Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunotoxicol ; 20(1): 2176952, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-36788724

RESUMO

Cytotoxic T-lymphocytes (CTL) are a subset of T-cells that play a critical role in protecting against intracellular infections and cancer, and have the ability to identify and kill infected or transformed cells expressing non-self peptides associated with major histocompatibility (MHC) Class I molecules. Conversely, aberrant CTL activity can contribute to immune-related pathology under conditions of overwhelming infection or autoimmunity. Disease-modifying therapeutics can have unintended effects on CTL, and a growing number of therapeutics are intended to either suppress or enhance CTL or their functions. The susceptibility of CTL to unintended effects from common therapeutic modalities underscores the need for a better understanding of the impact that such therapies have on CTL function and the associated safety implications. While there are reliable ways of quantifying CTL, notably via flow cytometric analysis of specific CTL markers, it has been a greater challenge to implement fit-for-purpose methods measuring CTL function in the context of safety studies of therapeutics. This review focuses on methods for measuring CTL responses in the context of drug safety and pharmacology testing, with the goals of informing the reader about current approaches, evaluating their pros and cons, and providing perspectives on the utility of these approaches for safety evaluation.


Assuntos
Neoplasias , Linfócitos T Citotóxicos , Animais , Primatas , Neoplasias/terapia , Citotoxicidade Imunológica
2.
Environ Health Perspect ; 130(10): 105001, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36201310

RESUMO

BACKGROUND: Key characteristics (KCs), properties of agents or exposures that confer potential hazard, have been developed for carcinogens and other toxicant classes. KCs have been used in the systematic assessment of hazards and to identify assay and data gaps that limit screening and risk assessment. Many of the mechanisms through which pharmaceuticals and occupational or environmental agents modulate immune function are well recognized. Thus KCs could be identified for immunoactive substances and applied to improve hazard assessment of immunodulatory agents. OBJECTIVES: The goal was to generate a consensus-based synthesis of scientific evidence describing the KCs of agents known to cause immunotoxicity and potential applications, such as assays to measure the KCs. METHODS: A committee of 18 experts with diverse specialties identified 10 KCs of immunotoxic agents, namely, 1) covalently binds to proteins to form novel antigens, 2) affects antigen processing and presentation, 3) alters immune cell signaling, 4) alters immune cell proliferation, 5) modifies cellular differentiation, 6) alters immune cell-cell communication, 7) alters effector function of specific cell types, 8) alters immune cell trafficking, 9) alters cell death processes, and 10) breaks down immune tolerance. The group considered how these KCs could influence immune processes and contribute to hypersensitivity, inappropriate enhancement, immunosuppression, or autoimmunity. DISCUSSION: KCs can be used to improve efforts to identify agents that cause immunotoxicity via one or more mechanisms, to develop better testing and biomarker approaches to evaluate immunotoxicity, and to enable a more comprehensive and mechanistic understanding of adverse effects of exposures on the immune system. https://doi.org/10.1289/EHP10800.


Assuntos
Substâncias Perigosas , Sistema Imunitário , Carcinógenos , Consenso , Substâncias Perigosas/toxicidade , Preparações Farmacêuticas
3.
Toxicol Sci ; 189(1): 32-50, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-35583313

RESUMO

Bispecific T-cell engager (BiTE) molecules have great potential to treat cancer. Nevertheless, dependent on the targeted tumor antigen, the mechanism of action that drives efficacy may also contribute to on-target/off-tumor toxicities. In this study, we characterize an anti-CD70 half-life extended BiTE molecule (termed N6P) which targets CD70, a TNF family protein detected in several cancers. First, the therapeutic potential of N6P was demonstrated using in vitro cytotoxicity assays and an orthotopic xenograft mouse study resulting in potent killing of CD70+ cancer cells. Next, in vitro characterization demonstrated specificity for CD70 and equipotent activity against human and cynomolgus monkey CD70+ cells. To understand the potential for on-target toxicity, a tissue expression analysis was performed and indicated CD70 is primarily restricted to lymphocytes in normal healthy tissues and cells. Therefore, no on-target toxicity was expected to be associated with N6P. However, in a repeat-dose toxicology study using cynomolgus monkeys, adverse N6P-mediated inflammation was identified in multiple tissues frequently involving the mesothelium and epithelium. Follow-up immunohistochemistry analysis revealed CD70 expression in mesothelial and epithelial cells in some tissues with N6P-mediated injury, but not in control tissues or those without injury. Collectively, the data indicate that for some target antigens such as CD70, BiTE molecules may exhibit activity in tissues with very low antigen expression or the antigen may be upregulated under stress enabling molecule activity. This work illustrates how a thorough understanding of expression and upregulation is needed to fully address putative liabilities associated with on-target/off-tumor activity of CD3 bispecific molecules.


Assuntos
Anticorpos Biespecíficos , Neoplasias , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Complexo CD3/metabolismo , Meia-Vida , Humanos , Macaca fascicularis , Camundongos , Neoplasias/metabolismo , Linfócitos T
4.
Andrology ; 10(4): 789-799, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35224888

RESUMO

BACKGROUND: Glucose-dependent insulinotropic polypeptide receptor (Gipr) gene expression has been reported in mouse spermatids and Gipr knockout male mice have previously been reported to have decreased in vitro fertilization, although the role of Gipr signaling in male mouse fertility is not well understood. OBJECTIVES: The purposes of these studies were to determine the role of glucose-dependent insulinotropic polypeptide receptor in male fertility using Gipr knockout mice and anti-glucose-dependent insulinotropic polypeptide receptor antibody-treated wild-type mice and to determine if the expression of Gipr in mouse testes is similar in non-human and human primates. METHODS AND MATERIALS: Adiponectin promoter-driven Gipr knockout male mice (GiprAdipo-/- ) were assessed for in vitro and in vivo fertility, sperm parameters, and testicular histology. CD1 male mice were administered an anti-glucose-dependent insulinotropic polypeptide receptor antibody (muGIPR-Ab) prior to and during mating for assessment of in vivo fertility and sperm parameters. Expression of Gipr/GIPR mRNA in the mouse, cynomolgus monkey, and human testes was assessed by in situ hybridization methods using species-specific probes. RESULTS: GiprAdipo-/- male mice are infertile in vitro and in vivo, despite normal testis morphology, sperm counts, and sperm motility. In contrast, administration of muGIPR-Ab to CD1 male mice did not impact fertility. While Gipr mRNA expression is detectable in the mouse testes, GIPR mRNA expression is not detectable in monkey or human testes. DISCUSSION: The infertility of GiprAdipo-/- male mice correlated with the lack of Gipr expression in the testis and/or adipocyte tissue. However, as administration of muGIPR-Ab did not impact the fertility of adult male mice, it is possible that the observations in genetically deficient male mice are related to Gipr deficiency during development. CONCLUSION: Our data support a role for Gipr expression in the mouse testis during the development of sperm fertilization potential, but based on gene expression data, a similar role for glucose-dependent insulinotropic polypeptide receptor in non-human primate or human male fertility is unlikely.


Assuntos
Polipeptídeo Inibidor Gástrico , Testículo , Animais , Feminino , Fertilidade , Polipeptídeo Inibidor Gástrico/genética , Polipeptídeo Inibidor Gástrico/metabolismo , Expressão Gênica , Humanos , Macaca fascicularis/genética , Macaca fascicularis/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores dos Hormônios Gastrointestinais , Motilidade dos Espermatozoides , Testículo/metabolismo
5.
Regul Toxicol Pharmacol ; 127: 105064, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34656748

RESUMO

Over the last decade, immunotherapy has established itself as an important novel approach in the treatment of cancer, resulting in a growing importance in oncology. Engineered T cell therapies, namely chimeric antigen receptor (CAR) T cells and T cell receptor (TCR) T cell therapies, are platform technologies that have enabled the development of products with remarkable efficacy in several hematological malignancies and are thus the focus of intense research and development activity. While engineered T cell therapies offer promise in addressing currently intractable cancers, they also present unique challenges, including their nonclinical safety assessment. A workshop organized by HESI and the US Food and Drug Administration (FDA) was held to provide an interdisciplinary forum for representatives of industry, academia and regulatory authorities to share information and debate on current practices for the nonclinical safety evaluation of engineered T cell therapies. This manuscript leverages what was discussed at this workshop to provide an overview of the current important nonclinical safety assessment considerations for the development of these therapeutic modalities (cytokine release syndrome, neurotoxicity, on-target/off-tumor toxicities, off-target effects, gene editing or vector integration-associated genomic injury). The manuscript also discusses approaches used for hazard identification or risk assessment and provides a regulatory perspective on such aspects.


Assuntos
Engenharia Celular/métodos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Linfócitos T/imunologia , Síndrome da Liberação de Citocina/fisiopatologia , Edição de Genes , Imunoterapia Adotiva/efeitos adversos , Síndromes Neurotóxicas/fisiopatologia , Receptores de Antígenos de Linfócitos T/fisiologia , Medição de Risco
6.
J Immunotoxicol ; 18(1): 154-162, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34714999

RESUMO

The immunotoxic potential of drug candidates is assessed through the examination of results from a variety of in vitro and in vivo immunophenotyping and functional study endpoints in pre-clinical studies. CD8+ cytotoxic T-lymphocyte (CTL) activity impairment by immunosuppressive agents is recognized to be a potentiating factor for decreased antiviral defense and increased cancer risk. A bi-specific T-cell engager (BiTE®)-mediated CTL activity assay that applies to ex vivo experimentation in non-human primates in the context of toxicology studies was successfully developed and applied in cynomolgus monkey regulatory studies. While an ex vivo analysis conducted in the context of repeat-dose toxicology studies focuses on the long-term impact on CTL function, an in vitro assay with the same experimental design captures acute effects in the presence of the test article. Here, the in vitro assay was applied to a list of drugs with known clinical immunomodulatory impact to understand the applicability of the assay. The results showed this assay was sensitive to a wide range of immunosuppressants directly targeting cell-intrinsic signaling pathways in activated CTL. However, agents executing immuno-modulation through inhibiting cytokines/cytokine receptors, co-stimulatory molecules, and cell adhesion and migration pathways did not impair the CTL activity in this short-term in vitro culture. In addition, anti-PD-1/PD-L1 immune checkpoint blockers enhanced the CTL activity. Taken together, the results here demonstrate that in concordance with their mechanism of action, the in vitro BiTE®-mediated CTL assay is applicable and sensitive to immunomodulatory agents acting via a variety of mechanisms.


Assuntos
Agentes de Imunomodulação , Linfócitos T Citotóxicos , Animais , Linfócitos T CD8-Positivos , Imunofenotipagem , Macaca fascicularis
7.
Biologicals ; 72: 42-53, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34303595

RESUMO

ABP 798 is a biosimilar to Rituxan® (rituximab reference product [RP]). Non-clinical assessments relevant to the primary and secondary mechanisms of action (MOA) contribute to the totality of the evidence (TOE) in supporting biosimilarity and are critical in providing scientific evidence for extrapolation of indications. Similarity of ABP 798 with rituximab RP was investigated across a range of biological activities which have potential impact on pharmacokinetics and clinical efficacy with non-clinical assessments relevant to MOA such as CD20 internalization, trogocytosis, binding to primary human natural killer (NK) cells as well as the ability to induce antibody-dependent cellular phagocytosis (ADCP) in peripheral blood mononuclear cells. Additionally, in vitro synergy of ABP 798 or RP with chemotherapeutic agents, in vivo xenograft studies in mice, and toxicological assessments in cynomolgus monkeys (including B cell depletion and toxicokinetics) were also conducted. Results from these non-clinical assessments contribute to the TOE supporting the biosimilarity between ABP 798 and rituximab RP across a range of primary and secondary MOAs and support justification for extrapolation to all indications of use for ABP 798 for which the RP is approved.


Assuntos
Antineoplásicos , Medicamentos Biossimilares , Rituximab , Animais , Antineoplásicos/farmacologia , Medicamentos Biossimilares/farmacologia , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Camundongos , Padrões de Referência , Rituximab/farmacologia
8.
Toxicol Sci ; 177(1): 94-107, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32589753

RESUMO

Feline McDonough Sarcoma-like tyrosine kinase 3 (FLT3), a tyrosine-protein kinase involved in hematopoiesis, is detectable on the cell surface of approximately 80% of leukemia isolates from adult patients with acute myeloid leukemia (AML). AMG 553 is an investigational chimeric antigen receptor (CAR) T-cell immunotherapy for the treatment of AML. FLT3 expression analysis and in vitro and in vivo studies were leveraged to evaluate the nonclinical safety of AMG 553. Cynomolgus monkeys administered autologous anti-FLT3 CAR T cells demonstrated no evidence of CAR T-cell-mediated toxicity, expansion, or persistence, likely due to restricted cell surface FLT3 protein expression in healthy animals. This highlights the limited value of such in vivo studies for safety assessment of the CAR T-cell modality when directed against a target with restricted expression. To complement these studies and directly evaluate the potential toxicities of eliciting T-cell-mediated cytotoxicity against cells with surface expression of FLT3 protein in vivo, data from cynomolgus monkey toxicology studies with 2 bispecific T-cell engager molecules targeting FLT3 were leveraged; findings were consistent with the targeted killing of bone marrow cells expressing cell surface FLT3. Potential AMG 553-induced cytotoxicity was assessed against a wide range of normal human primary cells and cell lines; cytotoxicity was observed against FLT3-positive AML cell lines and a percentage of primary bone marrow CD34+ cells. In conclusion, the nonclinical safety data suggest that AMG 553 can target FLT3 protein on AML cells, whereas only affecting a percentage of normal hematopoietic stem and progenitor cells, supporting clinical development.


Assuntos
Leucemia Mieloide Aguda , Receptores de Antígenos Quiméricos , Adulto , Animais , Gatos , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Macaca fascicularis
9.
Cancer Epidemiol Biomarkers Prev ; 29(10): 1887-1903, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32152214

RESUMO

The key characteristics (KC) of human carcinogens provide a uniform approach to evaluating mechanistic evidence in cancer hazard identification. Refinements to the approach were requested by organizations and individuals applying the KCs. We assembled an expert committee with knowledge of carcinogenesis and experience in applying the KCs in cancer hazard identification. We leveraged this expertise and examined the literature to more clearly describe each KC, identify current and emerging assays and in vivo biomarkers that can be used to measure them, and make recommendations for future assay development. We found that the KCs are clearly distinct from the Hallmarks of Cancer, that interrelationships among the KCs can be leveraged to strengthen the KC approach (and an understanding of environmental carcinogenesis), and that the KC approach is applicable to the systematic evaluation of a broad range of potential cancer hazards in vivo and in vitro We identified gaps in coverage of the KCs by current assays. Future efforts should expand the breadth, specificity, and sensitivity of validated assays and biomarkers that can measure the 10 KCs. Refinement of the KC approach will enhance and accelerate carcinogen identification, a first step in cancer prevention.See all articles in this CEBP Focus section, "Environmental Carcinogenesis: Pathways to Prevention."


Assuntos
Biomarcadores/metabolismo , Carcinógenos/metabolismo , Neoplasias/diagnóstico , Humanos , Neoplasias/patologia
10.
J Immunotoxicol ; 17(1): 67-85, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32100588

RESUMO

Currently, there is a multitude of CD3 bispecifics with different molecular designs and binding properties in preclinical and clinical development for the treatment of liquid or solid tumors. The key safety concerns with CD3 bispecifics are excessive release of cytokines, which may translate to potentially life-threating cytokine release syndrome (CRS), target organ toxicity due to redirection of T-cells to normal tissues expressing the tumor-associated antigen (TAA) (off-tumor/on-target cytotoxicity), and, in some instances, neurotoxicity. Another key challenge is to arrive at a safe clinical starting dose and an efficient escalating strategy that allows patients in early dose cohorts the potential for clinical benefit in Phase 1 trials. To expand the therapeutic index and bring more treatment options to patients, there are intense efforts to overcome these challenges through improvements in molecular design, preclinical safety assessment strategies, and clinical management practices. A recent workshop at the U.S. Food and Drug Administration (FDA) with industry, academic, and regulatory agency representation was held to discuss the challenges and explore where such improvements to the development of CD3 bispecifics can be implemented. Here, the content of the presentations and the discussion that occurred during this workshop are summarized.


Assuntos
Anticorpos Biespecíficos/toxicidade , Antígenos de Neoplasias/metabolismo , Antineoplásicos/toxicidade , Complexo CD3/antagonistas & inibidores , Síndrome da Liberação de Citocina/prevenção & controle , Animais , Anticorpos Biespecíficos/administração & dosagem , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Complexo CD3/imunologia , Complexo CD3/metabolismo , Consenso , Conferências de Consenso como Assunto , Síndrome da Liberação de Citocina/induzido quimicamente , Síndrome da Liberação de Citocina/imunologia , Citocinas/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais/normas , Europa (Continente) , Humanos , Japão , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Pesquisa Translacional Biomédica/normas , Estados Unidos , United States Food and Drug Administration
11.
Toxicol Sci ; 170(1): 10-19, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31020324

RESUMO

Natural killer (NK) cells are lymphocytes capable of cytotoxicity against virally infected cells and tumor cells. The display of effector function by NK cells is the result of interactions between germline encoded activating/inhibitory NK cell receptors and their ligands (major histocompatibility complex class I, major histocompatibility complex class I-like, viral, and cellular stress-related surface molecules) expressed on target cells. Determination of NK cell number and function is a common element of the immunotoxicology assessment paradigm for the development of certain classes of pharmaceuticals across a range of modalities. This article summarizes the evidence associating NK cell dysfunction with infectious and cancer risks, reviews emerging NK cell biology, including the impact of immunogenetics on NK cell education and function, and provides perspectives about points to consider when assessing NK cell function in different species in the context of safety assessment.


Assuntos
Resistência à Doença/imunologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/imunologia , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Receptores de Células Matadoras Naturais/metabolismo , Animais , Citotoxicidade Imunológica , Resistência à Doença/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ligantes , Preparações Farmacêuticas/administração & dosagem
12.
J Immunotoxicol ; 15(1): 119-125, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30241454

RESUMO

The immunotoxic potential of drug candidates is assessed through the examination of results from a variety of studies and endpoints. While the functional assessment of CD8+ cytotoxic T-lymphocytes (CTL) is well-characterized in the clinic, the lack of a robust macaque CTL functional assay has been an important hurdle in evaluating and accurately quantifying cell-mediated CD8+ T-cell effector responses in the nonclinical setting. This paper describes the development of an assay to measure CTL activity in peripheral blood mononuclear cells (PBMC) isolated from Cynomolgus macaques. A human EGFR/CD3 Bispecific T-cell Engager (BiTE®) was used to mount a robust CD8+ T-cell response in the presence of target-expressing cells. Upon target engagement, degranulation of CD107a and production of interferon (IFN)-γ both reliably indicated a robust functional response in CD8+ T-cells. The BiTE®-mediated stimulation method proved to be favorable when compared to other methods of stimulation in the absence of target cells. These studies demonstrated acceptable longitudinal variability of the functional assay and sensitivity to dexamethasone-mediated immunosuppression. Taken together, the results indicated an assay leveraging CD3-bispecific antibodies and target-expressing cells can provide a robust approach to the in vitro or ex vivo assessment of CTL function in Cynomolgus macaques. Because the impairment of CTL activity by immunomodulators is recognized to be an important contributor to decreased antiviral defense and increased carcinogenicity risk, we believe that this novel assay to be a valuable addition to the immunotoxicology assessment of therapeutic drug candidates.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunoensaio/métodos , Macaca fascicularis/imunologia , Animais , Anticorpos Biespecíficos/metabolismo , Complexo CD3/imunologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Receptores ErbB/imunologia , Humanos , Interferon gama/metabolismo , Leucócitos Mononucleares/citologia , Ativação Linfocitária , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Variações Dependentes do Observador
13.
Trends Pharmacol Sci ; 39(3): 232-247, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29242029

RESUMO

Cancer risk assessment of therapeutics is plagued by poor translatability of rodent models of carcinogenesis. In order to overcome this fundamental limitation, new approaches are needed that enable us to evaluate cancer risk directly in humans and human-based cellular models. Our enhanced understanding of the mechanisms of carcinogenesis and the influence of human genome sequence variation on cancer risk motivates us to re-evaluate how we assess the carcinogenic risk of therapeutics. This review will highlight new opportunities for applying this knowledge to the development of a battery of human-based in vitro models and biomarkers for assessing cancer risk of novel therapeutics.


Assuntos
Carcinógenos/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Neoplasias/prevenção & controle , Farmacovigilância , Biomarcadores Farmacológicos/análise , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Predisposição Genética para Doença , Humanos
14.
Curr Med Res Opin ; 31(3): 557-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25651481

RESUMO

OBJECTIVE: Tumor necrosis factor (TNF) is a highly pleiotropic cytokine with multiple activities other than its originally discovered role of tumor necrosis in rodents. TNF is now understood to play a contextual role in driving either tumor elimination or promotion. Using both animal and human data, this review examines the role of TNF in cancer development and the effect of TNF and TNF inhibitors (TNFis) on malignancy risk. RESEARCH DESIGN: A literature review was performed using relevant search terms for TNF and malignancy. RESULTS: Although administration of TNF can cause tumor regression in specific rodent tumor models, human expression polymorphisms suggest that TNF can be a tumor-promoting cytokine, whereas blocking the TNF pathway in a variety of tumor models inhibits tumor growth. In addition to direct effects of TNF on tumors, TNF can variously affect immunity and the tumor microenvironment. Whereas TNF can promote immune surveillance designed to eliminate tumors, it can also drive chronic inflammation, autoimmunity, angiogenesis, and other processes that promote tumor initiation, growth, and spread. Key players in TNF signaling that shape this response include NF-κB and JNK, and malignant-inflammatory cell interactions, each of which may have different responses to TNF signaling. Focusing on rheumatoid arthritis (RA) patients, where clinical experience is most extensive, a review of the clinical literature shows no increased risk of overall malignancy or solid tumors such as breast and lung cancers with exposure to TNFis. Lymphoma rates are not increased with use of TNFis. Conflicting data exist regarding the risks of melanoma and nonmelanoma skin cancer. Data regarding the risk of recurrent malignancy are limited. CONCLUSIONS: Overall, the available data indicate that elevated TNF is a risk factor for cancer, whereas its inhibition in RA patients is not generally associated with an increased cancer risk. In particular, TNF inhibition is not associated with cancers linked to immune suppression. A better understanding of the tumor microenvironment, molecular events underlying specific tumors, and epidemiologic studies of malignancies within specific disease indications should enable more focused pharmacovigilance studies and a better understanding of the potential risks of TNFis.


Assuntos
Artrite Reumatoide/imunologia , Inflamação/imunologia , Neoplasias , Fatores de Necrose Tumoral , Animais , Modelos Animais de Doenças , Humanos , Recidiva Local de Neoplasia/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Fatores de Risco , Transdução de Sinais , Microambiente Tumoral , Inibidores do Fator de Necrose Tumoral , Fatores de Necrose Tumoral/metabolismo
15.
Toxicol Sci ; 142(1): 298-310, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25159132

RESUMO

The dysregulation of phosphatidylinositol 3-kinase (PI3K)-dependent pathways is implicated in several human cancers making it an attractive target for small molecule PI3K inhibitors. A series of potent pyridyltriazine-containing inhibitors of class Ia PI3Ks were synthesized and a subset of compounds was evaluated in exploratory repeat-dose rat toxicology studies. Daily oral dosing of compound 1: in Sprague Dawley rats for four consecutive days was associated with hepatobiliary toxicity that included biliary epithelial hyperplasia and hypertrophy, periductular edema, biliary stasis, and acute peribiliary inflammatory infiltrates. These histological changes were associated with clinical pathology changes that included increased serum liver enzymes, total bile acids, and bilirubin. The predominant clearance pathway of 1: was shown in vitro and in a bile-duct cannulated rat (14)C-ADME study to be P450-mediated oxidative metabolism. An O-demethylated pyridine metabolite, M3: , was identified as a candidate proximal metabolite that caused the hepatotoxicity. Co-administration of the pan-P450 inhibitor 1-aminobenzotriazole with 1: to rats significantly reduced the formation of M3: and prevented liver toxicity, whereas direct administration of M3: reproduced the toxicity. Structural changes were introduced to 1: to make the methoxypyridine ring less susceptible to P450 oxidation (compound 2: ), and addition of a methyl group to the benzylic carbon (compound 3: ) improved the pharmacokinetic profile. These changes culminated in the successful design of a clinical candidate 3: (AMG 511) that was devoid of liver toxicity in a 14-day rat toxicity study. Herein, we describe how a metabolism-based structure-activity relationship analysis allowed for the successful identification of a PI3K inhibitor devoid of off-target toxicity.


Assuntos
Sistema Biliar/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/toxicidade , Bibliotecas de Moléculas Pequenas/toxicidade , Triazinas/toxicidade , Animais , Sistema Biliar/enzimologia , Sistema Biliar/patologia , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Feminino , Espectrometria de Massas , Taxa de Depuração Metabólica , Metilação , Estrutura Molecular , Piridinas/química , Piridinas/farmacocinética , Ratos Sprague-Dawley , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacocinética , Distribuição Tecidual , Testes de Toxicidade , Triazinas/química , Triazinas/farmacocinética
16.
J Immunotoxicol ; 11(1): 1-12, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23746314

RESUMO

Observational and clinical studies have associated increased cancer risks with primary or acquired immunodeficiencies, autoimmunity, and use of immunotherapies to treat chronic inflammation (e.g. autoimmunity) or support organ engraftment. Understanding of the relationship between immune status and cancer risk is generally grounded in two juxtaposing paradigms: that the immune system protects the host via surveillance of tumors and oncogenic viruses (e.g. immunosurveillance model) and that chronic inflammation can augment tumor growth and metastasis (inflammation model). Whereas these models support a role of immune status in many cancers, they are insufficient to explain the disproportionate increase in B-cell lymphoma risk observed across patient populations with either chronic immunosuppression or inflammation. Evaluation for the presence of Epstein-Barr virus (EBV) in lymphomas obtained from various populations demonstrates a variable role for the virus in lymphomagenesis across patient populations. An evaluation of the DNA alterations found in lymphomas and an understanding of B-cell ontogeny help to provide insight into the unique susceptibility of lymphocytes, primarily B-cells, to oncogenic transformation. EBV-independent B-cell oncogenic transformation is driven by chronic antigenic stimulation due to either inflammation (as seen in patients with autoimmune disease or a tissue allograft) or to unresolved infection (as seen in immunosuppressed patients), and the transformation arises as a result of DNA damage from genomic recombination and mutation during class switching and somatic hypermutation. This model explains the increased background rate of lymphoma in some patients with autoimmunity, and highlights the challenge of resolving the confounding that occurs between disease severity and use of targeted immunotherapies to treat chronic inflammation. The ability to distinguish between disease- and treatment-related risk of lymphoma and an appreciation of the etiology of B-cell transformation is central to an improved risk assessment by scientists, clinicians and regulators, including the approval, labeling, and chronic use of immunotherapies.


Assuntos
Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/terapia , Herpesvirus Humano 4/imunologia , Imunoterapia/métodos , Linfoma de Células B/terapia , Animais , Linfócitos B/virologia , Transformação Celular Neoplásica , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/imunologia , Humanos , Hospedeiro Imunocomprometido , Imunomodulação , Linfoma de Células B/etiologia , Linfoma de Células B/imunologia , Medição de Risco
17.
Toxicol Pathol ; 41(3): 548-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22886347

RESUMO

The immune system has been recognized for decades as a potential "target organ" of toxicity. Immune system activation can result in cytokine release resulting in severe systemic toxicity. Immunosuppression can result in impaired host defense and an increase in opportunistic infection, reemergence of latent infection, poor responses to vaccination, or increased risk of certain cancers. Several regulatory documents have addressed various aspects of immunotoxicity assessments. Nonhuman primates (NHPs) and in particular macaques are often the only relevant species for biotechnology-derived investigational new drugs based on cross-reactivity with human and NHP targets. This article reviews the challenges and opportunities associated with monitoring immune function in NHPs in the context of regulatory expectations. The article emphasizes how a comprehensive assessment of immunotoxicity remains a challenge due to interanimal variability associated with certain parameters (e.g., T-dependent antibody response)and it identifies gaps, such as the stage of development of certain assays (e.g., cytotoxic T-cell function). Despite these challenges, a thorough assessment of target biology-driven theoretical risks, in combination with proper integration of all information from the standard toxicology studies, and the refinement of certain assays should enable proper risk assessment. To this effect, emphasis should be placed on leveraging predictive in vitro assays using human cells.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Sistema Imunitário/efeitos dos fármacos , Macaca mulatta , Testes de Toxicidade/métodos , Adjuvantes Imunológicos/toxicidade , Animais , Feminino , Imunossupressores/toxicidade , Masculino , Primatas , Toxicologia
18.
J Immunotoxicol ; 6(1): 1-10, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19519157

RESUMO

The number of anti-inflammatory and immunomodulatory drugs being developed in the pharmaceutical industry has increased considerably in the past decade. This increase in research and development has been paralleled by questions from both regulatory agencies and industry on how best to assess decreased host resistance to infections or adverse immunostimulation caused by immunomodulatory agents such as anti-cytokine antibodies (e.g., the tumor necrosis factor-alpha inhibitors), anti-adhesion molecule antibodies (e.g., anti-alpha-4 integrin inhibitors) and immunostimulatory molecules (e.g., anti-CD28 antibodies). Although several methods have been developed for nonclinical assessment of immunotoxicity, highly publicized adverse events have brought to light significant gaps in the application of nonclinical immunotoxicity testing in assessing potential risk in humans. Confounding this problem is inconsistent application of immunotoxicology methods for risk assessment within the scientific community, limited understanding of appropriate immunotoxicity testing strategy for immunomodulators and inconsistent testing requests by regulatory agencies. To address these concerns, The Immunotoxicology Technical Committee (ITC) of the International Life Science Institute (ILSI) Health and Environmental Sciences Institute (HESI) organized a workshop on Immunomodulators and Clinical Immunotoxicology in May 2007. The Workshop was convened to identify key gaps in nonclinical and clinical immunotoxicity testing of anti-inflammatory and immunomodulatory agents and to begin to develop consistent approaches for immunotoxicity testing and risk assessment. This paper summarizes the outcome of the HESI ITC Immunomodulators and Clinical Immunotoxicology Workshop. Topics not discussed at the Workshop were outside the scope of this report. Although more work is needed to develop consistent approaches for immunotoxicity assessment of immunomodulators, this Workshop provided the foundation for future discussion.


Assuntos
Ensaios Clínicos como Assunto/tendências , Avaliação Pré-Clínica de Medicamentos/tendências , Fatores Imunológicos/efeitos adversos , Testes de Toxicidade/tendências , Animais , Anti-Inflamatórios/efeitos adversos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/toxicidade , Ensaios Clínicos como Assunto/normas , Consenso , Coleta de Dados , Aprovação de Drogas/legislação & jurisprudência , Avaliação Pré-Clínica de Medicamentos/normas , Indústria Farmacêutica/normas , Indústria Farmacêutica/tendências , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/imunologia , Fatores Imunológicos/farmacologia , Fatores Imunológicos/toxicidade , Modelos Animais , Medição de Risco/normas , Medição de Risco/tendências , Testes de Toxicidade/normas
19.
J Biomol Screen ; 11(6): 575-85, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16760371

RESUMO

The authors describe an assay to quantitate DNA fragmentation using terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling (TUNEL) stain, adapted to a 96-well microplate format for adherent cells, and an automated high-content screening imager. The apoptotic responses to actinomycin D (a known antineoplastic agent) to imiquimod (a small-molecule toll-like receptor [TLR] 7 agonist used in skin cancer treatment) and to several structurally related TLR 7/8 agonists were evaluated in squamous carcinoma SCC15 and SCC25 cells and normal human keratinocytes. Potent proapoptotic and growth-impairing (as determined by reduced cell numbers) actions of actinomycin D (1-300 ng/mL) were discerned with the assay. Consistent with previous reports, imiquimod (at 300 microM; approximately 75 microg/mL) induced TUNEL positivity of malignant cell cultures, but this effect also occurred in normal keratinocytes. Two related TLR agonists induced apoptosis at lower concentrations. However, the concentrations of these and the imiquimod necessary to elicit cancer cell apoptosis were 300 to 1000 times higher relative to their ability to induce the secretion of an antineoplastic protein, interferon-alpha, from human blood monocytes. This TUNEL analysis allows the quantitative comparison of compounds' apoptotic activity toward adherent malignant and normal cells and may be useful for hit characterization after a screen.


Assuntos
Aminoquinolinas/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Células Epiteliais/efeitos dos fármacos , Receptores Toll-Like/agonistas , Antineoplásicos/farmacologia , Apoptose/imunologia , Automação , Carcinoma de Células Escamosas/imunologia , Linhagem Celular , Técnicas Citológicas , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imiquimode , Queratinócitos/efeitos dos fármacos , Queratinócitos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA