Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Am J Pathol ; 192(4): 687-700, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35063406

RESUMO

Skin toxicity is a common safety concern associated with drugs that inhibit epidermal growth factor receptors as well as other targets involved in epidermal growth and differentiation. Recently, the use of a three-dimensional reconstructed human epidermis model enabled large-scale drug screening and showed potential for predicting skin toxicity. Although a decrease in epidermal thickness was often observed when the three-dimensional reconstructed tissues were exposed to drugs causing skin toxicity, the thickness evaluation of epidermal layers from a pathologist was subjective and not easily reproducible or scalable. In addition, the subtle differences in thickness among tissues, as well as the large number of samples tested, made cross-study comparison difficult when a manual evaluation strategy was used. The current study used deep learning and image-processing algorithms to measure the viable epidermal thickness from multiple studies and found that the measured thickness was not only significantly correlated with a pathologist's semi-quantitative evaluation but was also in close agreement with the quantitative measurement performed by pathologists. Moreover, a sensitivity of 0.8 and a specificity of 0.75 were achieved when predicting the toxicity of 18 compounds with clinical observations with these epidermal thickness algorithms. This approach is fully automated, reproducible, and highly scalable. It not only shows reasonable accuracy in predicting skin toxicity but also enables cross-study comparison and high-throughput compound screening.


Assuntos
Aprendizado Profundo , Dermatopatias , Algoritmos , Epiderme , Humanos , Processamento de Imagem Assistida por Computador , Pele
2.
Toxicol Appl Pharmacol ; 300: 47-54, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27078884

RESUMO

Bromo and extra terminal (BET) proteins (BRD2, BRD3, BRD4 and BRDT) are epigenetic transcriptional regulators required for efficient expression of growth promoting, cell cycle progression and antiapoptotic genes. Through their bromodomain, these proteins bind to acetylated lysine residues of histones and are recruited to transcriptionally active chromatin. Inhibition of the BET-histone interaction provides a tractable therapeutic strategy to treat diseases that may have epigenetic dysregulation. JQ1 is a small molecule that blocks BET interaction with histones. It has been shown to decrease proliferation of patient-derived multiple myeloma in vitro and to decrease tumor burden in vivo in xenograft mouse models. While targeting BET appears to be a viable and efficacious approach, the nonclinical safety profile of BET inhibition remains to be well-defined. We report that mice dosed with JQ1 at efficacious exposures demonstrate dose-dependent decreases in their lymphoid and immune cell compartments. At higher doses, JQ1 was not tolerated and due to induction of significant body weight loss led to early euthanasia. Flow cytometry analysis of lymphoid tissues showed a decrease in both B- and T-lymphocytes with a concomitant decrease in peripheral white blood cells that was confirmed by hematology. Further investigation with the inactive enantiomer of JQ1 showed that these in vivo effects were on-target mediated and not elicited through secondary pharmacology due to chemical structure.


Assuntos
Azepinas/farmacologia , Sistema Imunitário/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Triazóis/farmacologia , Animais , Azepinas/administração & dosagem , Relação Dose-Resposta a Droga , Epigenômica , Sistema Imunitário/patologia , Linfócitos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Reticulócitos/efeitos dos fármacos , Triazóis/administração & dosagem
3.
Toxicol Pathol ; 44(4): 536-44, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26992887

RESUMO

The skin has a relatively limited range of responses to injury regardless of the specific mechanism underlying the insult. When the skin's barrier function is disrupted, it mounts an inflammatory and proliferative response in an effort to restore this essential function. The epidermal keratinocyte is central to the initiation of the skin's response, triggering an immunologic cascade that leads to the stereotypic morphologic responses that we encounter as pathologists. Drug-induced immune-mediated cutaneous injuries or "drug eruptions" are relatively common, sometimes with overlapping mechanisms, and it is often possible to classify these based on the classical hypersensitivity-type reactions. A specific type of immune-mediated skin injury is psoriasis. The pathogenesis of psoriasis is multifactorial but involves the interaction of environmental factors with a genetic predisposition. The initial stimulus triggering the development of psoriatic lesions involves activation of epidermal keratinocytes, with subsequent amplification driven by cross talk between the adaptive and innate immune systems. Several cytokines produced by Th17 T helper cells have recently been shown to be important in the pathogenesis of psoriasis, namely, interleukin-23 (IL-23) and IL-17, due to demonstrated clinical efficacy of cytokine blockade; and IL-22, based on its effects in both in vitro and in vivo models.


Assuntos
Dermatopatias/imunologia , Dermatopatias/fisiopatologia , Humanos
4.
Toxicol Pathol ; 44(4): 555-63, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26940714

RESUMO

In vitro skin model systems are increasingly being used both in the early evaluation of therapeutic drug candidates and in confirmatory mechanistic studies. The most commonly used of these in vitro model systems are reconstituted human epidermis (RHE) models. These RHE models consist solely of epidermal keratinocytes, which comes with some limitations but also with the advantage of focusing toxicologic and pharmacologic evaluation on keratinocytes alone. RHE models can generally be implemented more quickly, easily, and reproducibly than in vivo models and can thus be used for high throughput compound screening while potentially reducing the need for animal studies. Histologic evaluation of RHE sections can be done quite easily, and the sections are very amenable to quantification via image analysis, including automated analysis. RHE model systems can provide very valuable early indications of therapeutic candidate biology, pharmacology, and toxicity; and early results have demonstrated that RHE models have been quite predictive for in vivo pharmacologic and toxicologic effects on the skin, including clinical skin toxicity.


Assuntos
Epiderme , Queratinócitos , Modelos Biológicos , Humanos , Técnicas In Vitro , Técnicas de Cultura de Órgãos , Pele
5.
Nature ; 445(7128): 648-51, 2007 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-17187052

RESUMO

Psoriasis is a chronic inflammatory skin disease characterized by hyperplasia of the epidermis (acanthosis), infiltration of leukocytes into both the dermis and epidermis, and dilation and growth of blood vessels. The underlying cause of the epidermal acanthosis in psoriasis is still largely unknown. Recently, interleukin (IL)-23, a cytokine involved in the development of IL-17-producing T helper cells (T(H)17 cells), was found to have a potential function in the pathogenesis of psoriasis. Here we show that IL-22 is preferentially produced by T(H)17 cells and mediates the acanthosis induced by IL-23. We found that IL-23 or IL-6 can directly induce the production of IL-22 from both murine and human naive T cells. However, the production of IL-22 and IL-17 from T(H)17 cells is differentially regulated. Transforming growth factor-beta, although crucial for IL-17 production, actually inhibits IL-22 production. Furthermore, IL-22 mediates IL-23-induced acanthosis and dermal inflammation through the activation of Stat3 (signal transduction and activators of transcription 3) in vivo. Our results suggest that T(H)17 cells, through the production of both IL-22 and IL-17, might have essential functions in host defence and in the pathogenesis of autoimmune diseases such as psoriasis. IL-22, as an effector cytokine produced by T cells, mediates the crosstalk between the immune system and epithelial cells.


Assuntos
Interleucina-23/metabolismo , Interleucinas/metabolismo , Psoríase/complicações , Psoríase/patologia , Linfócitos T Auxiliares-Indutores/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Orelha/patologia , Inflamação/imunologia , Inflamação/patologia , Interleucina-17/biossíntese , Interleucina-17/metabolismo , Interleucina-23/imunologia , Interleucina-23/farmacologia , Interleucina-6/deficiência , Interleucina-6/genética , Interleucina-6/farmacologia , Interleucinas/biossíntese , Interleucinas/deficiência , Interleucinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Psoríase/imunologia , Fator de Transcrição STAT3/metabolismo , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Interleucina 22
6.
Toxicol Pathol ; 40(2): 272-87, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22222884

RESUMO

A number of therapeutic immunomodulatory biologics, including antibodies, fusion proteins, and recombinant proteins, have been causally linked with serious adverse effects in humans. In nearly all cases, these serious adverse effects have been directly associated with the immunomodulatory biologic's intended pharmacologic activity or exaggerated pharmacology. Examples of immunomodulatory biologics known to cause serious adverse effects in the clinic ranging from immunostimulation and cytokine release syndrome (e.g., TGN1412) to immunosuppression with increased risk of opportunistic infections (e.g., TNF-α antagonists, anti-integrins) are presented. Specific examples of the nonclinical testing strategy used for the clinical risk assessment of these immunomodulatory biologics are discussed, with an emphasis on the clinical relevance and predictivity of the models. Infectious challenge animal models, in particular, were critically evaluated for their utility in evaluating clinical risk assessment versus understanding mechanism of action. The nonclinical safety testing strategy for an immunomodulatory biologic should be custom tailored to interrogate the biology of the immunologic target in order to best assess potential clinical risk. This nonclinical strategy should include mechanistic and efficacy models of pharmacologic activity and immunologic signaling pathways, in vitro immunologic assays such as cytokine release, and immunophenotypic assessment by flow cytometry, immunohistochemistry, and/or immunofluorescence, as appropriate.


Assuntos
Produtos Biológicos/efeitos adversos , Avaliação de Medicamentos/métodos , Fatores Imunológicos/efeitos adversos , Animais , Humanos , Medição de Risco , Yin-Yang
7.
J Immunol ; 184(8): 4307-16, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20231694

RESUMO

It has been suggested that IL-17RC forms a complex with IL-17RA to mediate the functions of IL-17A and IL-17F homodimers as well as IL-17A/F heterodimers. It is still unclear whether IL-17RC is absolutely required for the signaling of IL-17 cytokines in vivo. By using Il-17rc-deficient mice, we show that IL-17RC is essential for the signaling of IL-17A, IL-17F, and IL-17A/F both in vitro and in vivo. IL-17RC does not preassociate with IL-17RA on the cell surface; rather IL-17A can induce the formation of an IL-17RC and IL-17RA complex. This process is not dependent on the intracellular similar expression to fibroblast growth factor genes and IL-17Rs (SEFIR) domain of IL-17RC, but the SEFIR is essential in IL-17A signal transduction. Finally, Il-17rc(-/-) mice develop much milder disease in an experimental autoimmune encephalomyelitis model, supporting an essential role for IL-17RC in mediating immune-mediated CNS inflammation.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Interleucina-17/fisiologia , Receptores de Interleucina/fisiologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Técnicas de Cocultura , Encefalomielite Autoimune Experimental/patologia , Humanos , Interleucinas/fisiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Receptores de Interleucina/deficiência , Receptores de Interleucina/genética
8.
J Immunol ; 185(12): 7487-97, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21076065

RESUMO

Engagement of TCRs induces actin rearrangements, which are critical for T cell activation. T cell responses require new actin polymerization, but the significance of higher-order actin structures, such as microfilament bundles, is unknown. To determine the role of the actin-bundling protein leukocyte-plastin (L-plastin; LPL) in this process, T cells from LPL(-/-) mice were studied. LPL(-/-) T cells were markedly defective in TCR-mediated cytokine production and proliferation. LPL(-/-) T cells also spread inefficiently on surfaces with immobilized TCR ligands and formed smaller immunological synapses with APCs, likely due to defective formation of lamellipodia. LPL(-/-) mice showed delayed rejection of skin allografts after release from immunosuppression. Moreover, LPL(-/-) mice developed much less severe neurologic symptoms in experimental autoimmune encephalomyelitis, which correlated with impaired T cell responses to Ag, manifested by reduced proliferation and production of IFN-γ and IL-17. Thus, LPL-dependent actin bundling facilitates the formation of lamellipodia and normal immunological synapses and thereby enables T cell activation.


Assuntos
Sinapses Imunológicas/imunologia , Ativação Linfocitária/imunologia , Fosfoproteínas/imunologia , Linfócitos T/imunologia , Actinas/genética , Actinas/imunologia , Actinas/metabolismo , Animais , Antígenos/genética , Antígenos/imunologia , Antígenos/metabolismo , Proliferação de Células , Proteínas do Citoesqueleto , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Rejeição de Enxerto/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Sinapses Imunológicas/genética , Sinapses Imunológicas/metabolismo , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas dos Microfilamentos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Pseudópodes/genética , Pseudópodes/imunologia , Pseudópodes/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transplante de Pele/imunologia , Linfócitos T/metabolismo , Transplante Homólogo
9.
Ann Neurol ; 68(3): 369-83, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20641064

RESUMO

OBJECTIVE: Clinical studies indicate that anti-CD20 B-cell depletion may be an effective multiple sclerosis (MS) therapy. We investigated mechanisms of anti-CD20-mediated immune modulation using 2 paradigms of experimental autoimmune encephalomyelitis (EAE). METHODS: Murine EAE was induced by recombinant myelin oligodendrocyte glycoprotein (rMOG), a model in which B cells are considered to contribute pathogenically, or MOG peptide (p)35-55, which does not require B cells. RESULTS: In EAE induced by rMOG, B cells became activated and, when serving as antigen-presenting cells (APCs), promoted differentiation of proinflammatory MOG-specific Th1 and Th17 cells. B-cell depletion prevented or reversed established rMOG-induced EAE, which was associated with less central nervous system (CNS) inflammation, elimination of meningeal B cells, and reduction of MOG-specific Th1 and Th17 cells. In contrast, in MOG p35-55-induced EAE, B cells did not become activated or efficiently polarize proinflammatory MOG-specific T cells, similar to naive B cells. In this setting, anti-CD20 treatment exacerbated EAE, and did not impede development of Th1 or Th17 cells. Irrespective of the EAE model used, B-cell depletion reduced the frequency of CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), and increased the proinflammatory polarizing capacity of remaining myeloid APCs. INTERPRETATION: Our study highlights distinct roles for B cells in CNS autoimmunity. Clinical benefit from anti-CD20 treatment may relate to inhibition of proinflammatory B cell APC function. In certain clinical settings, however, elimination of unactivated B cells, which participate in regulation of T cells and other APC, may be undesirable. Differences in immune responses to MOG protein and peptide may be important considerations when choosing an EAE model for testing novel B cell-targeting agents for MS.


Assuntos
Anticorpos/uso terapêutico , Antígenos CD20/imunologia , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/fisiopatologia , Ativação Linfocitária/imunologia , Animais , Antígenos CD20/genética , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Citometria de Fluxo/métodos , Fatores de Transcrição Forkhead/metabolismo , Glicoproteínas/efeitos adversos , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/efeitos adversos , Estatísticas não Paramétricas , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
10.
Blood ; 113(9): 1919-28, 2009 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-19109565

RESUMO

Hematopoietic stem cells (HSCs) have the capacity to self-renew and continuously differentiate into all blood cell lineages throughout life. At each branching point during differentiation, interactions with the environment are key in the generation of daughter cells with distinct fates. Here, we examined the role of the cell adhesion molecule JAM-C, a protein known to mediate cellular polarity during spermatogenesis, in hematopoiesis. We show that murine JAM-C is highly expressed on HSCs in the bone marrow (BM). Expression correlates with self-renewal, the highest being on long-term repopulating HSCs, and decreases with differentiation, which is maintained longest among myeloid committed progenitors. Inclusion of JAM-C as a sole marker on lineage-negative BM cells yields HSC enrichments and long-term multilineage reconstitution when transferred to lethally irradiated mice. Analysis of Jam-C-deficient mice showed that two-thirds die within 48 hours after birth. In the surviving animals, loss of Jam-C leads to an increase in myeloid progenitors and granulocytes in the BM. Stem cells and myeloid cells from fetal liver are normal in number and homing to the BM. These results provide evidence that JAM-C defines HSCs in the BM and that JAM-C plays a role in controlling myeloid progenitor generation in the BM.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/fisiologia , Diferenciação Celular/genética , Imunoglobulinas/genética , Imunoglobulinas/fisiologia , Células Progenitoras Mieloides/fisiologia , Animais , Moléculas de Adesão Celular/metabolismo , Linhagem da Célula/genética , Proliferação de Células , Feminino , Deleção de Genes , Hematopoese/genética , Imunoglobulinas/metabolismo , Imunofenotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Progenitoras Mieloides/metabolismo
12.
Clin Pharmacol Ther ; 105(1): 177-189, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29952004

RESUMO

Most treatments for epithelial injury target hematopoietic mechanisms, possibly causing immunosuppression. Interleukin (IL)-22 promotes tissue regeneration, acting directly on epithelial cells. UTTR1147A, a human IL-22Fc (immunoglobulin G (IgG)4) fusion protein, activates IL-22 signaling. This phase I placebo-controlled trial of single, ascending, i.v. (1-120 µg/kg) and s.c (3-120 µg/kg) doses of UTTR1147A analyzed its effects on safety, tolerability, pharmacokinetics, and pharmacodynamic biomarkers in healthy volunteers. Most adverse events (AEs) were mild or moderate. The maximum tolerated i.v. dose in healthy volunteers was 90 µg/kg. Predominant AEs were dose-dependent reversible skin effects consistent with IL-22 pharmacology. UTTR1147A exposure increased approximately dose-proportionally, with a half-life of ~1 week. IL-22 biomarkers (regenerating islet protein 3A (REG3A), serum amyloid A (SAA), and C-reactive protein (CRP)) increased dose-dependently. Neither inflammatory symptoms and signs nor cytokines increased with CRP elevations. UTTR1147A demonstrated acceptable safety, pharmacokinetics, and IL-22R engagement, supporting further clinical development.


Assuntos
Células Epiteliais/efeitos dos fármacos , Imunoglobulina G/administração & dosagem , Interleucinas/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Adolescente , Adulto , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Voluntários Saudáveis , Humanos , Imunoglobulina G/metabolismo , Interleucinas/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/metabolismo , Método Simples-Cego , Adulto Jovem , Interleucina 22
13.
J Pharmacol Exp Ther ; 327(2): 308-15, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18728239

RESUMO

B cell-activating factor receptor 3 (BR3)-Fc is an IgG1-receptor dimeric fusion protein that has multiple O-linked glycosylation sites and sialylation levels that can vary in the manufacturing process. Increased sialic acid levels resulted from increased site occupancy with the O-linked N-acetylgalactosamine (GalNAc-Gal), but because the ratio of sialic acid per mole of oligosaccharide remained approximately 1, this led to increased asialo terminal GalNAc. Previous studies have demonstrated an effect of terminal asialo Gal or GalNAc on the clearance of glycoproteins due to uptake and degradation by lectin receptors in the liver. However, the previous studies examined N-linked oligosaccharides, and there are less data regarding O-linked oligosaccharides. The objective of these studies was to determine the effects on the pharmacokinetics and distribution of the asialo terminal GalNAc and varying amounts of sialic acid residues on BR3-Fc. The results of the data presented here suggest that exposed Gal on the desialylated BR3-Fc led to rapid clearance due to uptake and degradation in the liver that was associated with nonparenchymal cells. It is interesting to note that the data indicated a decreased clearance and increased exposure of BR3-Fc as the sialic acid levels increased, even though increased sialic acid was associated with increased asialo GalNAc. Therefore, the exposed GalNAc did not seem to play a role in the clearance of BR3-Fc; although the Gal linked to the hydroxyl group at position 3 may have prevented an interaction. Because we did not see uptake of desialylated BR3-Fc in hepatocytes where the asialoglycoprotein receptor is localized, this nonparenchymal cell lectin may have preference for O-linked glycoproteins.


Assuntos
Receptor de Asialoglicoproteína/farmacocinética , Receptor do Fator Ativador de Células B/farmacocinética , Glicoproteínas/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Animais , Antígenos CD19/análise , Receptor do Fator Ativador de Células B/química , Receptor do Fator Ativador de Células B/farmacologia , Feminino , Fragmentos Fc das Imunoglobulinas/metabolismo , Fragmentos Fc das Imunoglobulinas/farmacologia , Radioisótopos do Iodo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/farmacologia , Distribuição Tecidual
14.
Biochem Pharmacol ; 152: 224-235, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29608910

RESUMO

Interleukin (IL)-22 plays protective roles in infections and in inflammatory diseases that have been linked to its meditation of innate immunity via multiple mechanisms. IL-22 binds specifically to its heterodimeric receptor, which is expressed on a variety of epithelial tissues. UTTR1147A is a recombinant fusion protein that links the human cytokine IL-22 with the Fc portion of human immunoglobulin (Ig) G4. Here, we report extensive in vitro and in vivo nonclinical studies that were conducted to characterize the pharmacological activity of UTTR1147A. The in vitro activity and potency of UTTR1147A were analyzed using primary human hepatocytes and human colonic epithelial cell lines. Assessment of in vivo efficacy was performed in a mouse colitis model and by measuring relevant pharmacodynamic biomarkers, including antimicrobial peptides REG3A/ß, serum amyloid protein A (SAA) and lipopolysaccharide binding protein (LBP). The pharmacokinetic and pharmacodynamic characteristics of UTTR1147A were assessed in healthy mice, rats and cynomolgus monkeys. UTTR1147A induced STAT3 activation through binding to IL-22 receptor expressed in primary human hepatocytes and human colon cell lines. In both, activation occurred in a concentration-dependent manner with similar potencies. In the mouse colitis model, murine IL-22Fc- (muIL-22Fc) treated groups at doses of 1.25 µg and above had statistically lower average histologic colitis scores compared to the control treated group. Administration of muIL-22Fc or UTTR1147A was associated with a dose-dependent induction of PD markers REG3ß and SAA in rodents as well as REG3A, SAA and LBP in cynomolgus monkeys. The combined data confirm pharmacological activity of IL-22Fc and support potential regenerative and protective mechanisms in epithelial tissues.


Assuntos
Imunoglobulina G/metabolismo , Interleucinas/metabolismo , Animais , Área Sob a Curva , Linhagem Celular , Colite/induzido quimicamente , Colite/terapia , Citocinas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Interleucina 22
15.
Pharmacol Res Perspect ; 6(6): e00434, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30464842

RESUMO

Although Interleukin-22 (IL-22) is produced by various leukocytes, it preferentially targets cells with epithelial origins. IL-22 exerts essential roles in modulating various tissue epithelial functions, such as innate host defense against extracellular pathogens, barrier integrity, regeneration, and wound healing. Therefore, IL-22 is thought to have therapeutic potential in treating diseases associated with infection, tissue injury or chronic tissue damage. A number of in vitro and in vivo nonclinical studies were conducted to characterize the pharmacological activity and safety parameters of UTTR1147A, an IL-22 recombinant fusion protein that links the human cytokine IL-22 with the Fc portion of a human immunoglobulin. To assess the pharmacological activity of UTTR1147A, STAT3 activation was evaluated in primary hepatocytes isolated from human, cynomolgus monkey, minipig, rat, and mouse after incubation with UTTR1147A. UTTR1147A activated STAT3 in all species evaluated, demonstrating that all were appropriate nonclinical species for toxicology studies. The nonclinical safety profile of UTTR1147A was evaluated in rats, minipigs, and cynomolgus monkeys to establish a safe clinical starting dose for humans in Phase I trials and to support clinical intravenous, subcutaneous and/or topical administration treatment regimen. Results demonstrate the cross-species translatability of the biological response in activating the IL-22 pathway as well as the translatability of findings from in vitro to in vivo systems. UTTR1147A was well tolerated in all species tested and induced the expected pharmacologic effects of epidermal hyperplasia and a transient increase in on-target acute phase proteins. These effects were all considered to be clinically predictable, manageable, monitorable, and reversible.


Assuntos
Hepatócitos/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Interleucinas/toxicidade , Proteínas Recombinantes de Fusão/toxicidade , Animais , Ensaios Clínicos Fase I como Assunto , Avaliação Pré-Clínica de Medicamentos , Feminino , Hepatócitos/metabolismo , Humanos , Interleucinas/administração & dosagem , Macaca fascicularis , Masculino , Camundongos , Cultura Primária de Células , Ratos , Proteínas Recombinantes de Fusão/administração & dosagem , Fator de Transcrição STAT3/metabolismo , Suínos , Porco Miniatura , Interleucina 22
16.
PLoS One ; 12(1): e0170639, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28125663

RESUMO

Diabetic foot ulcers (DFU) are one of the major complications in type II diabetes patients and can result in amputation and morbidity. Although multiple approaches are used clinically to help wound closure, many patients still lack adequate treatment. Here we show that IL-20 subfamily cytokines are upregulated during normal wound healing. While there is a redundant role for each individual cytokine in this subfamily in wound healing, mice deficient in IL-22R, the common receptor chain for IL-20, IL-22, and IL-24, display a significant delay in wound healing. Furthermore, IL-20, IL-22 and IL-24 are all able to promote wound healing in type II diabetic db/db mice. Mechanistically, when compared to other growth factors such as VEGF and PDGF that accelerate wound healing in this model, IL-22 uniquely induced genes involved in reepithelialization, tissue remodeling and innate host defense mechanisms from wounded skin. Interestingly, IL-22 treatment showed superior efficacy compared to PDGF or VEGF in an infectious diabetic wound model. Taken together, our data suggest that IL-20 subfamily cytokines, particularly IL-20, IL-22, and IL-24, might provide therapeutic benefit for patients with DFU.


Assuntos
Diabetes Mellitus Tipo 2/genética , Pé Diabético/genética , Interleucinas/genética , Receptores de Interleucina/genética , Cicatrização/genética , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/terapia , Pé Diabético/patologia , Pé Diabético/terapia , Regulação da Expressão Gênica , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Interleucinas/administração & dosagem , Ligantes , Camundongos , Camundongos Endogâmicos NOD , Cicatrização/efeitos dos fármacos , Infecção dos Ferimentos/genética , Infecção dos Ferimentos/terapia , Interleucina 22
17.
Br J Pharmacol ; 174(8): 628-640, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28009435

RESUMO

BACKGROUND AND PURPOSE: CD22 and CD79b are cell-surface receptors expressed on B-cell-derived malignancies such as non-Hodgkin's lymphoma (NHL). An anti-mitotic agent, monomethyl auristatin E, was conjugated to anti-CD22 and anti-CD79b antibodies to develop target-specific therapies for NHL. The mechanism of action (MOA) and pharmacological and pharmacokinetic (PK) profiles of these antibody-drug conjugates (ADCs) were investigated in cynomolgus monkeys. EXPERIMENTAL APPROACH: Animals were administered anti-CD22 or anti-CD79b ADCs, respective unconjugated antibodies or vehicle. Pharmacodynamic effects on total and proliferating B cells and serum PK were then assessed. Antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) of the ADCs were evaluated in vitro. KEY RESULTS: Depletion of B cells was observed after administration of either ADC or the respective unconjugated antibodies. An extended duration of depletion was observed in animals administered ADCs. Similarly, preferential depletion of proliferating B cells in blood and germinal centre B cells in spleen were only observed in animals administered ADCs. Serum PK profiles of ADCs and respective unconjugated antibodies were comparable. In vitro, anti-human CD22 and anti-human CD79b antibodies showed no or only moderate ADCC activity, respectively; neither antibody had CDC activity. CONCLUSIONS AND IMPLICATIONS: The findings support the proposed MOA: initial depletion of total B cells by antibody-mediated opsonization, followed by preferential, sustained depletion of proliferating B cells by the auristatin conjugate due to its anti-mitotic action. Delivering potent anti-mitotic agents to B cells via the specificity of monoclonal antibodies provides a means to eliminate pathogenic B cells in NHL with improved risk-benefit profiles over traditional chemotherapeutics.


Assuntos
Anticorpos/imunologia , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Antígenos CD79/imunologia , Oligopeptídeos/farmacologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Reações Antígeno-Anticorpo , Linfócitos B/imunologia , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Macaca fascicularis , Masculino , Relação Estrutura-Atividade
18.
Clin Pharmacol Ther ; 79(6): 558-69, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16765144

RESUMO

BACKGROUND: Palifermin is a recombinant human keratinocyte growth factor approved to decrease the incidence and duration of severe oral mucositis in patients with hematologic malignancies who received myelotoxic therapy requiring hematopoietic stem cell support. METHODS: This randomized, double-blind, placebo-controlled study investigated the pharmacokinetics, pharmacodynamics, and safety of palifermin in healthy volunteers after single, escalating doses (60 to 250 mug/kg). Pharmacodynamic measurements (Ki67 staining) assessing buccal mucosal epithelial proliferation were performed at baseline and at 48 or 72 hours after dosing. RESULTS: Exposure to palifermin increased approximately dose proportionally, with a 3-fold increase observed for a 4-fold increase in dose. Palifermin concentrations decreased sharply during the first 0.5 to 1.5 hours after dosing, slightly increased or plateaued between 1.5 and 6 hours, and subsequently decreased. The overall mean systemic clearance and volume of distribution at steady state were 590 mL/h/kg and 2000 mL/kg, respectively. The mean half-life ranged from 4.5 to 6 hours across the dose levels. The mean and SD ratio of Ki67 staining at 48 hours after dosing to baseline was 1.19 (0.24) for placebo, 2.02 (0.60) for 60 mug/kg, 3.04 (1.13) for 120 mug/kg, and 4.66 (0.77) for 250 mug/kg-treated groups. CONCLUSION: Palifermin exhibited linear pharmacokinetics and caused dose-dependent epithelial proliferation.


Assuntos
Células Epiteliais/efeitos dos fármacos , Fator 7 de Crescimento de Fibroblastos/farmacologia , Mucosa Bucal/citologia , Adolescente , Adulto , Idoso , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Fator 7 de Crescimento de Fibroblastos/administração & dosagem , Fator 7 de Crescimento de Fibroblastos/sangue , Fator 7 de Crescimento de Fibroblastos/farmacocinética , Humanos , Injeções Intravenosas , Masculino , Estomatite/tratamento farmacológico
19.
Toxicol Sci ; 151(2): 245-60, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26917699

RESUMO

PRO304186, a humanized monoclonal antibody targeting soluble interleukin-17 A and F, was developed for autoimmune and inflammatory disease indications. When administered to cynomolgus monkeys PRO304186 induced unexpected adverse effects characterized by clinical signs of hematemesis, hematochezia, and moribundity. Pathology findings included hemorrhage throughout the gastrointestinal tract without any evidence of vascular wall damage or inflammatory cellular infiltration. Mechanistic investigation of these effects revealed mild elevations of serum MCP-1 and IL-12/23 but without a classical proinflammatory profile in PRO304186-treated animals. In vitro studies demonstrated off-target effects on vascular endothelial cells including activation of nitric oxide synthase leading to production of nitric oxide (NO) accompanied by increased mitochondrial membrane depolarization, glutathione depletion, and increased paracellular permeability. Additionally, endothelial cell-PRO304186-conditioned medium reduced myosin light chain phosphorylation in vascular smooth muscle cells. Furthermore, an ex vivo study utilizing segments from cynomolgus aorta and femoral artery confirmed PRO304186-induced endothelium-dependent smooth muscle relaxation and vasodilation mediated via NO. Finally, a single dose of PRO304186 in cynomolgus monkeys induced a rapid and pronounced increase in NO in the portal circulation that preceded a milder elevation of NO in the systemic circulation and corresponded temporally with systemic hypotension; findings consistent with NO-mediated vasodilation leading to hypotension. These changes were associated with non-inflammatory, localized hemorrhage in the gastrointestinal tract consistent with hemodynamic vascular injury associated with intense local vasodilation. Together, these data demonstrate that PRO304186-associated toxicity in monkeys was due to an off-target effect on endothelium that involved regional NO release resulting in severe systemic vasodilation, hypotension, and hemorrhage.


Assuntos
Anticorpos Monoclonais Humanizados/toxicidade , Artérias/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Hemorragia Gastrointestinal/induzido quimicamente , Hipotensão/induzido quimicamente , Óxido Nítrico/metabolismo , Vasodilatação/efeitos dos fármacos , Animais , Anticorpos Monoclonais Humanizados/metabolismo , Artérias/metabolismo , Artérias/fisiopatologia , Células Cultivadas , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Hemorragia Gastrointestinal/metabolismo , Hemorragia Gastrointestinal/fisiopatologia , Hematemese/induzido quimicamente , Hematemese/metabolismo , Hematemese/fisiopatologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Hipotensão/metabolismo , Hipotensão/fisiopatologia , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Interleucina-17/metabolismo , Macaca fascicularis , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Cadeias Leves de Miosina/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Fatores de Tempo
20.
Toxicol Sci ; 152(1): 72-84, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27103662

RESUMO

CRTh2 is expressed on immune cells that drive asthma pathophysiology. Current treatment options for severe asthma are inadequate and therapeutic antibody-mediated depletion of CRTh2-expressing cells represents a promising new therapeutic strategy. Here we report for the first time that CRTh2 is not only expressed on immune cells, but also on microvasculature in the central nervous system (CNS) and gastric mucosa in humans. Microvascular expression of CRTh2 raises a safety concern because a therapeutic antiCRTh2 antibody with enhanced depletion capacity could lead to vascular damage. To evaluate this safety risk, we characterized microvascular expression in human and in transgenic mice expressing human CRTh2 protein (hCRTh2.BAC.Tg) and found that CRTh2 is not localized to microvascular endothelium that is directly exposed to circulating therapeutic antibody, but rather, to pericytes that in the CNS are shielded from direct circulatory exposure by the blood-brain barrier. Immunohistochemical visualization of an intravenously administered antiCRTh2 antibody in transgenic mice revealed localization to microvascular pericytes in the gastric mucosa but not in the CNS, suggesting the blood-brain barrier effectively limits pericyte exposure to circulating therapeutic antibody in the CNS. Repeated dosing with a depleting antiCRTh2 antibody in hCRTh2.BAC.Tg mice revealed linear pharmacokinetics and no drug-related adverse findings in any tissues, including the CNS and gastric mucosa, despite complete depletion of CRTh2 expressing circulating eosinophils and basophils. Collectively, these studies demonstrate that the likelihood of drug-related CNS or gastrointestinal toxicity in humans treated with a therapeutic depleting antiCRTh2 antibody is low despite pericyte expression of CRTh2 in these tissues.


Assuntos
Antiasmáticos/farmacologia , Anticorpos Monoclonais/farmacologia , Asma/tratamento farmacológico , Sistema Nervoso Central/efeitos dos fármacos , Mucosa Gástrica/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores , Animais , Antiasmáticos/administração & dosagem , Antiasmáticos/farmacocinética , Antiasmáticos/toxicidade , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Asma/imunologia , Asma/metabolismo , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Mucosa Gástrica/imunologia , Mucosa Gástrica/metabolismo , Humanos , Injeções Intravenosas , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pericitos/imunologia , Pericitos/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/imunologia , Receptores de Prostaglandina/metabolismo , Medição de Risco , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA