Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(2): e0298240, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38315680

RESUMEN

PF-07209960 is a novel bispecific fusion protein composed of an anti-PD-1 antibody and engineered IL-15 cytokine mutein with reduced binding affinity to its receptors. The pharmacokinetics (PK), pharmacodynamics (PD), and toxicity of PF-07209960 were evaluated following once every other week subcutaneous (SC) or intravenous (IV) administration to cynomolgus monkeys in a repeat-dose PKPD (0.01-0.3 mg/kg/dose) and GLP toxicity study (0.1-3 mg/kg/dose). PF-07209960 showed dose dependent pharmacokinetics with a terminal T1/2 of 8 and 13 hours following IV administration at 0.03 and 0.1 mg/kg, respectively. The clearance is faster than a typical IgG1 antibody. Slightly faster clearance was also observed following the second dose, likely due to increased target pool and formation of anti-drug antibodies (ADA). Despite a high incidence rate of ADA (92%) observed in GLP toxicity study, PD-1 receptor occupancy, IL-15 signaling (STAT5 phosphorylation) and T cell expansion were comparable following the first and second doses. Activation and proliferation of T cells were observed with largest increase in cell numbers found in gamma delta T cells, followed by CD4+ and CD8+ T cells, and then NK cells. Release of cytokines IL-6, IFNγ, and IL-10 were detected, which peaked at 72 hours postdose. There was PF-07209960-related mortality at ≥1 mg/kg. At scheduled necropsy, microscopic findings were generalized mononuclear infiltration in various tissues. Both the no observed adverse effect level (NOAEL) and the highest non severely toxic dose (HNSTD) were determined to be 0.3 mg/kg/dose, which corresponded to mean Cmax and AUC48 values of 1.15 µg/mL and 37.9 µg*h/mL, respectively.


Asunto(s)
Anticuerpos Monoclonales , Receptor de Muerte Celular Programada 1 , Animales , Macaca fascicularis , Interleucina-15 , Administración Intravenosa , Citocinas , Inhibidores de Puntos de Control Inmunológico
2.
Toxicol Sci ; 194(1): 53-69, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37228089

RESUMEN

The degradation tag (dTAG) system for target protein degradation can remove proteins from biological systems without the drawbacks of some genetic methods, such as slow kinetics, lack of reversibility, low specificity, and the inability to titrate dosage. These drawbacks can make it difficult to compare toxicity resulting from genetic and pharmacological interventions, especially in vivo. Because the dTAG system has not been studied extensively in vivo, we explored the use of this system to study the physiological sequalae resulting from CDK2 or CDK5 degradation in adult mice. Mice with homozygous knock-in of the dTAG sequence onto CDK2 and CDK5 were born at Mendelian ratios despite decreased CDK2 or CDK5 protein levels in comparison with wild-type mice. In bone marrow cells and duodenum organoids derived from these mice, treatment with the dTAG degrader dTAG-13 resulted in rapid and robust protein degradation but caused no appreciable change in viability or the transcriptome. Repeated delivery of dTAG-13 in vivo for toxicity studies proved challenging; we explored multiple formulations in an effort to maximize degradation while minimizing formulation-related toxicity. Degradation of CDK2 or CDK5 in all organs except the brain, where dTAG-13 likely did not cross the blood brain barrier, only caused microscopic changes in the testis of CDK2dTAG mice. These findings were corroborated with conditional CDK2 knockout in adult mice. Our results suggest that the dTAG system can provide robust protein degradation in vivo and that loss of CDK2 or CDK5 in adult mice causes no previously unknown phenotypes.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Proteínas , Masculino , Ratones , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas/metabolismo , Proteolisis
3.
Toxicol Pathol ; 50(7): 871-880, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36200575

RESUMEN

Mylotarg (Gemtuzumab ozogamicin [GO]), an antibody drug conjugate comprising a CD33-directed antibody linked to calicheamicin, is approved for use in certain acute myeloid leukemia patients. Following reports of prolonged thrombocytopenia and hemorrhagic events in a subset of patients, a detailed series of in vitro and ex vivo studies was performed at the request of regulators, both to look at the effects of GO on platelet production and to determine whether treatment with GO was likely to affect platelet aggregation under a variety of conditions. Treatment with GO resulted in cellular cytotoxicity and/or decreased differentiation during human megakaryocyte development. However, GO did not impair platelet aggregation under the experimental conditions evaluated. Ultimately, the effect of GO on megakaryocyte development observed in our studies was determined to have no impact on the risk-benefit assessment in the intended patient population, as thrombocytopenia is a known side effect of GO, and monitoring of platelet counts in patients is already strongly recommended.


Asunto(s)
Megacariocitos , Trombocitopenia , Humanos , Gemtuzumab , Aminoglicósidos/toxicidad , Anticuerpos Monoclonales Humanizados , Proliferación Celular , Trombocitopenia/inducido químicamente , Trombocitopenia/tratamiento farmacológico
4.
Toxicol Pathol ; 50(7): 867-870, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36305575

RESUMEN

Hematology and bone marrow analysis is central to our understanding of the hematopoietic system and how it responds to insults, and this session presented during the 2022 STP symposium provided a review of current and novel approaches for the evaluation of the hematopoietic system in the context of nonclinical investigations. This publication summarizes the information presented on novel approaches for evaluation of the hematopoietic system using automated hematology analyzers, including details around the quantitative assessment of bone marrow cell suspensions as well as introducing several newly available hematology parameters. It was followed by a discussion on intravital microscopy and live cell imaging and how these methods can assist with de-risking hematopoiesis-associated safety concerns, and a review of recent assays using artificial intelligence for the evaluation of bone marrow.


Asunto(s)
Inteligencia Artificial , Sistema Hematopoyético , Hematopoyesis , Células de la Médula Ósea , Médula Ósea
5.
Toxicol Pathol ; 50(7): 890-894, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36117463

RESUMEN

The session on the hemostatic system focused on new developments in coagulation and platelet biology as well as how therapeutic agents may affect hemostasis. The classic cascade model of coagulation was compared with the more recent models of cell-based and vascular-based coagulation, which may provide better insight on how the coagulation cascade works in vivo. A review of platelet biology highlighted that, as platelets age, desialylated platelets form and are recognized by Ashwell-Morell receptor (AMR), leading to hepatic uptake and subsequent increase in thrombopoietin (TPO) production. Administration of therapeutics that induce thrombocytopenia was also discussed, including Mylotarg, which is an antibody-drug conjugate that was shown to decrease human megakaryocyte development but had no effect on platelet aggregation. An acetyl co-A carboxylase inhibitor was shown to cause thrombocytopenia by inhibiting de novo lipogenesis, which is critical for the formation of the megakaryocyte demarcation membrane system responsible for platelet production. It was also illustrated how preclinical translation models have been very helpful in the development of adeno-associated virus (AAV) hemophilia B gene therapy and what old and new preclinical tools we have that can predict the risk of a prothrombotic state in people.


Asunto(s)
Hemostáticos , Trombocitopenia , Humanos , Hemostasis , Trombopoyetina/genética , Trombocitopenia/inducido químicamente , Plaquetas
6.
Int J Toxicol ; 41(4): 276-290, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35603517

RESUMEN

COVID-19 is a potentially fatal infection caused by the SARS-CoV-2 virus. The SARS-CoV-2 3CL protease (Mpro) is a viral enzyme essential for replication and is the target for nirmatrelvir. Paxlovid (nirmatrelvir co-administered with the pharmacokinetic enhancer ritonavir) showed efficacy in COVID-19 patients at high risk of progressing to hospitalization and/or death. Nonclinical safety studies with nirmatrelvir are essential in informing benefit-risk of Paxlovid and were conducted to support clinical development. In vivo safety pharmacology assessments included a nervous system/pulmonary study in rats and a cardiovascular study in telemetered monkeys. Potential toxicities were assessed in repeat dose studies of up to 1 month in rats and monkeys. Nirmatrelvir administration (1,000 mg/kg, p.o.) to male rats produced transient increases in locomotor activity and respiratory rate but did not affect behavioral endpoints in the functional observational battery. Cardiovascular effects in monkeys were limited to transient increases in blood pressure and decreases in heart rate, observed only at the highest dose tested (75 mg/kg per dose b.i.d; p.o.). Nirmatrelvir did not prolong QTc-interval or induce arrhythmias. There were no adverse findings in repeat dose toxicity studies up to 1 month in rats (up to 1,000 mg/kg daily, p.o.) or monkeys (up to 600 mg/kg daily, p.o.). Nonadverse, reversible clinical pathology findings without clinical or microscopic correlates included prolonged coagulation times at ≥60 mg/kg in rats and increases in transaminases at 600 mg/kg in monkeys. The safety pharmacology and nonclinical toxicity profiles of nirmatrelvir support clinical development and use of Paxlovid for treatment of COVID-19.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Animales , Antivirales/efectos adversos , Masculino , Ratas
7.
Vet Clin Pathol ; 48(3): 389-399, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31556157

RESUMEN

Clinical pathology reporting practices are diverse among individuals and organizations involved in nonclinical toxicology studies. Clear, informative, and consistent reporting of clinical pathology results increases their value and avoids misinterpretation, resulting in decreased drug development costs. In recent years, certain common practices in clinical pathology reporting have been embraced by industry leaders and more consistently utilized across the pharmaceutical industry. The purpose of this manuscript is to review current clinical pathology reporting practices and to provide nonbinding suggestions to improve consistency, quality, and value of clinical pathology reports generated in support of nonclinical toxicology studies.


Asunto(s)
Patología Clínica , Informe de Investigación , Toxicología , Animales , Exactitud de los Datos , Desarrollo de Medicamentos , Humanos , Informe de Investigación/normas , Escritura/normas
8.
Vet Clin Pathol ; 48(3): 383-388, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31549731

RESUMEN

The interpretation of clinical pathology results from nonclinical safety studies is a fundamental component in hazard identification of new drug candidates. The ever-increasing complexity of nonclinical safety studies and sophistication of modern analytical methods have made the interpretation of clinical pathology information by a highly trained subject matter expert imperative. Certain interpretive techniques are particularly effective in the identification and characterization of clinical pathology effects. The purpose of this manuscript is to provide an overview of contemporary interpretive practices for clinical pathology results and to provide nonbinding recommendations aimed at improving consistency, quality, and overall value of clinical pathology interpretations generated in support of nonclinical toxicology studies.


Asunto(s)
Patología Clínica , Toxicología , Medicina Veterinaria , Animales , Investigación Biomédica
9.
Clin Cancer Res ; 25(15): 4735-4748, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31085720

RESUMEN

PURPOSE: Immune checkpoint inhibitors (ICI) targeting PD1, PDL1, or CTLA4 are associated with immune-related adverse events (irAE) in multiple organ systems including myocarditis. The pathogenesis and early diagnostic markers for ICI-induced myocarditis are poorly understood, and there is currently a lack of laboratory animal model to enhance our understanding. We aimed to develop such a model using cynomolgus monkeys. EXPERIMENTAL DESIGN: Chinese-origin cynomolgus monkeys were dosed intravenously with vehicle or nivolumab 20 mg/kg plus ipilimumab 15 mg/kg once weekly and euthanized on day 29. RESULTS: Multiple organ toxicities were observed in cynomolgus monkeys, and were characterized by loose feces, lymphadenopathy, and mononuclear cell infiltrations of varying severity in heart, colon, kidneys, liver, salivary glands, and endocrine organs. Increased proliferation of CD4+ and CD8+ T lymphocytes as well as an increase in activated T cells and central memory T cells in the blood, spleen, and lymph nodes, were observed. Transcriptomic analysis suggested increased migration and activation of T cells and increased phagocytosis and antigen presentation in the heart. Mononuclear cell infiltration in myocardium was comprised primarily of T cells, with lower numbers of macrophages and occasional B cells, and was associated with minimal cardiomyocyte degeneration as well as increases in cardiac troponin-I and NT-pro-BNP. Morphologically, cardiac lesions in our monkey model are similar to the reported ICI myocarditis in humans. CONCLUSIONS: We have developed a monkey model characterized by multiple organ toxicities including myocarditis. This model may provide insight into the immune mechanisms and facilitate biomarker identification for ICI-associated irAEs.


Asunto(s)
Antineoplásicos Inmunológicos/toxicidad , Factores Inmunológicos/toxicidad , Inflamación/etiología , Linfocitos Infiltrantes de Tumor/inmunología , Miocarditis/inducido químicamente , Neoplasias/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/patología , Ipilimumab/toxicidad , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Macaca fascicularis , Miocarditis/inmunología , Miocarditis/patología , Neoplasias/inmunología , Neoplasias/patología , Nivolumab/toxicidad
10.
PLoS One ; 14(1): e0210995, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30677061

RESUMEN

Chemotherapy-induced peripheral neuropathy (CiPN) is a serious adverse effect in the clinic, but nonclinical assessment methods in animal studies are limited to labor intensive behavioral tests or semi-quantitative microscopic evaluation. Hence, microRNA (miRNA) biomarkers and automated in-life behavioral tracking were assessed for their utility as non-invasive methods. To address the lack of diagnostic biomarkers, we explored miR-124, miR-183 and miR-338 in a CiPN model induced by paclitaxel, a well-known neurotoxic agent. In addition, conventional and Vium's innovative Digital Vivarium technology-based in-life behavioral tests and postmortem microscopic examination of the dorsal root ganglion (DRG) and the sciatic nerve were performed. Terminal blood was collected on days 8 or 16, after 20 mg/kg paclitaxel was administered every other day for total of 4 or 7 doses, respectively, for plasma miRNA quantification by RT-qPCR. DRG and sciatic nerve samples were collected from mice sacrificed on day 16 for miRNA quantification. Among the three miRNAs analyzed, only miR-124 was statistically significantly increased (5 fold and 10 fold on day 8 and day 16, respectively). The increase in circulating miR-124 correlated with cold allodynia and axonal degeneration in both DRG and sciatic nerve. Automated home cage motion analysis revealed for the first time that nighttime motion was significantly decreased (P < 0.05) in paclitaxel-dosed animals. Although both increase in circulating miR-124 and decrease in nighttime motion are compelling, our results provide positive evidence warranting further testing using additional peripheral nerve toxicants and diverse experimental CiPN models.


Asunto(s)
Antineoplásicos/toxicidad , MicroARN Circulante/sangre , Enfermedades del Sistema Nervioso Periférico/sangre , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Animales , Antineoplásicos Fitogénicos/toxicidad , Automatización , Conducta Animal/efectos de los fármacos , Biomarcadores/sangre , MicroARN Circulante/genética , MicroARN Circulante/metabolismo , Modelos Animales de Enfermedad , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Hiperalgesia/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/sangre , MicroARNs/genética , MicroARNs/metabolismo , Movimiento , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/patología , Paclitaxel/toxicidad , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología
11.
Mol Cancer Res ; 15(11): 1531-1541, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28760782

RESUMEN

Genetic deletion of cyclin-dependent kinase 4 (Cdk4) is associated with pancreatic beta cell loss and glucose dysregulation in rodents. Palbociclib, one of the first selective CDK4/6 inhibitors approved for the treatment of advanced breast cancer, is currently being investigated as an adjuvant treatment in patients with early-stage breast cancer and in a variety of cancers covering a wide-range of patient populations. Hence, longer chronic toxicity studies were necessary to further examine its safety profile. The effects of different doses and duration of palbociclib administration on glucose and beta cell homeostasis in young (two months) versus aged (12 months) rats was compared. Glucose dysregulation, due to pancreatic beta cell degeneration, was observed in young rats administered the highest dose of palbociclib for 6 months. Abnormal pancreatic islet histology and activation of the endoplasmic reticulum stress response in beta cells were detected after shorter administration with high-dose palbociclib in young rats. To test the hypothesis that palbociclib-associated inhibition of beta cell proliferation will more profoundly affect younger animals that have not achieved replicative quiescence, we administered high-dose palbociclib to aged rats for 6 months. In contrast to the young rats, despite equivalent exposures to palbociclib, no evidence of impaired glucose tolerance, hypoinsulinemia, beta cell vacuolization, or beta cell loss was seen in aged rats. Palbociclib administration induces beta cell failure in young but not aged rats.Implications: Although adult humans receiving palbociclib have not displayed detectable adverse effects on glucose metabolism, the risk of beta cell failure in children remains unexplored. Mol Cancer Res; 15(11); 1531-41. ©2017 AACR.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antineoplásicos/administración & dosificación , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Piperazinas/administración & dosificación , Piridinas/administración & dosificación , Envejecimiento/metabolismo , Animales , Antineoplásicos/efectos adversos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Estrés del Retículo Endoplásmico/efectos de los fármacos , Homeostasis/efectos de los fármacos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Piperazinas/efectos adversos , Piridinas/efectos adversos , Ratas , Ratas Sprague-Dawley
12.
Toxicol Pathol ; 45(2): 260-266, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28056663

RESUMEN

There is limited direction in the literature or regulatory guidance on determination of adversity for clinical pathology (CP) biomarkers in preclinical safety studies. Toxicologic clinical pathologists representing the American Society for Veterinary Clinical Pathology-Regulatory Affairs Committee and Society of Toxicologic Pathology-Clinical Pathology Interest Group identified principles, overall approach, and unique considerations for assessing adversity in CP data interpretation to provide a consensus opinion. Emphasized is the need for pathophysiologic context and a weight-of-evidence approach. Most CP biomarkers do not have the potential to be adverse in isolation, regardless of magnitude of change. Rather, they quantify or describe the impact of effects, provide adjunct or supportive information regarding a process or pathogenesis, and provide translational biomarkers of effect. Most often, CP changes are part of a constellation of findings that collectively are adverse. Thus, most CP changes must be interpreted in conjunction with other study findings and require contextual and integrative interpretation. Exceptions include critical CP changes without correlates that indicate a health risk in the tested species. Overall, CP changes should not be interpreted in isolation and their adversity is best addressed with an integrated approach.


Asunto(s)
Biomarcadores/análisis , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Guías como Asunto , Patología Clínica/normas , Patología Veterinaria/normas , Pruebas de Toxicidad/normas , Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/veterinaria , Humanos , Nivel sin Efectos Adversos Observados , Control de Calidad , Medición de Riesgo , Pruebas de Toxicidad/veterinaria
13.
Toxicol Pathol ; 45(2): 302-320, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27189632

RESUMEN

Gastrointestinal toxicity is dose limiting with many therapeutic and anticancer agents. Real-time, noninvasive detection of markers of toxicity in biofluids is advantageous. Ongoing research has revealed microRNAs as potential diagnostic and predictive biomarkers for the detection of select organ toxicities. To study the potential utility of microRNA biomarkers of intestinal injury in a preclinical toxicology species, we evaluated 3 rodent models of drug-induced intestinal toxicity, each with a distinct mechanism of toxicity. MiR-215 and miR-194 were identified as putative intestinal toxicity biomarkers. Both were evaluated in plasma and feces and compared to plasma citrulline, an established intestinal injury biomarker. Following intestinal toxicant dosing, microRNA changes in feces and plasma were detected noninvasively and correlated with histologic evidence of intestinal injury. Fecal miR-215 and miR-194 levels increased, and plasma miR-215 decreased in a dose- and time-dependent manner. Dose-dependent decreases in plasma miR-215 levels also preceded and correlated positively with plasma citrulline modulation, suggesting miR-215 is a more sensitive biomarker. Moreover, during the drug-free recovery phase, plasma miR-215 returned to predose levels, supporting a corresponding recovery of histologic lesions. Despite limitations, this study provides preliminary evidence that select microRNAs have the potential to act as noninvasive, sensitive, and quantitative biomarkers of intestinal injury.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/diagnóstico , Heces/química , Mucosa Intestinal/efectos de los fármacos , MicroARNs/sangre , Pruebas de Toxicidad/normas , Animales , Biomarcadores/análisis , Biomarcadores/sangre , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/sangre , Masculino , MicroARNs/análisis , Ratas Wistar , Sensibilidad y Especificidad , Factores de Tiempo
14.
Toxicol Pathol ; 45(2): 321-334, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28013573

RESUMEN

Detecting and monitoring exocrine pancreatic damage during nonclinical and clinical testing is challenging because classical biomarkers amylase and lipase have limited sensitivity and specificity. Novel biomarkers for drug-induced pancreatic injury are needed to improve safety assessment and reduce late-stage attrition rates. In a series of studies, miR-216a and miR-217 were evaluated as potential biomarkers of acute exocrine pancreatic toxicity in rats. Our results revealed that miR-216a and miR-217 were almost exclusively expressed in rat pancreas and that circulating miR-216a and miR-217 were significantly increased in rats following administration of established exocrine pancreatic toxicants caerulein (CL) and 1-cyano-2-hydroxy-3-butene (CHB) as well as in rats administered a proprietary molecule known to primarily affect the exocrine pancreas. Conversely, neither microRNA was increased in rats administered a proprietary molecule known to cause a lesion at the pancreatic endocrine-exocrine interface (EEI) or in rats administered an established renal toxicant. Compared with amylase and lipase, increases in miR-216a and miR-217 were of greater magnitude, persisted longer, and/or correlated better with microscopic findings within the exocrine pancreas. Our findings demonstrate that in rats, miR-216a and miR-217 are sensitive and specific biomarkers of acute exocrine pancreatic toxicity that may add value to the measurement of classical pancreatic biomarkers.


Asunto(s)
Insuficiencia Pancreática Exocrina/sangre , MicroARNs/sangre , Páncreas Exocrino/efectos de los fármacos , Enfermedad Aguda , Alquenos/toxicidad , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Ceruletida/toxicidad , Insuficiencia Pancreática Exocrina/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Nitrilos/toxicidad , Especificidad de Órganos , Páncreas Exocrino/metabolismo , Páncreas Exocrino/patología , Ratas Sprague-Dawley , Ratas Wistar , Sensibilidad y Especificidad
15.
Toxicol Pathol ; 44(2): 163-72, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26879687

RESUMEN

The Society of Toxicologic Pathology formed a working group in collaboration with the American Society for Veterinary Clinical Pathology to provide recommendations for the appropriate inclusion of clinical pathology evaluation in recovery arms of nonclinical toxicity studies but not on when to perform recovery studies. Evaluation of the recovery of clinical pathology findings is not required routinely but provides useful information on risk assessment in nonclinical toxicity studies and is recommended when the ability of the organ to recover is uncertain. The study design generally requires inclusion of concurrent controls to separate procedure-related changes from test article-related changes, but return of clinical pathology values toward baseline may be sufficient in some cases. Evaluation of either a select or full panel of standard hematology, coagulation, and serum and urine chemistry biomarkers can be scientifically justified. It is also acceptable to redesignate dosing phase animals to the recovery phase or vice versa to optimize data interpretation. Assessment of delayed toxicity during the recovery phase is not required but may be appropriate in development programs with unique concerns. Evaluation of the recovery of clinical pathology data for vaccine development is required and, for efficacy markers, is recommended if it furthers pharmacologic understanding.


Asunto(s)
Investigación Biomédica , Patología Clínica , Pruebas de Toxicidad/normas , Animales , Investigación Biomédica/legislación & jurisprudencia , Investigación Biomédica/normas , Perros , Haplorrinos , Ratones , Patología Clínica/legislación & jurisprudencia , Patología Clínica/normas , Ratas , Proyectos de Investigación , Medición de Riesgo
16.
Mol Genet Metab ; 114(2): 217-25, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25092414

RESUMEN

Recombinant human acid sphingomyelinase (rhASM) is being developed as an enzyme replacement therapy for patients with acid sphingomyelinase deficiency (Niemann-Pick disease types A and B), which causes sphingomyelin to accumulate in lysosomes. In the acid sphingomyelinase knock-out (ASMKO) mouse, intravenously administered rhASM reduced tissue sphingomyelin levels in a dose-dependent manner. When rhASM was administered to normal rats, mice, and dogs, no toxicity was observed up to a dose of 30mg/kg. However, high doses of rhASM≥10mg/kg administered to ASMKO mice resulted in unexpected toxicity characterized by cardiovascular shock, hepatic inflammation, adrenal hemorrhage, elevations in ceramide and cytokines (especially IL-6, G-CSF, and keratinocyte chemoattractant [KC]), and death. The toxicity could be completely prevented by the administration of several low doses (3mg/kg) of rhASM prior to single or repeated high doses (≥20mg/kg). These results suggest that the observed toxicity involves the rapid breakdown of large amounts of sphingomyelin into ceramide and/or other toxic downstream metabolites, which are known signaling molecules with cardiovascular and pro-inflammatory effects. Our results suggest that the nonclinical safety assessment of novel therapeutics should include the use of specific animal models of disease whenever feasible.


Asunto(s)
Perros , Terapia de Reemplazo Enzimático , Enfermedad de Niemann-Pick Tipo A/tratamiento farmacológico , Esfingomielina Fosfodiesterasa/administración & dosificación , Esfingomielina Fosfodiesterasa/deficiencia , Administración Intravenosa , Glándulas Suprarrenales , Animales , Ceramidas/sangre , Ceramidas/metabolismo , Citocinas/sangre , Citocinas/inmunología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Hígado/metabolismo , Hígado/patología , Lisosomas/metabolismo , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad de Niemann-Pick Tipo A/metabolismo , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/toxicidad , Esfingomielina Fosfodiesterasa/toxicidad , Esfingomielinas/metabolismo
17.
Toxicol Pathol ; 42(4): 765-73, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24616262

RESUMEN

Two 6-month repeat-dose toxicity studies in cynomolgus monkeys illustrated immune complex-mediated adverse findings in individual monkeys and identified parameters that potentially signal the onset of immune complex-mediated reactions following administration of RN6G, a monoclonal antibody (mAb). In the first study, 3 monkeys exhibited nondose-dependent severe clinical signs accompanied by decreased erythrocytes with increased reticulocytes, neutrophilia, monocytosis, thrombocytopenia, coagulopathy, decreased albumin, azotemia, and increased serum levels of activated complement products, prompting unscheduled euthanasia. Histologically, immunohistochemical localization of RN6G was associated with monkey immunoglobulin and complement components in glomeruli and other tissues, attributable to immune complex disease (ICD). All 3 animals also had anti-RN6G antibodies and decreased plasma levels of RN6G. Subsequently, an investigational study was designed and conducted with regulatory agency input to detect early onset of ICD and assess reversibility to support further clinical development. Dosing of individual animals ceased when biomarkers of ICD indicated adverse findings. Of the 12 monkeys, 1 developed anti-RN6G antibodies and decreased RN6G exposure that preceded elevations in complement products, interleukin-6, and coagulation parameters and decreases in albumin and fibrinogen. All findings in this monkey, except for antidrug antibody (ADA), reversed after cessation of dosing without progressing to adverse sequelae typically associated with ICD.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Biomarcadores/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Enfermedades del Complejo Inmune/sangre , Animales , Anticuerpos Monoclonales/sangre , Proteína C-Reactiva/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Enfermedades del Complejo Inmune/inducido químicamente , Enfermedades del Complejo Inmune/patología , Inmunohistoquímica , Interferón gamma/sangre , Interleucina-1beta/sangre , Interleucina-6/sangre , Macaca fascicularis , Masculino , Microscopía Electrónica de Transmisión , Factor de Necrosis Tumoral alfa/sangre , Urinálisis
18.
Toxicol Sci ; 135(2): 451-64, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23872713

RESUMEN

The fibroblast growth factor receptors (FGFR) play a major role in angiogenesis and are desirable targets for the development of therapeutics. Groups of Wistar Han rats were dosed orally once daily for 4 days with a small molecule pan-FGFR inhibitor (5mg/kg) or once daily for 6 days with a small molecule MEK inhibitor (3mg/kg). Serum phosphorous and FGF23 levels increased in all rats during the course of the study. Histologically, rats dosed with either drug exhibited multifocal, multiorgan soft tissue mineralization. Expression levels of the sodium phosphate transporter Npt2a and the vitamin D-metabolizing enzymes Cyp24a1 and Cyp27b1 were modulated in kidneys of animals dosed with the pan-FGFR inhibitor. Both inhibitors decreased ERK phosphorylation in the kidneys and inhibited FGF23-induced ERK phosphorylation in vitro in a dose-dependent manner. A separate cardiovascular outcome study was performed to monitor hemodynamics and cardiac structure and function of telemetered rats dosed with either the pan-FGFR inhibitor or MEK inhibitor for 3 days. Both compounds increased blood pressure (~+ 17 mmHg), decreased heart rate (~-75 bpm), and modulated echocardiography parameters. Our data suggest that inhibition of FGFR signaling following administration of either pan-FGFR inhibitor or MEK inhibitor interferes with the FGF23 pathway, predisposing animals to hyperphosphatemia and a tumoral calcinosis-like syndrome in rodents.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Transducción de Señal , Animales , Secuencia de Bases , Sistema Cardiovascular/metabolismo , Línea Celular , Cartilla de ADN , Factor-23 de Crecimiento de Fibroblastos , Humanos , Fosforilación , Ratas , Ratas Wistar
19.
Toxicol Pathol ; 41(3): 508-18, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22968286

RESUMEN

Animal models of human disease are commonly utilized to gain insight into the potential efficacy and mode of action of novel pharmaceuticals. However, conventional (healthy) rodent and nonrodent models are generally utilized in nonclinical safety testing. Animal models of human disease may be helpful in understanding safety risks of compounds in nonclinical or clinical development, with their greatest value being in targeted or hypothesis-driven studies to help understand the mechanism of a particular toxicity. Limitations of animal models of disease in nonclinical safety testing include a lack of historical control, heterogeneity in disease expression, a limited life span, and confounding effects of the disease. In most instances, animal models of human disease should not be utilized to supplant testing in conventional animal models. While of potential benefit, testing in an animal model of human disease should only be taken after adequate consideration of relevance along with benefits and limitations of the proposed model.


Asunto(s)
Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Pruebas de Toxicidad/métodos , Animales , Humanos , Ratones , Ratas , Medición de Riesgo
20.
Int J Toxicol ; 31(6): 529-36, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23117654

RESUMEN

Kidney injury biomarkers have been utilized by pharmaceutical companies as a means to assess the potential of candidate drugs to induce nephrotoxicity. Multiple platforms and assay methods exist, but the comparison of these methods has not been described. Millipore's Kidney Toxicity panel, EMD/Novagen's Widescreen Kidney Toxicity panel, and Meso Scales Kidney Injury panel were selected based on published information. Kidney injury molecule 1, cystatin C, clusterin, and osteopontin were the 4 biomarkers common among all kits tested and the focus of this study. Rats were treated with a low and high dose of para-aminophenol, a known nephrotoxicant, and urine samples were collected and analyzed on the Bio-Plex 200 or MSD's Sector Imager 6000, according to manufacturers specifications. Comparatively, of the 3 kits, Millipore was the most consistent in detecting elevations of 3 out of the 4 biomarkers at both dose levels and indicated time points.


Asunto(s)
Lesión Renal Aguda/orina , Moléculas de Adhesión Celular/metabolismo , Clusterina/metabolismo , Cistatina C/metabolismo , Osteopontina/metabolismo , Xenobióticos/toxicidad , Lesión Renal Aguda/etiología , Aminofenoles/toxicidad , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Receptor Celular 1 del Virus de la Hepatitis A , Enfermedades Renales , Masculino , Glicoproteínas de Membrana , Mutágenos/toxicidad , Ratas , Ratas Wistar , Juego de Reactivos para Diagnóstico , Receptores Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...