Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Toxicol ; 40(4): 344-354, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33866838

RESUMEN

Phosphatidylinositol 3-kinase (PI3K) δ is a lipid kinase primarily found in leukocytes, which regulates important cell functions. AMG2519493 was a PI3K δ-specific inhibitor in development for treatment of various inflammatory diseases. AMG2519493-related changes in the male and/or female reproductive organs were observed in the 1- and 3-month oral repeat dose toxicology studies in the rat and cynomolgus monkey. Hemorrhagic corpora lutea cysts and increased incidence of corpora lutea cysts without hemorrhage were observed in the ovaries at supra pharmacological doses in the rat. A decrease in seminiferous germ cells in the testis, indicative of spermatogenesis maturation arrest, was observed in both the rat and cynomolgus monkey. Although the characteristics were comparable, the drug systemic exposures associated with the testicular changes were very different between the 2 species. In the rat, the testicular change was only observed at supra pharmacologic exposure. Isotype assessment of PI3K signaling in rat spermatogonia in vitro indicated a role for PI3K ß, but not δ, in the c Kit/PI3K/protein kinase B signaling pathway. Therefore, changes in both the ovary and testis of the rat were considered due to off target effect as they only occurred at suprapharmacologic exposure. In contrast, the testicular changes in the cynomolgus monkey (decrease in seminiferous germ cells) occurred at very low doses associated with PI3K δ-specific inhibition, indicating that the PI3K δ isoform may be important in spermatogenesis maturation in the cynomolgus monkey. Our results suggest species-related differences in PI3K isoform-specific control on reproductive organs.


Asunto(s)
Ovario/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Pirimidinas/farmacología , Quinolinas/farmacología , Testículo/efectos de los fármacos , Animales , Femenino , Macaca fascicularis , Masculino , Ratones , Ovario/enzimología , Ratas , Ratas Sprague-Dawley , Espermatogonias/enzimología , Testículo/enzimología
2.
Regul Toxicol Pharmacol ; 106: 224-238, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31085251

RESUMEN

Calcitonin gene-related peptide (CGRP) and its receptor have been implicated as a key mediator in the pathophysiology of migraine. Thus, erenumab, a monoclonal antibody antagonist of the CGRP receptor, administered as a once monthly dose of 70 or 140 mg has been approved for the preventive treatment of migraine in adults. Due to the species specificity of erenumab, the cynomolgus monkey was used in the pharmacology, pharmacokinetics, and toxicology studies to support the clinical program. There were no effects of erenumab on platelets in vitro (by binding, activation or phagocytosis assays). Specific staining of human tissues with erenumab did not indicated any off-target binding. There were no erenumab-related findings in a cardiovascular safety pharmacology study in cynomolgus monkeys or in vitro in human isolated coronary arteries. Repeat-dose toxicology studies conducted in cynomolgus monkeys at dose levels up to 225 mg/kg (1 month) or up to 150 mg/kg (up to 6 months) with twice weekly subcutaneous (SC) doses showed no evidence of erenumab-mediated adverse toxicity. There were no effects on pregnancy, embryo-fetal or postnatal growth and development in an enhanced pre-postnatal development study in the cynomolgus monkey. There was evidence of placental transfer of erenumab based on measurable serum concentrations in the infants up to 3 months post birth. The maternal and developmental no-observed-effect level (NOEL) was the highest dose tested (50 mg/kg SC Q2W). These nonclinical data in total indicate no safety signal of concern to date and provide adequate margins of exposure between the observed safe doses in animals and clinical dose levels.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Trastornos Migrañosos/prevención & control , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Anticuerpos Monoclonales Humanizados/sangre , Relación Dosis-Respuesta a Droga , Humanos
3.
J Biol Chem ; 292(5): 1876-1883, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-27994063

RESUMEN

The stable effector functionLess (SEFL) antibody was designed as an IgG1 antibody with a constant region that lacks the ability to interact with Fcγ receptors. The engineering and stability and pharmacokinetic assessments of the SEFL scaffold is described in the accompanying article (Jacobsen, F. W., Stevenson, R., Li, C., Salimi-Moosavi, H., Liu, L., Wen, J., Luo, Q., Daris, K., Buck, L., Miller, S., Ho, S-Y., Wang, W., Chen, Q., Walker, K., Wypych, J., Narhi, L., and Gunasekaran, K. (2017) J. Biol. Chem 292). The biological properties of these SEFL antibodies were assessed in a variety of human and cynomolgus monkey in vitro assays. Binding of parent molecules and their SEFL variants to human and cynomolgus monkey FcγRs were evaluated using flow cytometry-based binding assays. The SEFL variants tested showed decreased binding affinity to human and cynomolgus FcγRs compared with the wild-type IgG1 antibody. In addition, SEFL variants demonstrated no antibody-dependent cell-mediated cytotoxicity in vitro against Daudi cells with cynomolgus monkey peripheral blood mononuclear cells, and had minimal complement-dependent cytotoxicity activity similar to that of the negative control IgG2 in a CD20+ human Raji lymphoma cell line. SEFL mutations eliminated off-target antibody-dependent monocyte phagocytosis of cynomolgus monkey platelets, and cynomolgus platelet activation in vitro These experiments demonstrate that the SEFL modifications successfully eliminated Fc-associated effector binding and functions.


Asunto(s)
Anticuerpos Monoclonales , Plaquetas/inmunología , Inmunoglobulina G , Monocitos/inmunología , Fagocitosis/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Receptores de IgG , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Macaca fascicularis , Ratones , Fagocitosis/inmunología , Activación Plaquetaria/inmunología , Receptores de IgG/genética , Receptores de IgG/inmunología
4.
Bone ; 64: 314-25, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24727159

RESUMEN

RANKL is a key regulator of bone resorption and osteoclastogenesis. Denosumab is a fully human IgG2 monoclonal antibody that inhibits bone resorption by binding and inhibiting the activity of RANKL. To determine the effects of denosumab on pre- and postnatal skeletal growth and development, subcutaneous injections of 0 (control) or 50 mg/kg/month denosumab were given to pregnant cynomolgus monkeys from approximately gestation day (GD) 20 until parturition (up to 6 doses). For up to 6 months postpartum (birth day [BD] 180/181), evaluation of the infants included skeletal radiographs, bone biomarkers, and oral examinations for assessment of tooth eruption. Infant bones were collected at necropsy for densitometry, biomechanical testing, and histopathologic evaluation from control and denosumab-exposed infants on BD1 (or within 2 weeks of birth) and BD181, and from infants that died or were euthanized moribund from BD5 to BD69. In all denosumab-exposed infants, biomarkers of bone resorption and formation were markedly decreased at BD1 and BD14 and slightly greater at BD91 vs. control, then similar to control values by BD181. Spontaneous long bone fractures were detected clinically or radiographically in 4 denosumab-exposed infants at BD28 and BD60, with evidence of radiographic healing at ≥BD60. In BD1 infants exposed to denosumab in utero, radiographic evaluations of the skeleton revealed decreased long bone length; a generalized increased radio-opacity of the axial and appendicular skeleton and bones at the base of the skull with decreased or absent marrow cavities, widened growth plates, flared/club-shaped metaphysis, altered jaw/skull shape, and reduced jaw length; and delayed development of secondary ossification centers. Densitometric evaluations in these infants demonstrated a marked increase in bone mineral density at trabecular sites, but cortical bone mineral density was decreased. Histologically, long bone cortices were attenuated and there was an absence of osteoclasts. Bones with active endochondral ossification consisted largely of a dense network of retained primary spongiosa with reduced marrow space consistent with an osteopetrotic phenotype. A minimal increase in growth plate thickness largely due to the expansion of the hypertrophic zone was present. Retained woven bone was observed in bones formed by intramembranous ossification, consistent with absence of bone remodeling. These changes in bone tissue composition and geometry were reflected in reduced biomechanical strength and material properties of bones from denosumab-exposed infants. Material property changes were characterized by increased tissue brittleness reflected in reductions in calculated material toughness at the femur diaphysis and lack of correlation between energy and bone mass at the vertebra; these changes were likely the basis for the increased skeletal fragility (fractures). Although tooth eruption was not impaired in denosumab-exposed infants, the reduced growth and increased bone density of the mandible resulted in dental abnormalities consisting of tooth malalignment and dental dysplasia. Radiographic changes at BD1 persisted at BD28, with evidence of resumption of bone resorption and remodeling observed in most infants at BD60 and/or BD90. In 2 infants euthanized on BD60 and BD69, there was histologic and radiographic evidence of subphyseal/metaphyseal bone resorption accompanied by multiple foci of ossification in growth plates that were markedly increased in thickness. In infants necropsied at BD181, where systemic exposure to denosumab had been below limits of quantitation for approximately 3months, there was largely full recovery from all bone-related changes observed earlier postpartum, including tissue brittleness. Persistent changes included dental dysplasia, decreased bone length, reduced cortical thickness, and decreased peak load and ultimate strength at the femur diaphysis. In conclusion, the skeletal and secondary dental effects observed in infant monkeys exposed in utero to denosumab are consistent with the anticipated pharmacological activity of denosumab as a monoclonal antibody against RANKL and inhibitor of osteoclastogenesis. The resulting inhibition of resorption impaired both bone modeling and remodeling during skeletal development and growth. The skeletal phenotype of these infant monkeys resembles human infants with osteoclast-poor osteopetrosis due to inactivating mutations of RANK or RANKL.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Osteoclastos/patología , Osteopetrosis/patología , Efectos Tardíos de la Exposición Prenatal , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Remodelación Ósea , Denosumab , Femenino , Macaca fascicularis , Osteopetrosis/diagnóstico por imagen , Fenotipo , Embarazo , Tomografía Computarizada por Rayos X , Erupción Dental
5.
Reprod Toxicol ; 42: 27-40, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23886817

RESUMEN

Denosumab is a monoclonal antibody that inhibits bone resorption by targeting RANKL, an essential mediator of osteoclast formation, function, and survival. Reproductive toxicity of denosumab was assessed in cynomolgus monkeys in an embryofetal development study (dosing GD20-50) and a pre-postnatal toxicity study (dosing GD20-parturition). In the embryofetal toxicity study, denosumab did not elicit maternal toxicity, fetal harm or teratogenicity. In the pre-postnatal toxicity study, there were increased stillbirths, and one maternal death due to dystocia. There was no effect on maternal mammary gland histomorphology, lactation, or fetal growth. In infants exposed in utero, there was increased postnatal mortality, decreased body weight gain, and decreased growth/development. Denosumab-related effects in infants were present in bones and lymph nodes. There was full recovery at 6 months of age from most bone-related changes observed earlier postpartum. The effects observed in mothers and infants were consistent with the pharmacological action of denosumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Conservadores de la Densidad Ósea/toxicidad , Animales , Denosumab , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Macaca fascicularis , Masculino , Intercambio Materno-Fetal , Embarazo , Reproducción/efectos de los fármacos , Mortinato
6.
Regul Toxicol Pharmacol ; 66(3): 269-78, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23680404

RESUMEN

According to ICH S6(R1), mating studies are not practical for assessing effects on female fertility of biopharmaceuticals that are pharmacologically active only in non-human primates (NHPs). Instead, fertility should be assessed by evaluating histopathology and organ weights of the reproductive tract in studies of at least 3 months dosing duration using sexually mature NHPs. An assessment of the menstrual cycle in females can be included if there is cause for concern based on pharmacological mode of action or relevant findings in previous studies. However, many factors unrelated to the molecule under evaluation can impact cycle length and thus affect data interpretation. Assessment of a monoclonal antibody in a 6 month repeat dose toxicity study is used as an example in this manuscript to review potential sources of background variability, identify strategies to minimize its impact and recommend optimal ways to collect, present and analyze menstrual cycle data. Experimental variables include the amount of time required for menses to normalize following the transport of animals to the testing facility, stress-related effects on the cycle length due to socialization issues with new cagemates, and the normal background irregularity of cycle length in NHPs. Study related procedures (i.e., animal handling for dosing, blood draws, body weights, ECGs, etc.) did not affect cycle lengths in this study. We show that introducing a number of key experimental control procedures to minimize cycle variability can enable a robust assessment of the effects of a biotherapeutic on menstrual cycling within a chronic toxicity study in NHPs.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Biofarmacia/métodos , Determinación de Punto Final , Fertilidad/efectos de los fármacos , Ciclo Menstrual/efectos de los fármacos , Pruebas de Toxicidad Subcrónica/métodos , Animales , Biofarmacia/normas , Femenino , Macaca fascicularis , Medición de Riesgo , Factores de Tiempo , Pruebas de Toxicidad Subcrónica/normas
7.
Regul Toxicol Pharmacol ; 59(3): 493-502, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20951178

RESUMEN

A multinational pharmaceutical and biotechnology company survey was conducted to gain a better understanding of the use and value of the tissue cross-reactivity (TCR) assay in the development of biotherapeutic molecules. The majority of the molecules did not use TCR data as the only basis for determining species selection for toxicity studies (73%). For 95% of the molecules, the TCR data had no impact on the development strategy. For 2% of the molecules (1/56), TCR data was the sole source of information indicating a potential risk to patients. Unexpected or off-target binding was seen with 35% of the molecules, with the majority of this binding occurring in the CNS and reproductive organs. Tissues that were known or presumed to contain the target stained positively in 22% and 10% of molecules tested in non-human primate and human tissues, respectively. Tissues that were known or presumed to lack the target were negative for staining in 39% and 50% of molecules for non-human primate and human tissue, respectively. For 5% (6/110) of all the molecules, companies stated that toxicities would have been missed in animal studies or the clinic (i.e., not identified by clinical signs, histopathology, etc.) if the TCR studies had not been performed.


Asunto(s)
Biofarmacia , Encuestas Epidemiológicas , Animales , Biofarmacia/métodos , Biofarmacia/tendencias , Biotecnología/métodos , Biotecnología/tendencias , Reacciones Cruzadas/efectos de los fármacos , Reacciones Cruzadas/fisiología , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/tendencias , Industria Farmacéutica/métodos , Industria Farmacéutica/tendencias , Encuestas Epidemiológicas/métodos , Humanos , Inmunohistoquímica , Internet , Macaca fascicularis , Ratones , Preparaciones Farmacéuticas , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
8.
Birth Defects Res B Dev Reprod Toxicol ; 86(6): 446-62, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20025046

RESUMEN

Developmental and reproductive toxicology testing in nonhuman primates (NHPs) has become more common due to the increasing number of biopharmaceuticals in drug development, since NHPs are frequently the only species to express pharmacologic responses similar to humans. NHPs may also be used to help resolve issues associated with small-molecule reproductive toxicology in traditional species (rodents and rabbits). Adequate designs in NHP are presented for developmental toxicity (embryo-fetal development, pre-postnatal development, enhanced pre-postnatal development), reproductive toxicity (male and female), and juvenile toxicity studies. Optional parameters that may be included in these studies are discussed, as are new study designs that consolidate multiple aspects of the reproductive assessment and thereby conserve the limited supply of sexually mature NHPs available for testing. The details described will assist scientists in pharmaceutical, regulatory, and contract research organizations who are involved in conducting these unique studies to optimize their design based on case-by-case considerations.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Haplorrinos/fisiología , Reproducción/efectos de los fármacos , Pruebas de Toxicidad/métodos , Animales , Animales de Laboratorio , Callithrix/fisiología , Femenino , Macaca fascicularis/fisiología , Macaca mulatta/fisiología , Masculino , Preparaciones Farmacéuticas/clasificación , Conejos , Ratas , Reproducción/fisiología , Especificidad de la Especie
9.
Int J Toxicol ; 28(3): 230-53, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19546261

RESUMEN

Although toxicology studies should always be conducted in pharmacologically relevant species, the specificity of many biopharmaceuticals can present challenges in identification of a relevant species. In certain cases, that is, when the clinical product is active only in humans or chimpanzees, or if the clinical candidate is active in other species but immunogenicity limits the ability to conduct a thorough safety assessment, alternative approaches to evaluating the safety of a biopharmaceutical must be considered. Alternative approaches, including animal models of disease, genetically modified mice, or use of surrogate molecules, may improve the predictive value of preclinical safety assessments of species-specific biopharmaceuticals, although many caveats associated with these models must be considered. Because of the many caveats that are discussed in this article, alternative approaches should only be used to evaluate safety when the clinical candidate cannot be readily tested in at least one relevant species to identify potential hazards.


Asunto(s)
Biofarmacia/métodos , Evaluación Preclínica de Medicamentos/métodos , Drogas en Investigación/toxicidad , Pruebas de Toxicidad/métodos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/toxicidad , Biofarmacia/economía , Pruebas de Carcinogenicidad , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/economía , Evaluación Preclínica de Medicamentos/normas , Etiquetado de Medicamentos , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Transgénicos , Embarazo , Proteínas Recombinantes de Fusión/toxicidad , Especificidad de la Especie , Pruebas de Toxicidad/economía , Pruebas de Toxicidad/normas
10.
Regul Toxicol Pharmacol ; 53(3): 226-34, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19217923

RESUMEN

The effects of treatment with a soluble IL-4 receptor (sIL-4R) on reproduction and neonatal development were assessed in pregnant cynomolgus monkeys and mice. When pregnant cynomolgus monkeys were administered a human sIL-4R intravenously twice a week during organogenesis (GD 20-51) at 0, 0.2 or 2.0mg/kg, there was an increase in abortion/embryo-fetal death in the 0.2 (42.9%) and 2.0 (26.3%) mg/kg groups compared to controls (17.6%). All fetuses removed at cesarean sectioning on GD 100-102 were alive and no abnormalities were noted. There were three stillborn neonates (2.0mg/kg group), which were determined to have died before birth. No neonates died after birth and no abnormalities were noted. Due to the unanticipated results in the monkey study, a mouse developmental study with a murine surrogate molecule was conducted. When pregnant Crl:CD-1((R))(ICR)BR mice were administered murine sIL-4R intravenously once daily during the organogenesis period (GD 6-15) at 0, 25, 75, 250, or 625microg/mouse ( approximately 20mg/kg), there were no test-article-related abnormalities in any parameters. Antibody development to the drug did not influence toxicity in the monkey or mouse. In conclusion, evaluation of reproductive effects in mice administered murine soluble IL-4R was not predictive of reproductive effects noted in cynomolgus monkeys administered human soluble IL-4R.


Asunto(s)
Exposición Materna/efectos adversos , Receptores de Interleucina-4 , Proteínas Recombinantes , Reproducción/efectos de los fármacos , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/patología , Animales , Animales Recién Nacidos , Peso Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Humanos , Masculino , Ratones , Tamaño de los Órganos/efectos de los fármacos , Organogénesis/efectos de los fármacos , Embarazo , Receptores de Interleucina-4/administración & dosificación , Receptores de Interleucina-4/química , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/toxicidad , Solubilidad , Especificidad de la Especie
11.
Expert Opin Drug Metab Toxicol ; 4(7): 871-7, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18624676

RESUMEN

BACKGROUND: Preclinical efficacy and safety studies, especially chronic studies, can be difficult to perform when the candidate therapeutic agent is a human protein due to the specificity of these molecules for the human target. The main issues are: i) the human protein or target may not be pharmacologically active in rodents or dogs, the standard toxicology species; or ii) the therapeutic agent may be so immunogenic in these species, that longer duration studies are not possible due to the formation of neutralizing antibodies. Thus, preclinical safety testing of biotherapeutics poses a particular challenge in selecting a relevant animal species for use in toxicology studies. OBJECTIVE: This article will discuss the considerations that are unique to safety assessment of biotherapeutics and will provide alternatives to the standard toxicity testing which is conducted for small molecules. METHODS: This article is based on published information with regards to species selection considerations as well as information from the FDA website on several marketed compounds. In addition, discussions of this topic that have occurred in public forums as well as the experience of the author are considered. CONCLUSION: The most important consideration in species selection for a biotherapeutic is that the drug is pharmacologically active in the preclinical species. This is a key consideration as biotherapeutics are highly targeted and rarely, if ever, demonstrate off-target toxicity. Because of this species specificity, nonhuman primates are often the only relevant species that can be used to assess the safety of a biotherapeutic. Other alternatives such as use of a homologous protein in rodents or the use of transgenic or knockout mice can also be used to assess safety although the caveats to these approaches must be considered.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inducido químicamente , Pruebas de Toxicidad/métodos , Toxicología/métodos , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Humanos , Ratones , Ratones Transgénicos , Primates , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA