Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(10): e2123002119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35235456

RESUMEN

Therapeutic human IgG antibodies are routinely tested in mouse models of oncologic, infectious, and autoimmune diseases. However, assessing the efficacy and safety of long-term administration of these agents has been limited by endogenous anti-human IgG immune responses that act to clear human IgG from serum and relevant tissues, thereby reducing their efficacy and contributing to immune complex­mediated pathologies, confounding evaluation of potential toxicity. For this reason, human antibody treatment in mice is generally limited in duration and dosing, thus failing to recapitulate the potential clinical applications of these therapeutics. Here, we report the development of a mouse model that is tolerant of chronic human antibody administration. This model combines both a human IgG1 heavy chain knock-in and a full recapitulation of human Fc receptor (FcγR) expression, providing a unique platform for in vivo testing of human monoclonal antibodies with relevant receptors beyond the short term. Compared to controls, hIgG1 knock-in mice mount minimal anti-human IgG responses, allowing for the persistence of therapeutically active circulating human IgG even in the late stages of treatment in chronic models of immune thrombocytopenic purpura and metastatic melanoma.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Inmunoglobulina G/inmunología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/toxicidad , Formación de Anticuerpos/genética , Enfermedad Crónica , Humanos , Tolerancia Inmunológica , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Ratones , Ratones Transgénicos , Modelos Animales , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/terapia
2.
Toxicol Pathol ; 50(8): 910-919, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36329562

RESUMEN

Removal of the core fucose from the Fc region of humanized monoclonal antibodies (afucosylated antibodies) enhances their antibody-dependent cell cytotoxicity activities in killing cancer cells. Based on the authors' experience and literature, administrations of afucosylated antibodies have been associated with neutropenia in cynomolgus monkeys. However, in a recent general toxicology study conducted with an afucosylated antibody in cynomolgus monkeys, transient neutropenia was observed and correlated with the emergence of anti-drug antibodies (ADAs) in the affected animals. To further explore the relationship between neutropenia, afucosylated antibodies, and ADAs in cynomolgus monkeys, we performed an investigational retrospective meta-analysis of data from general toxicology studies conducted with Genentech's therapeutic antibodies administered to cynomolgus monkeys between 2005 and 2021. In this analysis, transient neutropenia strongly correlated with ADA-induced inflammation in cynomolgus monkeys administered afucosylated antibodies. This may reflect the simultaneous occurrence of two distinct processes of neutrophil elimination and utilization, thus overwhelming bone marrow reserve capacity leading to transient neutropenia. The integrated analysis of immunogenicity, and anatomic and clinical pathology results from these studies highlights the correlation of transient neutropenia in cynomolgus monkeys with ADA-related inflammation, potentially exacerbated by enhanced effector function of afucosylated antibodies.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Neutropenia , Animales , Macaca fascicularis , Estudios Retrospectivos , Anticuerpos Monoclonales Humanizados/toxicidad , Neutropenia/inducido químicamente , Inflamación
3.
Andrologia ; 54(10): e14557, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36177829

RESUMEN

Pembrolizumab is a monoclonal antibody. Anastrozole is an infertility inhibitor of aromatase. Resveratrol is an antioxidant polyphenol in the reproductive system. This study was planned to demonstrate the protective effects of anastrozole and resveratrol against pembrolizumab-induced reproductive damage. Forty-two Sprague-Dawley rats were used in the study. Groups: The control, Pembrolizumab (PEMB), PEMB + Anastrazol (ANAST), PEMB + Resveratrol (RES), RES, and ANAST groups. At the end of the experiment, rats were euthanased under anaesthesia. Tissue samples were taken from rats for biochemical, histological, and ELISA evaluations. Tissues were subjected to routine tissue follow-up for histological analysis. Biochemically, thiobarbituric acid reactive substance (TBARS), glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) levels were measured. Sperm motility, abnormal sperm rate, and epididymal sperm concentration were examined spermatologically. Serum testosterone and programmed cell death-1 (PD-1) levels were measured using the ELISA. TBARS levels were significantly increased and GSH, SOD, GPx, and CAT levels were mitigated in PEMB-treated rats. Histologically; Control, ANAST, and RES groups testis samples were observed with normal histological appearance. Histological damage was detected in seminiferous tubule structures in testicular tissue in the PEMB group. In treatment groups, this damage was decreased. In addition, PD-1 and testosterone levels were evaluated by the ELISA method. ANAST and RES have therapeutic effects against reproductive damage caused by PEMB.


Asunto(s)
Antioxidantes , Inhibidores de la Aromatasa , Testículo , Anastrozol/farmacología , Animales , Anticuerpos Monoclonales Humanizados/toxicidad , Antioxidantes/farmacología , Aromatasa , Inhibidores de la Aromatasa/farmacología , Catalasa/farmacología , Glutatión , Glutatión Peroxidasa , Masculino , Polifenoles/farmacología , Receptor de Muerte Celular Programada 1/metabolismo , Ratas , Ratas Sprague-Dawley , Resveratrol/farmacología , Semen , Motilidad Espermática , Superóxido Dismutasa/metabolismo , Testosterona , Sustancias Reactivas al Ácido Tiobarbitúrico
4.
Toxicol Appl Pharmacol ; 422: 115557, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33932462

RESUMEN

CSL112 (apolipoprotein A-I, apo AI [human]) is an investigational drug in Phase 3 development for risk reduction of early recurrent cardiovascular events following an acute myocardial infarction (AMI). Although CSL112 is known to be well tolerated with a regimen of four weekly 6 g intravenous infusions after AMI, high doses of reconstituted apo AI preparations can transiently elevate liver enzymes in rats, raising the possibility of additive liver toxicity and toxicokinetic (TK) effects upon co-administration with cholesterol-lowering drugs, i.e., HMG-CoA reductase and proprotein convertase subtilisin/kexin type 9 inhibitors. We performed a toxicity and TK study in CD rats assigned to eleven treatment groups, including two dose levels of intravenous (IV) CSL112 (140 mg/kg, low-dose; 600 mg/kg, high-dose) administered as a single dose, alone or with intravenous alirocumab 50 mg/kg/week and/or oral atorvastatin 10 mg/kg/day. In addition, control groups of atorvastatin and alirocumab alone and in combination were investigated. Results showed some liver enzyme elevations (remaining <2-fold of baseline) related to administration of CSL112 alone. There was limited evidence of an additive effect of CSL112 on liver enzymes when combined, at either dose level, with alirocumab and/or atorvastatin, and histology revealed no evidence of an increased incidence or severity of hepatocyte vacuolation compared to the control treatments. Co-administration of the study drugs had minimal effect on their respective exposure levels, and on levels of total cholesterol and high-density lipoprotein cholesterol. These data support concomitant use of CSL112 with alirocumab and/or atorvastatin with no anticipated negative impact on liver safety and TK.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Anticolesterolemiantes/toxicidad , Atorvastatina/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Lipoproteínas HDL/toxicidad , Hígado/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticolesterolemiantes/farmacocinética , Atorvastatina/farmacocinética , Biomarcadores/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Colesterol/sangre , Interacciones Farmacológicas , Femenino , Lipoproteínas HDL/farmacocinética , Hígado/metabolismo , Hígado/patología , Masculino , Ratas Sprague-Dawley , Medición de Riesgo , Pruebas de Toxicidad , Toxicocinética
5.
BMC Cancer ; 21(1): 270, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33711962

RESUMEN

BACKGROUND: Epidermal growth factor receptor (EGFR) is a target for cancer therapy as it is overexpressed in a wide variety of cancers. Therapeutic antibodies that bind EGFR are being evaluated in clinical trials as imaging agents for positron emission tomography and image-guided surgery. However, some of these antibodies have safety concerns such as infusion reactions, limiting their use in imaging applications. Nimotuzumab is a therapeutic monoclonal antibody that is specific for EGFR and has been used as a therapy in a number of countries. METHODS: Formulation of IRDye800CW-nimotuzumab for a clinical trial application was prepared. The physical, chemical, and pharmaceutical properties were tested to develop the specifications to determine stability of the product. The acute and delayed toxicities were tested and IRDye800CW-nimotuzumab was determined to be non-toxic. Non-compartmental pharmacokinetics analysis was used to determine the half-life of IRDye800CW-nimotuzumab. RESULTS: IRDye800CW-nimotuzumab was determined to be non-toxic from the acute and delayed toxicity study. The half-life of IRDye800CW-nimotuzumab was determined to be 38 ± 1.5 h. A bi-exponential analysis was also used which gave a t1/2 alpha of 1.5 h and t1/2 beta of 40.8 h. CONCLUSIONS: Here, we show preclinical studies demonstrating that nimotuzumab conjugated to IRDye800CW is safe and does not exhibit toxicities commonly associated with EGFR targeting antibodies.


Asunto(s)
Drogas en Investigación/administración & dosificación , Inmunoconjugados/administración & dosificación , Neoplasias/diagnóstico por imagen , Imagen Óptica/métodos , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacocinética , Anticuerpos Monoclonales Humanizados/toxicidad , Bencenosulfonatos/administración & dosificación , Bencenosulfonatos/farmacocinética , Bencenosulfonatos/toxicidad , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Estabilidad de Medicamentos , Drogas en Investigación/farmacología , Drogas en Investigación/toxicidad , Receptores ErbB/antagonistas & inhibidores , Femenino , Semivida , Humanos , Inmunoconjugados/farmacocinética , Inmunoconjugados/toxicidad , Indoles/administración & dosificación , Indoles/farmacocinética , Indoles/toxicidad , Aplicación de Nuevas Drogas en Investigación , Masculino , Ratones , Neoplasias/patología , Neoplasias/cirugía , Cirugía Asistida por Computador/métodos , Pruebas de Toxicidad Aguda , Ensayos Antitumor por Modelo de Xenoinjerto
6.
South Med J ; 114(9): 614-619, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34480197

RESUMEN

The development of immune checkpoint inhibitors (ICIs) has changed the treatment paradigm for cancer. The ICIs nivolumab, pembrolizumab, and cemiplimab target programmed cell death protein 1, and durvalumab, avelumab, and atezolizumab target programmed death ligand 1. Ipilimumab targets cytotoxic T lymphocyte-associated antigen-4. Used as monotherapy or in combination, they have shown remarkable efficacy in melanoma, lung cancer, and many other solid tumors, and indications continue to evolve. These checkpoint inhibitors are typically well tolerated; however, they may cause immune-mediated adverse effects, resulting in inflammation of any organ system. Pulmonary toxicity is vital to recognize, and it can be more challenging to diagnose in patients with lung cancer, given the nature of the disease course and treatment.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/toxicidad , Pulmón/efectos de los fármacos , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/toxicidad , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/toxicidad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos
7.
Toxicol Appl Pharmacol ; 369: 1-16, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30822437

RESUMEN

Adrenomedullin (ADM) is a vasoactive peptide in sepsis. The non-neutralizing ADM-binding antibody Adrecizumab improved outcome in animal models of systemic inflammation and sepsis. Herein, we evaluated the preclinical safety of Adrecizumab in various animal species. First, Wistar rats received vehicle, 100, 200 or 400 mg/kg/day of Adrecizumab intravenously (n = 20 each) on days 1, 4, 8 and 14. An additional set of rats received vehicle or 400 mg/kg/day (n = 10 each) on the same days and were followed for 42 days. For toxicokinetics, satellite animals received vehicle (n = 6), 100, 200, or 400 mg/kg/day Adrecizumab intravenously (n = 18 each). A hemodynamic study was performed in Beagle dogs (n = 3) receiving vehicle (day 1), 2 mg/kg (day 3), 10 mg/kg (day 5), 50 mg/kg (day 8) and 10 mg/kg Adrecizumab intravenously (day 29). In final experiments, cynomolgus monkeys received vehicle, 25, 50 or 100 mg/kg/day Adrecizumab intravenously (n = 6 each) on days 1, 4, 8 and 14. Additional groups of monkeys received vehicle or 100 mg/kg/day Adrecizumab intravenously (n = 4 each) on the same days and were followed for 42 days. No mortality or moribund conditions occurred and no toxicologically relevant effects were attributed to Adrecizumab. Adrecizumab significantly increased circulating concentrations of its target peptide ADM, consistent with previous studies and mechanistically relevant. Toxicokinetic analyses showed immediate and dose-dependent peak concentrations, slow elimination and no gender differences. In conclusion, intravenous, repeated administration of high doses of Adrecizumab appeared well-tolerated across species. These results pave the way for further investigation of Adrecizumab in humans (intended dose of 2 mg/kg).


Asunto(s)
Adrenomedulina/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/toxicidad , Adrenomedulina/sangre , Adrenomedulina/inmunología , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacocinética , Perros , Femenino , Hemodinámica/efectos de los fármacos , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Ratas Wistar , Medición de Riesgo , Especificidad de la Especie , Toxicocinética
8.
Toxicol Appl Pharmacol ; 372: 57-69, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30914376

RESUMEN

Cytokine release syndrome (CRS) is a serious and potentially life-threatening complication typically associated with biological drug products. Pre-clinical testing in vitro and in vivo studies using non-human primates had failed to reliably predict CRS. To determine if bone marrow-thymus-liver (BLT) humanized mice with a fully engrafted human immune system or a CD34-humanized mouse model could predict CRS, we tested an anti-CD28 monoclonal antibody (mAb) similar to TGN1412. This TGN1412 analogue (TGN1412A) was initially tested in vitro and found to produce significant dose-dependent increases in cytokine production. For in vivo studies, adalimumab, an anti-tumor necrosis factor-alpha antibody known not to cause CRS, served as a negative control. We evaluated immune cell activation and cytokine expression in three independent experiments. In BLT humanized mice, significant increases in levels of human cytokines were identified in animals treated with anti-CD28 mAb. As expected, CD28+ cell detection was strongly reduced in the anti-CD28 treated group. Increased T cell activation was also observed. The control group did not show reductions in CD28+ T-cells and did not experience increased cytokine levels. Responses by CD34-humanized mice showed no significant differences between adalimumab and anti-CD28 treatment at doses used to test BLT-humanized mice. These results suggest that the TGN1412A produces similar results in vitro to the original TGN1412 monoclonal antibody. The BLT immune humanized mice but not the CD34 humanized mice produce both robust and specific cytokine responses and may represent a pre-clinical model to identify CRS.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Antígenos CD28/antagonistas & inhibidores , Síndrome de Liberación de Citoquinas/etiología , Citocinas/sangre , Linfocitos T/efectos de los fármacos , Animales , Antígenos CD34/inmunología , Antígenos CD28/sangre , Antígenos CD28/inmunología , Células Cultivadas , Síndrome de Liberación de Citoquinas/sangre , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Trasplante de Hígado , Ratones Endogámicos NOD , Ratones SCID , Medición de Riesgo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Timo/embriología , Timo/trasplante
9.
Acta Pharmacol Sin ; 40(7): 971-979, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30643210

RESUMEN

Emerging evidence demonstrates that a c-Met antibody-drug conjugate (ADC) has superior efficacy and safety profiles compared with those of currently available small molecules or antibody inhibitors for the treatment of c-Met-overexpressing cancers. Here we described both the in vitro and in vivo efficacies of SHR-A1403, a novel c-Met ADC composed of a humanized IgG2 monoclonal antibody against c-Met conjugated to a novel cytotoxic microtubule inhibitor. SHR-A1403 showed high affinity to c-Met proteins derived from human or monkey and potent inhibitory effects in cancer cell lines with high c-Met protein expression. In mice bearing tumors derived from cancer cell lines or patient HCC tissues with confirmed c-Met overexpression, SHR-A1403 showed excellent anti-tumor efficacy. Antibody binding with c-Met contributed to SHR-A1403 endocytosis; the subsequent translocation to lysosomes and cytotoxicity of the released toxin are speculated to be predominant mechanisms underlying the anti-tumor activity of SHR-A1403. In conclusion, SHR-A1403 showed significant anti-tumor activity in cancer cell lines, xenograft mouse models and an HCC PDX model, which all have high c-Met levels. These data provide references for SHR-A1403 as a potential therapy for the treatment of cancers with c-Met overexpression.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Moduladores de Tubulina/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/toxicidad , Antineoplásicos/inmunología , Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Inmunoconjugados/inmunología , Inmunoconjugados/toxicidad , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Microtúbulos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/inmunología , Moduladores de Tubulina/inmunología , Moduladores de Tubulina/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Regul Toxicol Pharmacol ; 108: 104476, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31536773

RESUMEN

Tildrakizumab (also known as MK-3222), is a high-affinity, humanized, immunoglobin G1κ monoclonal antibody targeting the p19 subunit of interleukin-23 recently approved for the treatment of moderate to severe plaque psoriasis in the US, Europe, and Australia. The safety profile of tildrakizumab was characterized in nonclinical studies using a pharmacologically relevant cynomolgus monkey model. In repeat-dose toxicity studies, cynomolgus monkeys were chronically treated with subcutaneous (SC) injections of 100 mg/kg of tildrakizumab every 2 weeks up to 9 months. Tildrakizumab was well tolerated, with no toxicological findings (including assessment of reproductive organs; hormonal effects; and cardiovascular, respiratory, and central nervous system function) at systemic exposures approximately 90 times higher than the recommended human dose of 100 mg. An embryofetal developmental study conducted in pregnant monkeys revealed no treatment-related effects to the developing fetus following SC administration of tildrakizumab 100 mg/kg. In a pre- and postnatal development study, 2 neonatal deaths due to potential viral infection at 100 mg/kg were considered of uncertain relationship to the treatment based on a lack of historical data on the occurrence of viral infection in neonate cynomolgus monkeys. The results of this comprehensive nonclinical safety program support the safe use of tildrakizumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Animales , Anticuerpos Monoclonales Humanizados/sangre , Anticuerpos Monoclonales Humanizados/farmacocinética , Desarrollo Embrionario/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Subunidad p19 de la Interleucina-23/sangre , Subunidad p19 de la Interleucina-23/inmunología , Macaca fascicularis , Masculino , Intercambio Materno-Fetal , Leche/química , Embarazo , Psoriasis/tratamiento farmacológico , Pruebas de Toxicidad Crónica
11.
Int J Toxicol ; 38(5): 395-404, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31364424

RESUMEN

Vedolizumab, a humanized monoclonal antibody approved for the treatment of adults with moderately to severely active ulcerative colitis or Crohn disease, targets α4ß7 integrin and selectively blocks gut-specific lymphocyte trafficking. The potential effects of vedolizumab on development were assessed by standard preclinical toxicity studies in rabbits and cynomolgus monkeys. A single infusion of vedolizumab (0, 10, 30, or 100 mg/kg) was administered intravenously to pregnant rabbits on gestational day 7; rabbits were monitored to gestational day 29. Vedolizumab (0, 10, or 100 mg/kg) was administered intravenously every 2 weeks to pregnant cynomolgus monkeys beginning on gestational day 20 with the last dose on gestational day 132 (9 doses total). In rabbits, vedolizumab did not affect maternal net body weight or net gains, gravid uterine weights, or mean maternal food consumption, nor did it affect intrauterine growth or fetal survival. There were also no vedolizumab effects on embryo-fetal development compared to controls. In cynomolgus monkeys, there was no increase in prenatal loss/death or stillbirth and no maternal toxicity associated with vedolizumab. On day 28 postpartum, low levels of vedolizumab were detected in the breast milk of 3 of 11 monkeys in the 100 mg/kg group. No vedolizumab-related effects on the number of infants born, infant development, or animal hematology or clinical chemistry were noted. Administration of vedolizumab to pregnant rabbits and cynomolgus monkeys did not show any potential for maternal or developmental effects.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Fármacos Gastrointestinales/toxicidad , Integrinas/antagonistas & inhibidores , Intercambio Materno-Fetal , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Peso Corporal/efectos de los fármacos , Femenino , Desarrollo Fetal/efectos de los fármacos , Fármacos Gastrointestinales/farmacocinética , Fuerza de la Mano , Integrinas/metabolismo , Macaca fascicularis , Masculino , Embarazo , Conejos
12.
Int J Toxicol ; 38(3): 173-182, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30852923

RESUMEN

Fulranumab, an anti-human nerve growth factor antibody, was evaluated in a series of nonclinical toxicology studies. No treatment effects were observed in adolescent cynomolgus monkeys in standard design, repeat-dose toxicology studies of up to 6 months. Adverse effects on the developing nervous system were observed in offspring of pregnant cynomolgus monkeys treated with fulranumab. Subsequent studies including detailed morphologic investigations of the nervous system did reveal fulranumab-related changes in adult cynomolgus monkeys; this article is focused on those findings. A single dose of ≥1 mg/kg fulranumab administered subcutaneously (SC) caused a decrease in neuron and sympathetic ganglion size (superior cervical ganglion), observed morphologically and stereologically, with a resulting appearance of increased glial cell density. Similar results were observed in repeat-dose (15 to 52 weeks) toxicity studies at ≤50 mg/kg/wk fulranumab SC. These effects recovered after a 3-month treatment-free period. Fulranumab did not cause any neuronal death, necrosis, apoptosis, or any apparent decrease in function of sympathetic neurons/ganglia at any time point examined. A no observed effect level (NOEL) was established at 0.25 mg/kg fulranumab SC every 4 weeks for 28 weeks.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Sistema Nervioso Central/efectos de los fármacos , Femenino , Macaca fascicularis , Masculino , Neuronas/efectos de los fármacos , Nivel sin Efectos Adversos Observados , Sistema Nervioso Periférico/efectos de los fármacos , Embarazo , Pruebas de Toxicidad Crónica
13.
Emerg Med J ; 36(5): 306-309, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30910911

RESUMEN

OBJECTIVES: We sought to estimate the prevalence of patients with cancer presenting to the emergency department (ED) who are undergoing treatment with immune checkpoint blockade (ICB) therapy; report their chief complaints; describe and estimate the prevalence of immune-related adverse events (IRAEs). METHODS: Four abstractors reviewed the medical records of patients with cancer treated with ICB who presented to an ED in Paris, France between January 2012 and June 2017. Chief complaints, underlying malignancy and ICB characteristics, and the final diagnoses according to the emergency physician were recorded. Abstractors noted if an emergency physician identified that a patient was receiving an ICB and if the emergency physician considered the possibility of an IRAE. The gold standard as to whether an IRAE was the cause was the patients' referring oncologist's opinion that the ED symptoms were attributed to ICB and IRAE according to post-ED medical records. Descriptive statistics were reported. RESULTS: Among the 409 patients treated with ICB at our institution, 139 presented to the ED. Chief complaints were fatigue (25.2%), fever (23%), vomiting (13.7%), diarrhoea (13.7%), dyspnoea (12.2%), abdominal pain (11.5%), confusion (8.6%) and headache (7.9%). Symptoms were due to IRAEs in 20 (14.4%) cases. The most frequent IRAEs were colitis (40%), endocrine toxicity (30%), hepatitis (25%) and pulmonary toxicity (5%). Patients with IRAEs compared with those without them more frequently had melanoma; had received more distinct courses of ICB treatment, an increased number of ICB medications and ICB cycles; and had a shorter time course since the last infusion of ICB. Emergency physicians considered the possibility of an IRAE in 24 (17.3%) of cases and diagnosed IRAE in 10 (50%) of those with later confirmed IRAE. IRAE was more likely to be missed when the referring oncologist was not contacted or when the patient had respiratory symptoms, fatigue or fever. CONCLUSIONS: ICB exposes patients to potentially severe IRAEs. Emergency physicians must identify patients treated with ICB and consider their toxicity when patients present to the ED with symptoms compatible with IRAEs.


Asunto(s)
Antineoplásicos Inmunológicos/toxicidad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Neoplasias/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales/toxicidad , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/toxicidad , Antineoplásicos Inmunológicos/uso terapéutico , Servicio de Urgencia en Hospital/organización & administración , Servicio de Urgencia en Hospital/estadística & datos numéricos , Fatiga/etiología , Femenino , Fiebre/etiología , Humanos , Ipilimumab/uso terapéutico , Ipilimumab/toxicidad , Masculino , Persona de Mediana Edad , Neoplasias/complicaciones , Nivolumab/uso terapéutico , Nivolumab/toxicidad , Paris/epidemiología , Prevalencia , Estudios Retrospectivos , Vómitos/etiología
14.
Toxicol Pathol ; 46(4): 408-420, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29768985

RESUMEN

Tanezumab, an anti-nerve growth factor (NGF) antibody, is in development for management of chronic pain. During clinical trials of anti-NGF antibodies, some patients reported unexpected adverse events requiring total joint replacements, resulting in a partial clinical hold on all NGF inhibitors. Three nonclinical toxicology studies were conducted to evaluate the effects of tanezumab or the murine precursor muMab911 on selected bone and joint endpoints and biomarkers in cynomolgus monkeys, Sprague-Dawley rats, and C57BL/6 mice. Joint and bone endpoints included histology, immunohistochemistry, microcomputed tomography (mCT) imaging, and serum biomarkers of bone physiology. Responses of bone endpoints to tanezumab were evaluated in monkeys at 4 to 30 mg/kg/week for 26 weeks and in rats at 0.2 to 10 mg/kg twice weekly for 28 days. The effects of muMab911 at 10 mg/kg/week for 12 weeks on selected bone endpoints were determined in mice. Tanezumab and muMab911 had no adverse effects on any bone or joint parameter. There were no test article-related effects on bone or joint histology, immunohistochemistry, or structure. Reversible, higher osteocalcin concentrations occurred only in the rat study. No deleterious effects were observed in joints or bones in monkeys, rats, or mice administered high doses of tanezumab or muMab911.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Huesos/efectos de los fármacos , Articulaciones/efectos de los fármacos , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/toxicidad , Macaca fascicularis , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Tomografía Computarizada por Rayos X
15.
Ann Rheum Dis ; 76(1): 295-302, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27381034

RESUMEN

OBJECTIVE: To investigate whether the effects of nerve growth factor (NGF) inhibition with tanezumab on rats with medial meniscal tear (MMT) effectively model rapidly progressive osteoarthritis (RPOA) observed in clinical trials. METHODS: Male Lewis rats underwent MMT surgery and were treated weekly with tanezumab (0.1, 1 or 10 mg/kg), isotype control or vehicle for 7, 14 or 28 days. Gait deficiency was measured to assess weight-bearing on the operated limb. Joint damage was assessed via histopathology. A second arm, delayed onset of treatment (starting 3-8 weeks after MMT surgery) was used to control for analgesia early in the disease process. A third arm, mid-tibial amputation, evaluated the dependency of the model on weight-bearing. RESULTS: Gait deficiency in untreated rats was present 3-7 days after MMT surgery, with a return to normal weight-bearing by days 14-28. Prophylactic treatment with tanezumab prevented gait deficiency and resulted in more severe cartilage damage. When onset of treatment with tanezumab was delayed to 3-8 weeks after MMT surgery, there was no increase in cartilage damage. Mid-tibial amputation completely prevented cartilage damage in untreated MMT rats. CONCLUSIONS: These data suggest that analgesia due to NGF inhibition during the acute injury phase is responsible for increased voluntary weight-bearing and subsequent cartilage damage in the rat MMT model. This model failed to replicate the hypotrophic bone response observed in tanezumab-treated patients with RPOA.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Cartílago Articular/lesiones , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Lesiones de Menisco Tibial/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/toxicidad , Artritis Experimental/inducido químicamente , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Marcha , Masculino , Meniscos Tibiales/diagnóstico por imagen , Meniscos Tibiales/patología , Radiografía , Ratas Endogámicas Lew , Lesiones de Menisco Tibial/diagnóstico por imagen , Lesiones de Menisco Tibial/patología , Lesiones de Menisco Tibial/fisiopatología , Soporte de Peso , Microtomografía por Rayos X
16.
Toxicol Pathol ; 45(5): 676-686, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28830332

RESUMEN

Obinutuzumab (GA101, Gazyva™, Gazyvaro®, F. Hoffmann-La Roche AG, Basel, Switzerland) is a humanized, glycoengineered type II antibody targeted against CD20. The preclinical safety evaluation required to support clinical development and marketing authorization of obinutuzumab included repeat-dose toxicity studies in cynomolgus monkeys for up to 6-month dosing with a 9-month recovery period. Results from those studies showed decreases in circulating B cells and corresponding B-cell depletion in lymphoid tissues, consistent with the desired pharmacology of obinutuzumab. Hypersensitivity reactions were noted at all doses in the 6-month study and were attributed to the foreign recognition of the drug construct in cynomolgus monkeys. Findings in monkeys were classified as acute hypersensitivity reactions that were evident immediately after dosing, such as excessive salivation, erythema, pruritus, irregular respiration, or ataxia, or chronic hypersensitivity reactions characterized by glomerulonephritis, arteritis/periarteritis, and inflammation in several tissues including serosal/adventitial inflammation. Immune complex deposits were demonstrated in tissues by immunohistochemistry, immunofluorescence, and electron microscopy. Some of, but not all, the animals that developed these reactions had detectable antidrug antibodies or circulating immune complexes accompanied by loss of drug exposure and pharmacodynamic effect. On the basis of clinical evidence to date, hypersensitivity reactions following obinutuzumab are rare, further supporting the general view that incidence and manifestation of immunogenicity in nonclinical species are generally not predictive for humans.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Hipersensibilidad a las Drogas , Macaca fascicularis , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/toxicidad , Antígenos CD20/análisis , Antígenos CD20/metabolismo , Linfocitos B/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Epidídimo/efectos de los fármacos , Femenino , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Masculino , Músculos/efectos de los fármacos , Pruebas de Toxicidad Crónica
17.
Int J Toxicol ; 36(6): 449-462, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29202623

RESUMEN

ANX005 is a humanized immunoglobulin G4 recombinant antibody against C1q that inhibits its function as the initiating molecule of the classical complement cascade. The safety and tolerability of ANX005 are currently being evaluated in a phase I trial in healthy volunteers ( www.clinicaltrials.gov Identifier: NCT03010046). Inhibition of C1q can be applied therapeutically in a broad spectrum of diseases, including acute antibody-mediated autoimmune disease, such as Guillain-Barré syndrome (GBS), and in chronic diseases of the central nervous system involving complement-mediated neurodegeneration, such as Alzheimer's disease (AD). To support the clinical development of ANX005, several studies were conducted to assess the pharmacology, pharmacokinetics, and potential toxicity of ANX005. ANX-M1, the murine precursor of ANX005, functionally inhibits the classical complement cascade both in vitro and in vivo, to protect against disease pathology in mouse models of GBS and AD. Toxicology studies with ANX005, itself, showed that intravenous administration once weekly for 4 weeks was well tolerated in rats and monkeys, with no treatment-related adverse findings. Serum levels of ANX005 in monkeys correlate with a reduction in free C1q levels both in the serum and in the cerebrospinal fluid. In summary, ANX005 has shown proof of concept in in vitro and in vivo nonclinical pharmacology models, with no toxicity in the 4-week repeat-dose studies in rats and monkeys. The no observed adverse effect level was 200 mg/kg/dose, which is 200-fold higher than the first-in-human starting dose of 1 mg/kg in healthy volunteers.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Enfermedades Autoinmunes/tratamiento farmacológico , Complemento C1q/inmunología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Enfermedades Autoinmunes/inmunología , Complemento C1q/metabolismo , Vía Clásica del Complemento/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Enfermedades Neurodegenerativas/inmunología , Ratas Sprague-Dawley , Especificidad de la Especie
18.
Biol Blood Marrow Transplant ; 22(9): 1608-1614, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27220263

RESUMEN

Mogamulizumab (MOG), a humanized anti-CC chemokine receptor 4 (CCR4) monoclonal antibody, has recently played an important role in the treatment of adult T cell leukemia/lymphoma (ATLL). Because CCR4 is expressed on normal regulatory T cells as well as on ATLL cells, MOG may accelerate graft-versus-host disease (GVHD) by eradicating regulatory T cells in patients with allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, there is limited information about its safety and efficacy in patients treated with MOG before allo-HSCT. In the present study, 25 patients with ATLL were treated with MOG before allo-HSCT, after which 18 patients (72%) achieved remission. The overall survival and progression-free survival at 1 year post-transplantation were 20.2% (95% CI, 6.0% to 40.3%) and 15.0% (95% CI, 4.3% to 32.0%), respectively. The cumulative incidence of acute GVHD was 64.0% (95% CI, 40.7% to 80.1%) for grade II-IV and 34.7% (95% CI, 15.8% to 54.4%) for grade III-IV. The cumulative incidence of transplantation-related mortality (TRM) was 49.0% (95% CI, 27.0% to 67.8%). Six of 7 patients with acute GVHD grade III-IV died from GVHD, which was the leading cause of death. In particular, a shorter interval from the last administration of MOG to allo-HSCT was associated with more severe GVHD. MOG use before allo-HSCT may decrease the ATLL burden; however, it is associated with an increase in TRM due to severe GVHD. Because MOG is a potent anti-ATLL agent, new treatment protocols should be developed to integrate MOG at suitable doses and timing of administration to minimize unwanted GVHD development.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Enfermedad Injerto contra Huésped/inducido químicamente , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Leucemia-Linfoma de Células T del Adulto/complicaciones , Leucemia-Linfoma de Células T del Adulto/tratamiento farmacológico , Enfermedad Aguda , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/mortalidad , Humanos , Leucemia-Linfoma de Células T del Adulto/mortalidad , Leucemia-Linfoma de Células T del Adulto/terapia , Masculino , Persona de Mediana Edad , Inducción de Remisión/métodos , Estudios Retrospectivos , Análisis de Supervivencia , Trasplante Homólogo
19.
Birth Defects Res B Dev Reprod Toxicol ; 107(4-5): 174-184, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27519584

RESUMEN

A humanized monoclonal antibody targeting transforming growth factor ß1 (TGF-ß1 mab) has been used in development for the treatment of chronic kidney disease. Embryo-fetal development studies were conducted in rats and rabbits using 30 and 25 animals per group, respectively. The TGF-ß1 mab was administered subcutaneously to rats at 0, 2, or 50 mg/kg/dose on gestation days (GDs) 6, 10, and 14 and intravenously to rabbits at 0 or 3 mg/kg/dose on GDs 7, 12 to 19, and at 30 mg/kg/dose on GDs 7, 12, 14, 16, and 18. Maternal reproductive endpoints and fetal viability, weight, and morphology were evaluated. There was no indication of maternal or embryo-fetal toxicity in the rat. Effects in the rabbit were limited to the fetus where the 30 mg/kg TGF-ß1 mab dose produced a slight decrease in fetal weight and an increase in the incidence of retrocaval ureter and an absent and/or malpositioned kidney/ureter in two fetuses. In conclusion, TGF-ß1 mab produced no adverse maternal or embryo-fetal findings in rats when administered ≤50 mg/kg on GDs 6, 10, and 14. TGF-ß1 mab did not demonstrate maternal toxicity or embryo-fetal lethality at doses as high as 30 mg/kg when administered on GDs 7, 12, 14, 16, and 18 in rabbits. Fetal growth and morphology were affected only at 30 mg/kg; thus, the no observed adverse effect level was 3 mg/kg in rabbits. The margin of safety for both rats and rabbits was ≥37-fold the clinical exposure level.


Asunto(s)
Anticuerpos Monoclonales Humanizados/toxicidad , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Fetal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Feto/efectos de los fármacos , Masculino , Nivel sin Efectos Adversos Observados , Conejos , Ratas , Ratas Sprague-Dawley , Reproducción/efectos de los fármacos , Pruebas de Toxicidad , Toxicocinética
20.
Retina ; 35(5): 1007-15, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25627091

RESUMEN

PURPOSE: To determine the permeability of bevacizumab and ranibizumab through highly-polarized retinal pigment epithelial (RPE) cells in vitro. METHODS: Highly-polarized RPE cells were cultured in the upper chamber of a Transwell culture system. Bevacizumab or ranibizumab was added to the upper chamber. After 3 hours, the concentrations of bevacizumab or ranibizumab were determined in the upper and lower chambers. The cytotoxicities of the two anti-vascular endothelial growth factor agents were determined histologically. The effects of inhibiting endocytosis by pharmacologic inhibitors were also evaluated. RESULTS: The concentration of ranibizumab was higher than that of bevacizumab in the lower chamber (P < 0.05). Vascular endothelial growth factor was found mainly in the lower chamber under normal conditions. However, the concentration of vascular endothelial growth factor in the lower chamber was significantly less when ranibizumab was added to the upper chamber than when bevacizumab was added. Histology showed no obvious changes in bevacizumab-exposed or ranibizumab-exposed RPE cells. Pretreatment with protein kinase C inhibitor staurosporine had significant negative effects on the permeability of bevacizumab and ranibizumab (P < 0.05). CONCLUSION: Ranibizumab is more permeable than bevacizumab through the highly-polarized RPE layer at clinically equivalent concentrations, and their permeability was partially protein kinase C-dependent. Ranibizumab might be more therapeutically effective than bevacizumab on choroidal neovascularization beneath the RPE layer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacocinética , Epitelio Pigmentado de la Retina/metabolismo , Inhibidores de la Angiogénesis/toxicidad , Animales , Anticuerpos Monoclonales Humanizados/toxicidad , Bevacizumab , Células Cultivadas , Impedancia Eléctrica , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Etiquetado Corte-Fin in Situ , Microscopía Electrónica de Transmisión , Permeabilidad , Ranibizumab , Epitelio Pigmentado de la Retina/efectos de los fármacos , Estaurosporina/farmacología , Porcinos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteína de la Zonula Occludens-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA