Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Toxicol Sci ; 183(1): 93-104, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34240189

RESUMEN

BMS-986251 is a retinoid-related orphan receptor γt (RORγt) inverse agonist that was in development for the treatment of autoimmune diseases. RORγt is a nuclear hormone receptor and transcription factor that is involved in the differentiation and function of T helper 17 cells. RORγt-deficient (constitutive or conditional) mice develop thymic lymphomas with >50% mortality at 4 months, whereas heterozygous mice are normal. A 6-month study was conducted in rasH2-Tg hemizygous mice to assess the potential carcinogenicity of BMS-986251. BMS-986251 was administered once daily by oral gavage to groups of 27 mice/sex at doses of 0 (water control), 0 (vehicle control), 5, 25, or 75 mg/kg. The positive control, N-methyl-N-nitrosourea, was administered by a single intraperitoneal injection to 15 mice/sex at a dose of 75 mg/kg. There were no tumors attributed to BMS-986251 except for thymic lymphomas. Thymic lymphoma was observed in 1 male (3.7%) and 3 females (11.1%) at the mid dose, and 6 females (22.2%) at the high dose. No lymphomas were observed in the negative control groups whereas the incidence of lymphomas in the positive control group was 47-60%. The incidence of thymic lymphomas in the BMS-986251-treated groups was higher than published literature and test facility historical control data. Furthermore, increased thymic lymphoid cellularity (lymphoid hyperplasia) was observed at the mid dose in males and at all doses in females. Since lymphoid hyperplasia may represent a preneoplastic change, a no-effect dose for potential tumor induction was not identified in this study. These results led to the discontinuation of BMS-986251 and underscore the challenges in targeting RORγt for drug development.


Asunto(s)
Linfoma , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Animales , Pruebas de Carcinogenicidad , Femenino , Hiperplasia , Linfoma/inducido químicamente , Linfoma/genética , Masculino , Ratones , Ratones Transgénicos
2.
Toxicol Appl Pharmacol ; 409: 115285, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33069749

RESUMEN

The OX40 receptor plays a crucial co-stimulatory role in T effector cell survival, expansion, cytokine production, and cytotoxicity to tumor cells; therefore, OX40 agonists are being evaluated as anti-cancer immunotherapies, especially in combination with checkpoint inhibitors. To support clinical development of BMS-986178 (an OX40 agonist antibody), two repeat-dose toxicity studies were conducted in cynomolgus monkeys. In the first study, BMS-986178 was administered intravenously (IV) once weekly for one month at doses from 30 to 120 mg/kg. BMS-986178 was well tolerated; surprisingly, immune function was suppressed rather than increased based on pharmacodynamic (PD) and flow cytometry readouts (e.g. T-cell dependent antibody response [TDAR]). To determine whether immune suppression was due to a bi-phasic response, a follow-up study was conducted at lower doses (1 and 10 mg/kg). Although receptor engagement was confirmed, immune function was still suppressed at both doses. In addition, treatment-emergent anti-drug antibodies (ADAs) at 1 mg/kg resulted in hypersensitivity reactions and reduced BMS-986178 exposure after repeated dosing, which precluded a full PD assessment at this dose. In conclusion, BMS-986178 was clinically well-tolerated by monkeys at weekly IV doses from 10 to 120 mg/kg (AUC[0-168] ≤ 712,000 µg●h/mL). However, despite target engagement, PD assays and other immune endpoints demonstrated immune suppression, not stimulation. Due to the inverted immune response at higher doses and the onset of ADAs, additional repeat-dose toxicity studies of BMS-986178 in monkeys (that would typically be required to support Phase 3 clinical trials and registration) would not add value for human safety assessment.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunidad/inmunología , Receptores OX40/inmunología , Linfocitos T/inmunología , Animales , Femenino , Estudios de Seguimiento , Humanos , Inmunoterapia/métodos , Macaca fascicularis , Masculino
3.
Regul Toxicol Pharmacol ; 100: 35-44, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30291877

RESUMEN

Protein therapeutics represent a rapidly growing proportion of new medicines being developed by the pharmaceutical industry. As with any new drug, an Occupational Exposure Limit (OEL) should be developed to ensure worker safety. Part of the OEL determination addresses bioavailability (BA) after inhalation, which is poorly understood for protein therapeutics. To explore this, male Sprague-Dawley rats were exposed intravenously or by nose-only inhalation to one of five test proteins of varying molecular size (10-150 kDa), including a polyethylene glycol-conjugated protein. Blood, lung tissue and bronchoalveolar lavage (BAL) fluid were collected over various time-points depending on the expected test protein clearance (8 minutes-56 days), and analyzed to determine the pharmacokinetic profiles. Since the BAL half-life of the test proteins was observed to be > 4.5 h after an inhalation exposure, accumulation and direct lung effects should be considered in the hazard assessment for protein therapeutics with lung-specific targets. The key finding was the low systemic bioavailability after inhalation exposure for all test proteins (∼≤1%) which did not appear molecular weight-dependent. Given that this study examined the inhalation of typical protein therapeutics in a manner mimicking worker exposure, a default 1% BA assumption is reasonable to utilize when calculating OELs for protein therapeutics.


Asunto(s)
Polietilenglicoles/farmacocinética , Proteínas/farmacocinética , Administración por Inhalación , Animales , Disponibilidad Biológica , Líquido del Lavado Bronquioalveolar/química , L-Lactato Deshidrogenasa/metabolismo , Pulmón/metabolismo , Masculino , Concentración Máxima Admisible , Ratas Sprague-Dawley , Receptores Fc/metabolismo
4.
AAPS J ; 20(2): 28, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29417397

RESUMEN

The development of novel therapies that can harnass the immune system to eradicate cancer is an area of intensive research. Several new biopharmaceuticals that target the immune system rather than the tumor itself have recently been approved and fundamentally transformed treatment of many cancer diseases. This success has intensified the search for new targets and modalities that could be developed as even more effective therapeutic agents either as monotherapy or in combination. While great benefits of novel immunotherapies in oncology are evident, the safety of these therapies has to also be addressed as their desired pharmacology, immune activation, can lead to "exaggerated" effects and toxicity. This review is focused on the unique challenges of the nonclinical safety assessment of monoclonal antibodies that target immune checkpoint inhibitors and costimulatory molecules. This class of molecules represents several approved drugs and many more drug candidates in clinical development, for which significant experience has been gained. Their development illustrates challenges regarding the predictivity of the animal models for assessing safety and setting starting doses for first-in-human trials as well as the translatability of nonclinical in vitro and in vivo data to the human findings. Based on learnings from the experience to date, factors to consider and novel approaches to explore are discussed to help address the unique safety issues of immuno-oncology drug development.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Inmunoterapia/efectos adversos , Modelos Animales , Neoplasias/terapia , Animales , Anticuerpos Monoclonales/efectos adversos , Productos Biológicos/efectos adversos , Receptores Coestimuladores e Inhibidores de Linfocitos T/agonistas , Receptores Coestimuladores e Inhibidores de Linfocitos T/antagonistas & inhibidores , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Evaluación Preclínica de Medicamentos/normas , Humanos , Inmunoterapia/métodos , Neoplasias/inmunología , Especificidad de la Especie
5.
J Immunotoxicol ; 11(1): 1-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23746314

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/terapia , Herpesvirus Humano 4/inmunología , Inmunoterapia/métodos , Linfoma de Células B/terapia , Animales , Linfocitos B/virología , Transformación Celular Neoplásica , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Humanos , Huésped Inmunocomprometido , Inmunomodulación , Linfoma de Células B/etiología , Linfoma de Células B/inmunología , Medición de Riesgo
6.
J Immunotoxicol ; 10(1): 106-17, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23173903

RESUMEN

Functional innate immune assessments, including phagocytosis and respiratory burst, are at the forefront of immunotoxicology evaluation in pre-clinical animal species. Although in the clinic and in academic science, phagocytosis, and respiratory burst assessments have been reported for over two decades, the implementation of phagocytosis and respiratory burst analyses in toxicology safety programs is just recently gaining publicity. Discussed herein are general methods, both microtiter plate-based and flow cytometric-based, for assessing phagocytosis and respiratory burst in pre-clinical species including mouse, rat, dog, and monkey. This methods-centric discussion includes a review of technologies and descriptions of method applications, with examples of results from analyses testing reported inhibitors (rottlerin, wortmannin, and SB203580) of phagocytosis and respiratory burst. Justification of implementation, strategic experimental design planning, and feasibility aspects of evaluating test article effects on phagocytosis and respiratory burst function are described within the context of a case study. The case study involves investigation of the effects of a small molecule p38 kinase inhibitor, BMS-582949, on phagocytosis and respiratory burst functions in rat and monkey neutrophils and monocytes in vitro, as well as ex vivo in these innate immune cells from monkeys administered BMS-582949 during a 1-week repeat dose investigative study. The results of the in vitro and ex vivo assessments demonstrated that BMS-582949 inhibited phagocytosis and respiratory burst. These findings correlated with incidences of opportunistic infections observed in rat and monkey toxicity studies.


Asunto(s)
Pruebas Inmunológicas , Fagocitosis/efectos de los fármacos , Pirroles/efectos adversos , Estallido Respiratorio/efectos de los fármacos , Triazinas/efectos adversos , Acetofenonas/farmacología , Androstadienos/farmacología , Animales , Benzopiranos/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Evaluación Preclínica de Medicamentos , Haplorrinos , Humanos , Imidazoles/farmacología , Inmunidad Innata/efectos de los fármacos , Ratones , Monocitos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Piridinas/farmacología , Pirroles/administración & dosificación , Ratas , Triazinas/administración & dosificación , Wortmanina
7.
Toxicol Sci ; 127(1): 159-68, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22331490

RESUMEN

The toxicokinetics and toxicity profile of belatacept (LEA29Y), which blocks the CD28 costimulation pathway to prevent T-cell activation, were evaluated in cynomolgus monkeys. In the current study, 30 monkeys (five monkeys per sex per group) received an intravenous dose of belatacept (10, 22, or 50 mg/kg) once weekly for 6 months. An additional five monkeys per sex received saline intravenously and served as controls. Systemic exposure to belatacept was dose proportional and similar for both sexes. Multiple dosing resulted in moderate belatacept accumulation (1.6- to 1.9-fold). Belatacept was clinically well tolerated in monkeys, with no drug-related laboratory parameter changes or target organ toxicity observed, including a lack of nephrotoxicity. Drug-related changes, which were reversible and related to the pharmacology, included dose-dependent minimal/mild reduction in the size and number of lymphoid germinal centers of the spleen and lymph nodes and minimal reductions in serum IgG levels. No antibodies specific for belatacept were detected during the treatment period. There were no changes in peripheral blood or splenic lymphocyte subpopulations or indications of autoimmune-like inflammation, infection, or malignancy, including preneoplastic changes. Functional recovery of the immune system was noted at all doses by a robust antibody response to keyhole limpet hemocyanin following immunization 2 months after the last belatacept dose was administered. Thus, belatacept was well tolerated in monkeys treated for 6 months at weekly doses up to 50 mg/kg, which represented a 20-fold increase above exposures achieved by the approved maintenance dose in kidney transplant recipients. These findings support the belatacept safety profile and demonstrate that belatacept does not result in adverse renal effects.


Asunto(s)
Formación de Anticuerpos/efectos de los fármacos , Inmunoconjugados/toxicidad , Inmunosupresores/toxicidad , Macaca fascicularis , Linfocitos T/efectos de los fármacos , Abatacept , Animales , Formación de Anticuerpos/inmunología , Área Bajo la Curva , Antígenos CD28/metabolismo , Femenino , Centro Germinal/efectos de los fármacos , Centro Germinal/patología , Hemocianinas/administración & dosificación , Hemocianinas/inmunología , Inmunoconjugados/farmacocinética , Inmunoglobulinas/sangre , Inmunosupresores/farmacocinética , Inyecciones Intravenosas , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/patología , Masculino , Transducción de Señal/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología , Linfocitos T/patología , Pruebas de Toxicidad Crónica
8.
J Rheumatol ; 34(12): 2365-73, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18050376

RESUMEN

OBJECTIVE: The immunogenicity of abatacept, a selective costimulation modulator, administered intravenously, was assessed across Phase II and III trials in patients with rheumatoid arthritis (RA). METHODS: Two direct-format enzyme-linked immunosorbent assays evaluated antibody responses [whole abatacept molecule (CTLA-4 and Ig portion) and CTLA-4 portion only (Assay A)] in the Phase II trials. During the Phase III trials and 2-year open-label periods, a similar, but more sensitive, Assay B was employed. Serum samples collected prestudy, during treatment, and 56 and/or 85 days following the last dose were evaluated. Seropositive samples with anti-CTLA-4 reactivity and sufficiently low drug levels were further characterized for neutralizing activity (cell-based bioassay). RESULTS: A total of 2237 patients with both pre- and post-baseline serum samples were eligible for assessment. Of these, 62 (2.8%) patients demonstrated an anti-abatacept or anti-CTLA-4 response, determined using either Assay A or B. Using the more sensitive Assay B, 60 of 1990 patients (3.0%) demonstrated an antibody response to the whole abatacept molecule (n = 41, 2.1%) or the CTLA-4 portion (n = 19, 1.0%). Of the 1764 RA patients evaluated in the Phase III studies, 203 discontinued therapy and had sera collected 56 and/or 85 days after discontinuation. Patients who discontinued had a higher incidence of immunogenicity versus patients who did not discontinue (7.4% vs 2.6%, respectively). Of 20 patients positive for anti-CTLA-4 reactivity, 13 were eligible for assessment with the neutralization bioassay. Of these, 8 patients exhibited neutralizing activity. Seroconversion occurred with no adverse safety outcomes or effect on pharmacokinetic parameters. No consistent pattern was observed between antibody response and loss of efficacy (American College of Rheumatology 20 and Health Assessment Questionnaire responses). CONCLUSION: Abatacept was associated with a low incidence of immunogenicity in patients with RA and lacked any adverse sequelae.


Asunto(s)
Artritis Reumatoide/inmunología , Inmunoconjugados/farmacología , Inmunosupresores/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Abatacept , Artritis Reumatoide/tratamiento farmacológico , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoconjugados/uso terapéutico , Inmunosupresores/uso terapéutico , Masculino
9.
Arthritis Rheum ; 56(8): 2557-65, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17665452

RESUMEN

OBJECTIVE: Treatment of rheumatoid arthritis and other autoimmune disorders with anti-tumor necrosis factor (anti-TNF) agents is associated with an increased risk of reactivation of latent Mycobacterium tuberculosis. While the mechanism of action of abatacept is fundamentally different from that of anti-TNF therapies, its effect on the protective response to latent tuberculosis is not known. We undertook this study to determine the effect of abatacept treatment in a murine model of chronic M tuberculosis infection. METHODS: Chronic M tuberculosis infection was established in C57BL/6 mice. Four months after infection, mice were treated for up to 16 weeks with abatacept, anti-murine TNF antibody, or vehicle. The primary end point was survival; body weight, bacterial load, histologic features, interferon-gamma (IFNgamma) production by T cells, and cellular infiltration were also assessed. RESULTS: Abatacept- and vehicle-treated groups both maintained control of M tuberculosis infection, with 100% survival after 16 weeks of treatment. These 2 groups had no significant differences in body weight, no clinically relevant differences in bacterial load in the lungs, lymph nodes, or spleen, and no differences in the mean percentage of total or activated T cells, macrophages, neutrophils, or B cells, or in IFNgamma production in the lung or lymph nodes. In contrast, 100% mortality was seen in the anti-TNF antibody-treated group by week 9, with significant body weight loss and increased bacterial load in the lungs, lymph nodes, and spleen. Furthermore, the anti-TNF antibody-treated group had increased pathology consistent with the exacerbation of M tuberculosis infection. CONCLUSION: Abatacept did not impair the ability of mice to control a chronic M tuberculosis infection. In contrast, mice treated with anti-TNF therapy showed increased pathology and bacterial load, with 100% mortality by week 9. The clinical significance of these findings has not yet been determined.


Asunto(s)
Antirreumáticos/efectos adversos , Inmunoconjugados/efectos adversos , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Abatacept , Animales , Anticuerpos/farmacología , Formación de Anticuerpos/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Interferón gamma/metabolismo , Longevidad/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/microbiología , Pulmón/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/microbiología , Bazo/patología , Tasa de Supervivencia , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología , Tuberculosis/mortalidad , Tuberculosis/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/inmunología
10.
J Immunotoxicol ; 4(2): 165-9, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18958726

RESUMEN

Evaluation of the immunotoxicity potential of some pharmaceuticals, including immunomodulatory chemicals and biologics, cannot be limited to testing in rodents. Thus, immune function tests have also been applied in studies with non-human primates and more recently dogs that assess various components of the immune system. These assays include TDAR responses with various immunogens, lymphocyte phenotyping, natural-killer cell activity, delayed-type hypersensitivity, and macrophage function assays. Approaches for incorporating immune function testing in non-rodent species, results from these tests, their interpretation and limitations with respect to drug safety assessment will be reviewed.

11.
J Immunotoxicol ; 1(3): 201-5, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18958653

RESUMEN

Immune-mediated drug hypersensitivity reactions (IDHR) are relatively rare reactions to drugs that can be observed in a limited population of patients, yet these reactions can have significant impacts on public health, clinical practice, and drug development. Despite the potentially significant impact of IDHR, research into the causes and mechanisms of action of these reactions has been limited. In order to identify and enhance potential research opportunities in IDHR, the Health and Environmental Sciences Institute (HESI) hosted a two-day workshop involving stakeholders from government, academia, and industry. Discussions focused on ways to increase IDHR research opportunities within both presently existing collaborative structures and new networks. Based on these discussions, workshop participants concluded that a volunteer organization of interested stakeholders could be established to provide for ongoing advocacy and coordination of efforts related to IDHR research. The primary objectives of such an organization would be to increase public awareness of the impact of IDHR, encourage multidisciplinary IDHR research and training, encourage the development and funding of IDHR research network and seed grants, and to establish a framework for the further exchange and dissemination of IDHR information.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...