia coli or yeast, produced different specificities was intriguing and more recently Next-Generation Sequencing has identified wide-ranging usage with highly diverse and unique repertoires. Another strategy is the combination of flow cytometry sorting of antigen-binding B lymphocytes and single-cell reverse transcription polymerase chain reaction followed by reexpression, which has identified many high-affinity mAbs.


Assuntos
Anticorpos Monoclonais/biossíntese , Linfócitos B/imunologia , Fusão Celular/métodos , Imunização/métodos , Animais , Animais Geneticamente Modificados , Citometria de Fluxo , Humanos
10.
Int Immunol ; 23(10): 625-36, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21930595

RESUMO

The rat is an important biomedical experimental model that benefited from the recent development of new transgenic and knockout techniques. With the goal to optimize rat mAb production and to analyze the impact of Bcl-2 on B-cell development, we generated bcl-2 transgenic rats. Transgenic rats showed Bcl-2 over-expression in B cells, increased B cell numbers in lymphoid organs, elevated production of immunoglobulins (Igs) and prolonged B-cell survival in vitro. Transgenic rats remained healthy, reproduced normally and did not develop autoimmunity. Fusions with bcl-2 transgenic splenocytes did not result in increased hybridoma generation. A comparison of on- and off-rates of 39 mAbs generated with bcl-2 transgenic and wild-type animals revealed no significant differences. Over-expression of Bcl-2 in hybridomas did not change cell proliferation but resulted in increased Ig production. Bcl-2 transgenic rats will be a useful tool for the generation of rat mAbs, the analysis of B cells in different pathophysiological models, such as autoimmunity, cancer or organ transplantation, and the study of rat B-cell biology.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Hibridomas/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Linfócitos B/citologia , Imunoglobulinas/biossíntese , Imunoglobulinas/imunologia , Ratos , Ratos Sprague-Dawley , Ratos Transgênicos
11.
Eur J Immunol ; 40(10): 2932-41, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21038471

RESUMO

The rat is a species frequently used in immunological studies but, until now, there were no models with introduced gene-specific mutations. In a recent study, we described for the first time the generation of novel rat lines with targeted mutations using zinc-finger nucleases. In this study, we compare immune development in two Ig heavy-chain KO lines; one with truncated Cµ and a new line with removed JH segments. Rats homozygous for IgM mutation generate truncated Cµ mRNA with a de novo stop codon and no Cγ mRNA. JH-deletion rats showed undetectable mRNA for all H-chain transcripts. No serum IgM, IgG, IgA and IgE were detected in these rat lines. In both lines, lymphoid B-cell numbers were reduced >95% versus WT animals. In rats homozygous for IgM mutation, no Ab-mediated hyperacute allograft rejection was encountered. Similarities in B-cell differentiation seen in Ig KO rats and ES cell-derived Ig KO mice are discussed. These Ig and B-cell-deficient rats obtained using zinc-finger nucleases-technology should be useful as biomedical research models and a powerful platform for transgenic animals expressing a human Ab repertoire.


Assuntos
Linfócitos B/imunologia , Transplante de Coração/imunologia , Regiões Constantes de Imunoglobulina/imunologia , Cadeias Pesadas de Imunoglobulinas/imunologia , Região de Junção de Imunoglobulinas/imunologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Linfócitos B/citologia , Diferenciação Celular/imunologia , Células-Tronco Embrionárias/imunologia , Sobrevivência de Enxerto/imunologia , Regiões Constantes de Imunoglobulina/genética , Cadeias Pesadas de Imunoglobulinas/genética , Isotipos de Imunoglobulinas/sangue , Região de Junção de Imunoglobulinas/genética , Tecido Linfoide/imunologia , Dados de Sequência Molecular , RNA/química , RNA/genética , Ratos , Ratos Endogâmicos Lew , Ratos Mutantes , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos , Dedos de Zinco/genética
12.
J Immunol ; 180(12): 7919-30, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523255

RESUMO

The induction of heme oxygenase 1 (HO-1) by a single treatment with cobalt protoporphyrin (CoPPIX) protects against inflammatory liver failure and ischemia reperfusion injury after allotransplantation. In this context, the HO-1-mediated inhibition of donor-derived dendritic cell maturation and migration is discussed as one of the key events of graft protection. To investigate the poorly understood mechanism of CoPPIX-induced HO-1 activity in more detail, we performed gene expression analysis in murine liver, revealing the up-regulation of STAT3 after CoPPIX treatment. By using wild-type and HO-1-deficient dendritic cells we demonstrated that LPS-induced maturation is dependent on STAT3 phosphorylation and independent of HO-1 activity. In summary, our observations revise our understanding of the anti-inflammatory properties of HO-1 and highlight the immunomodulatory capacity of STAT3, which might be of further interest for targeting undesired immune responses, including ischemia reperfusion injury.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Heme Oxigenase-1/fisiologia , Fator de Transcrição STAT3/fisiologia , Animais , Células da Medula Óssea/enzimologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/enzimologia , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/fisiologia , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/deficiência , Heme Oxigenase-1/genética , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Protoporfirinas/administração & dosagem , Protoporfirinas/farmacologia , Fator de Transcrição STAT3/biossíntese , Fator de Transcrição STAT3/metabolismo
13.
J Invest Dermatol ; 125(3): 473-81, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16117788

RESUMO

RDP58 is the first lead compound in a series of immunomodulating decapeptides discovered through activity-based screening and computer-aided, rational design. RDP58 disrupts cellular responses signaled through the Toll-like and tumor necrosis factor (TNF) receptor families and occludes important signal transduction pathways involved in inflammation, inhibiting the production of tumor necrosis factor alpha (TNFalpha), interferon-gamma, interleukin (IL)-2, IL-6, and IL-12. These pro-inflammatory cytokines are thought to be involved in the pathogenesis of several inflammatory and autoimmune diseases, including atopic dermatitis and psoriasis. The goal of this study was to determine the ability of RDP58 to inhibit skin inflammation following exposure to the well-characterized protein kinase C activator and tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA). Topical application of RDP58 to the epidermis following TPA treatment resulted in the amelioration of the phorbol ester-induced irritant contact dermatitis. Substantial reductions were observed in skin thickness and tissue weight, neutrophil-mediated myeloperoxidase activity, inflammatory cytokine production, and various histopathological indicators. We also found RDP58 to be effective in reducing the compounding inflammatory damage brought on by chronic TPA exposure, and that it is capable of targeting inflammatory mediators specifically in the keratinocyte. These results demonstrate that topically applied RDP58 is an effective anti-inflammatory treatment in the phorbol ester-induced dermatitis model, and suggest that it may have therapeutic potential in a variety of immune-related cutaneous diseases.


Assuntos
Dermatite Irritante/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Peptídeos/uso terapêutico , Administração Tópica , Animais , Citocinas/metabolismo , Dermatite Irritante/patologia , Fatores Imunológicos/administração & dosagem , Mediadores da Inflamação/antagonistas & inibidores , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Camundongos , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Peptídeos/administração & dosagem , Peroxidase/metabolismo , Acetato de Tetradecanoilforbol/toxicidade
14.
J Neuroimmunol ; 152(1-2): 33-43, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15223235

RESUMO

The therapeutic value of a novel immunomodulatory peptide, RDP58, was investigated in the acute experimental autoimmune encephalomyelitis (EAE) model of Multiple Sclerosis (MS). RDP58 is a 10-amino acid peptide with two major activities: (i) inhibition of inflammatory TH1 cytokines such as TNFalpha, IFNgamma, and IL12 and (ii) up-regulation of heme oxygenase-1 (HO-1) expression. Experiments in which EAE-induced Lewis rats exhibit an acute monophasic episode of disease demonstrated that a single intracerebroventricular injection of RDP58 is effective in preventing clinical signs of disease. The therapeutic effect on disease activity was observed at all pre-onset administration times and at all doses tested. Consistent with disease activity in vivo, RDP58-treated animals had reduced cellular infiltration within the spinal cord along with decreased TNFalpha expression levels. The data in this proof of concept study support the premise that RDP58, as a platform molecule, may be a promising new therapeutic intervention in autoimmune and inflammatory diseases.


Assuntos
Encefalomielite Autoimune Experimental/prevenção & controle , Peptídeos/administração & dosagem , Medula Espinal/efeitos dos fármacos , Animais , Western Blotting , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Injeções Intraventriculares , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/imunologia , Medula Espinal/patologia , Fator de Necrose Tumoral alfa/biossíntese
15.
Clin Cancer Res ; 10(8): 2851-9, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15102694

RESUMO

Cancer patients undergoing triple therapy (CPT-11, 5-fluorouracil, and leucovorin) often present with severe delayed diarrhea as a result of chemotherapy-induced gastrointestinal (GI) toxicity and inflammation. RDP58 is a novel, anti-inflammatory, D-amino acid decapeptide that inhibits the production of tumor necrosis factor alpha, IFN-gamma, and interleukin 12, and has been shown to effectively inhibit clinical symptoms and intestinal inflammation in several rodent models of chemically induced colitis, nonhuman primates with spontaneous colitis, and humans with mild to moderate ulcerative colitis. We evaluated RDP58 as a potential protective agent in chemotherapy-induced GI inflammation. Oral administration of RDP58 significantly decreased the incidence of diarrhea and improved the survival rates of mice treated with toxic doses of CPT-11 or 5-fluorouracil. Histological analysis showed that RDP58 significantly reduced the destruction of the intestinal mucosa by inhibiting local overproduction of tumor necrosis factor alpha, IFN-gamma, and interleukin 12 in vivo. Furthermore, RDP58 administration allowed the maximum tolerated dose of CPT-11 to be doubled in tumor-bearing mice resulting in significantly enhanced primary tumor responses and prolongation of time to relapse without a concomitant increase in GI toxicity. Our results suggest that RDP58 may have clinical utility in cancer therapy by preventing treatment-associated GI toxicity and potentially increasing the effectiveness of chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Camptotecina/análogos & derivados , Camptotecina/administração & dosagem , Antígenos de Histocompatibilidade Classe I , Peptídeos/administração & dosagem , Administração Oral , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Camptotecina/toxicidade , Linhagem Celular Tumoral , Citocinas/biossíntese , Citocinas/metabolismo , Sistema Digestório/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Fluoruracila/administração & dosagem , Mucosa Intestinal/metabolismo , Irinotecano , Jejuno/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Peptídeos/química , Peptídeos/toxicidade , Fatores de Tempo
16.
Transpl Int ; 16(12): 849-56, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12942166

RESUMO

There remains no treatment for chronic allograft rejection mainly manifested by progressive arteriosclerosis. We investigated the effect of Allotrap peptide RDP58 therapy on arteriosclerosis in an aortic allotransplant model. RDP58 was administered intraperitoneally at 0.1, 0.5, or 2.5 mg/kg, every other day after transplantation. RDP58 therapy markedly inhibited vascular intimal thickening, media necrosis, and adventitial cellular inflammation. The attenuation of arteriosclerosis was associated with the induction of heme oxygenase (HO)-1 expression, inhibition of TNF-alpha production in aortic allografts, as well as decreased specific complement-dependent cytotoxic antibodies in serum. RDP58 inhibited both smooth muscle cell (SMC) proliferation with an 80% inhibition at 100 microM without evidence of cytotoxicity and TNF-induced apoptosis of SMCs in a dose-dependent fashion. These data suggest that the suppressive effect of RDP58 on allograft arteriosclerosis is due to multiple actions of the peptide, including induction of HO-1, inhibition of TNF-alpha, and a direct effect on SMC proliferation.


Assuntos
Aorta Torácica/transplante , Arteriosclerose/tratamento farmacológico , Rejeição de Enxerto/tratamento farmacológico , Peptídeos/farmacologia , Animais , Aorta Torácica/imunologia , Aorta Torácica/patologia , Apoptose , Arteriosclerose/imunologia , Autoanticorpos/imunologia , Divisão Celular , Linhagem Celular , Doença Crônica , Proteínas do Sistema Complemento/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Ratos , Transplante Homólogo , Fator de Necrose Tumoral alfa/imunologia
17.
J Am Soc Nephrol ; 14(3): 745-54, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12595512

RESUMO

Heme oxygenase-1 (HO-1) overexpression using gene transfer protects rat livers against ischemia/reperfusion (I/R) injury. This study evaluates the effects of Ad-HO-1 gene transfer in a rat renal isograft model. Donor LEW kidneys were perfused with Ad-HO-1, Ad-beta-gal, or PBS, stored at 4 degrees C for 24 h, and transplanted orthotopically into LEW recipients, followed by contralateral native nephrectomy. Serum creatinine, urine protein/creatinine ratios, severity of histologic changes, HO-1 mRNA/protein expression, and HO enzymatic activity were analyzed. Ad-HO-1 gene transfer conferred a survival advantage when compared with PBS- and Ad-beta-gal-treated controls, with median survival of 100, 7, and 7 d, respectively (P < 0.01). Serum creatinine levels were elevated at day 7 in all groups (range, 2.2 to 5.8 mg/dl) but recovered to 1.0 mg/dl by day 14 (P < 0.01) in Ad-HO-1 group, which was sustained thereafter. Urine protein/creatinine ratio at day 7 was elevated in both PBS and Ad-beta-gal, as compared with the Ad-HO-1 group (12.0 and 9.8 versus 5.0; P < 0.005); histologically, ATN and glomerulosclerosis was more severe in Ad-beta-gal group at all time points. Reverse transcriptase-PCR-based HO-1 gene expression was significantly increased before reperfusion (P < 0.001) and remained increased in the Ad-HO-1-treated group for 3 d after transplantation. Concomitantly, HO enzymatic activity was increased at transplantation and at 3 d posttransplant in the Ad-HO-1 group, compared with Ad-beta-gal controls (P < 0.05); tubular HO-1 expression was discernible early posttransplant in the Ad-HO-1 group alone. These findings are consistent with protective effects of HO-1 overexpression using a gene transfer approach against severe renal I/R injury, with reduced mortality and attenuation of tissue injury.


Assuntos
Sobrevivência de Enxerto/fisiologia , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase (Desciclizante)/metabolismo , Transplante de Rim , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/terapia , Adenoviridae/genética , Animais , Temperatura Baixa , Regulação Enzimológica da Expressão Gênica , Técnicas de Transferência de Genes , Heme Oxigenase-1 , Imuno-Histoquímica , Rim/enzimologia , Rim/patologia , Necrose Tubular Aguda/metabolismo , Necrose Tubular Aguda/patologia , Necrose Tubular Aguda/terapia , Masculino , Ratos , Ratos Endogâmicos Lew , Traumatismo por Reperfusão/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Índice de Gravidade de Doença
18.
Curr Pharm Des ; 8(24): 2217-29, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12369864

RESUMO

We used a novel rational design approach in the development of novel immunomodulatory peptides, in particular RDP58, with at least two primary biological activities, inhibition of TNF production and upregulation of heme oxygenase-1 (HO-1) activity. The design strategy used a variety of topological and shape descriptors in combination with an analysis of molecular dynamics trajectories to identify potential drug candidates. The process was initiated using a panel of 19 lead molecules derived from a functionally-important region of HLA I, on the premise that the peptides might modulate immune responses by blocking HLA/T lymphocyte interactions. Each of these 19 peptides was tested in vivo for potential cardiac allograft protection in a mouse model. Outcome of graft survival was the primary data available to describe nine of the peptides as active and ten as inactive. A virtual combinatorial library of 279,936 peptides was then generated, based on physical properties associated with efficacy in the original learning set of 19 peptides--in particular, the distribution of lipophilic residues. The library was screened using descriptors that fell into 2 categories: static (physico-chemical) and dynamic (conformational). The screenings identified 5 peptides with theoretic potential efficacy. These were synthesized and tested for activity in vitro and in vivo. Besides prolonging graft survival in rodents, RDP58 was found to inhibit TNF and increase HO-1 activity; these biological activities were tested exhaustively in various in vivo disease models. RDP58 demonstrated convincing potential to alleviate disease symptoms in many of the experimental diseases.


Assuntos
Imunossupressores/farmacologia , Peptídeos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Doenças Autoimunes/tratamento farmacológico , Citocinas/antagonistas & inibidores , Desenho de Fármacos , Humanos , Imunossupressores/síntese química , Imunossupressores/química , Imunossupressores/farmacocinética , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacocinética
19.
Transplantation ; 74(5): 591-8, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12352873

RESUMO

Today, the major problem in organ transplantation is not acute graft rejection but chronic graft deterioration. In addition to alloantigen-specific events, alloantigen independent factors like donor age, previous diseases, consequences of brain death, and perioperative events of ischemia/reperfusion injury have a major impact on long-term graft function. The induction of the stress protein heme oxygenase-1 (HO-1) protects cells from injury and apoptosis. Here, we tested the protective effects of HO-1 induction in a clinically relevant kidney transplant model. Induction of HO-1 expression following cobalt-protoporphyrin (CoPP) treatment in organ donors prolonged graft survival and long-term function remarkably following extended periods of ischemia. Positive effects were observed with both optimal and marginal grafts from old donor animals. Structural changes characteristic for chronic rejection, as well as graft infiltration by monocytes/macrophages and CD8+ T cells, were substantially reduced following HO-1 induction. Up-regulation of HO-1 expression before organ transplantation was also associated with reduced levels for tumor necrosis factor (TNF)-alpha mRNA, increased levels for interferon (IFN)-gamma, and bcl-x, and insignificant differences for CD25, interleukin (IL)-2, IL-4, IL-6, and IL-10 mRNA levels. The significant improvement of long-term graft function following induction of HO-1 expression in donor organs suggests that this strategy may be a novel clinical treatment option with particular relevance for transplantation of marginal organs.


Assuntos
Cobalto/farmacologia , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/genética , Transplante de Rim/patologia , Protoporfirinas/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Indução Enzimática/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1 , Interleucinas/genética , Transplante de Rim/imunologia , Transplante de Rim/fisiologia , Masculino , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Receptores de Interleucina-2/genética , Linfócitos T/patologia , Transcrição Gênica , Falha de Tratamento
20.
Am J Transplant ; 2(7): 581-92, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12201358

RESUMO

The hallmark of chronic rejection is the occlusion of the artery lumen by intima hyperplasia as a consequence of leukocyte infiltration and vascular smooth muscle cell (VSMC) migration and proliferation. Heme oxygenase-1 (HO-1) is a tissue protective molecule which degrades heme into carbon monoxide (CO), free iron and biliverdin. We analyzed the effects of HO-1 gene transfer into the vessel wall using an adenoviral vector (AdHO-1) and of CO delivery in a model of chronic allogeneic aorta rejection in rats. Carbon monoxide treatment was achieved by a new pharmacological approach in transplantation using methylene chloride (MC), which releases CO after degradation. AdHO-1-mediated gene transfer into aorta endothelial cells (ECs) or CO delivery resulted in a significant reduction in intimal thickness compared to untreated or noncoding adenovirus-treated controls. Aortas transduced with AdHO-1 or treated with CO showed a reduction in the number of leukocytes as well as in the expression of adhesion molecules, costimulatory molecules and cytokines, with the gene transfer treatment displaying a more pronounced effect than the CO treatment. Conversely, CO inhibited VSMC accumulation in the intima more efficiently than AdHO-1 treatment. Gene transfer of HO-1 and pharmacological manipulation of CO are novel approaches to the analysis and treatment of chronic rejection.


Assuntos
Monóxido de Carbono/uso terapêutico , Rejeição de Enxerto/prevenção & controle , Heme Oxigenase (Desciclizante)/genética , Animais , Células COS/transplante , Carboxihemoglobina/farmacocinética , Chlorocebus aethiops , Doença Crônica , Técnicas de Transferência de Genes , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Masculino , Ratos , Ratos Endogâmicos Lew , Transplante Heterólogo , Transplante Homólogo , Transplante Isogênico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA