Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 814
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Regul Toxicol Pharmacol ; 138: 105329, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36592682

RESUMO

To support registration of monoclonal antibodies (mAbs) for chronic indications, 6-month toxicity studies have historically been conducted. Experience with mAb development has shown a relatively benign and well-understood safety profile for this class, with most toxicity findings anticipated based on pharmacology. We evaluated whether a 6-month toxicity study is necessary to assess the long-term safety of mAbs. Data on First-in-Human (FIH)-enabling and chronic toxicity studies were shared for 142 mAbs submitted by 11 companies. Opportunities to further optimize study designs to reduce animal usage were identified. For 71% of mAbs, no toxicities or no new toxicities were noted in chronic studies compared to FIH-enabling study findings. New toxicities of potential concern for human safety or that changed trial design were identified in 13.5% of cases, with 7% being considered critical and 2% leading to program termination. An iterative, weight-of-evidence model which considers factors that influence the overall risk for a mAb to cause toxicity was developed. This model enables an evidence-based justification, suggesting when 3-month toxicity studies are likely sufficient to support late-stage clinical development and registration for some mAbs.


Assuntos
Anticorpos Monoclonais , Projetos de Pesquisa , Animais , Humanos , Anticorpos Monoclonais/toxicidade
2.
Proc Natl Acad Sci U S A ; 117(11): 6086-6091, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32123080

RESUMO

Recombinant immunotoxins (RITs) are chimeric proteins composed of an Fv and a protein toxin being developed for cancer treatment. The Fv brings the toxin to the cancer cell, but most of the RITs do not reach the tumor and are removed by other organs. To identify cells responsible for RIT removal, and the pathway by which RITs reach these cells, we studied SS1P, a 63-kDa RIT that targets mesothelin-expressing tumors and has a short serum half-life. The major organs that remove RIT were identified by live mouse imaging of RIT labeled with FNIR-Z-759. Cells responsible for SS1P removal were identified by immunohistochemistry and intravital two-photon microscopy of kidneys of rats. The primary organ of SS1P removal is kidney followed by liver. In the kidney, SS1P passes through the glomerulus, is taken up by proximal tubular cells, and transferred to lysosomes. In the liver, macrophages are involved in removal. The short half-life of SS1P is due to its very rapid filtration by the kidney followed by degradation in proximal tubular cells of the kidney. In mice treated with SS1P, proximal tubular cells are damaged and albumin in the urine is increased. SS1P uptake by kidney is reduced by coadministration of l-lysine. Our data suggests that l-lysine administration to humans might prevent SS1P-mediated kidney damage, reduce albumin loss in urine, and alleviate capillary leak syndrome.


Assuntos
Albuminúria/patologia , Anticorpos Monoclonais/farmacocinética , Síndrome de Vazamento Capilar/patologia , Imunotoxinas/farmacocinética , Túbulos Renais Proximais/efeitos dos fármacos , Albuminúria/induzido quimicamente , Albuminúria/prevenção & controle , Albuminúria/urina , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Anticorpos Monoclonais/toxicidade , Síndrome de Vazamento Capilar/induzido quimicamente , Síndrome de Vazamento Capilar/prevenção & controle , Síndrome de Vazamento Capilar/urina , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes/química , Meia-Vida , Humanos , Imunotoxinas/administração & dosagem , Imunotoxinas/química , Imunotoxinas/toxicidade , Microscopia Intravital , Glomérulos Renais/metabolismo , Túbulos Renais Proximais/diagnóstico por imagem , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Lisina/administração & dosagem , Mesotelina , Camundongos , Microscopia de Fluorescência , Neoplasias/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/toxicidade , Eliminação Renal/efeitos dos fármacos , Albumina Sérica/análise , Albumina Sérica/metabolismo , Coloração e Rotulagem
3.
Blood ; 135(25): 2292-2301, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32157300

RESUMO

Immune thrombocytopenia (ITP) is an acquired bleeding disorder characterized by antibody-mediated platelet destruction. Different mechanisms have been suggested to explain accelerated platelet clearance and impaired thrombopoiesis, but the pathophysiology of ITP has yet to be fully delineated. In this study, we tested 2 mouse models of immune-mediated thrombocytopenia using the rat anti-mouse GPIbα monoclonal antibody 5A7, generated in our laboratory. After a single IV administration of high-dose (2 mg/kg) 5A7, opsonized platelets were rapidly cleared from the circulation into the spleen and liver; this was associated with rapid upregulation of thrombopoietin (TPO) messenger RNA. In contrast, subcutaneous administration of low-dose 5A7 (0.08-0.16 mg/kg) every 3 days gradually lowered the platelet count; in this case, opsonized platelets were observed only in the spleen, and TPO levels remained unaltered. Interestingly, in both models, the 5A7 antibody was found on the surface of, as well as internalized to, bone marrow megakaryocytes. Consequently, platelets generated in the chronic phase of repeated subcutaneous 5A7 administration model showed reduced GPIbα membrane expression on their surface. Our findings indicate that evaluation of platelet surface GPIbα relative to platelet size may be a useful marker to support the diagnosis of anti-GPIbα antibody-induced ITP.


Assuntos
Anticorpos Monoclonais/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Púrpura Trombocitopênica Idiopática/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/toxicidade , Reações Antígeno-Anticorpo , Plaquetas/imunologia , Modelos Animais de Doenças , Injeções Intravenosas , Injeções Subcutâneas , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Opsonizantes/imunologia , Agregação Plaquetária/imunologia , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Púrpura Trombocitopênica Idiopática/etiologia , RNA Mensageiro/biossíntese , Ratos , Baço/patologia , Trombopoetina/biossíntese , Trombopoetina/genética , Regulação para Cima
4.
Int J Toxicol ; 41(5): 389-401, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35672934

RESUMO

Bococizumab is an anti-PCSK9 monoclonal antibody that was intended for the treatment of hypercholesterolemia. After reviewing the 6-month rat toxicity study data, in which there was a low spontaneous tumor incidence, unrelated to bococizumab administration, the U.S. FDA granted a carcinogenicity waiver request based on a weight-of-evidence assessment of low carcinogenic risk. Subsequently, after reviewing 6-month rat toxicity study data from another anti-PCSK9 antibody, RN317, with a similar low tumor incidence (unrelated to RN317), the U.S. FDA rescinded the bococizumab carcinogenicity study waiver and requested a full 2-year rat carcinogenicity study be conducted. The resulting 2-year carcinogenicity study demonstrated no bococizumab-related increase in tumors, confirming the weight-of-evidence evaluation and alleviating concerns regarding the carcinogenic potential. Here we report the scientific and regulatory background that led to the request for a rat carcinogenicity study, the feedback on the design of the carcinogenicity study, and the results from this study which affirmed the original weight-of-evidence assessment of low carcinogenic risk.


Assuntos
Carcinógenos , Hipercolesterolemia , Animais , Anticorpos Monoclonais/toxicidade , Testes de Carcinogenicidade , Carcinógenos/toxicidade , LDL-Colesterol , Pró-Proteína Convertase 9 , Ratos
5.
J Intern Med ; 290(1): 141-156, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33342002

RESUMO

BACKGROUND: Phosphorylcholine (PC) is an important pro-inflammatory damage-associated molecular pattern. Previous data have shown that natural IgM anti-PC protects against cardiovascular disease. We aimed to develop a monoclonal PC IgG antibody with anti-inflammatory and anti-atherosclerotic properties. METHODS: Using various techniques PC antibodies were validated and optimized. In vivo testing was performed in a femoral artery cuff model in ApoE3*Leiden mice. Safety studies are performed in rats and cynomolgus monkeys. RESULTS: A chimeric anti-PC (PC-mAb(T15), consisting of a human IgG1 Fc and a mouse T15/E06 Fab) was produced, and this was shown to bind specifically to epitopes in human atherosclerotic tissues. The cuff model results in rapid induction of inflammatory genes and altered expression of genes associated with ER stress and choline metabolism in the lesions. Treatment with PC-mAb(T15) reduced accelerated atherosclerosis via reduced expression of endoplasmic reticulum stress markers and CCL2 production. Recombinant anti-PC Fab fragments were identified by phage display and cloned into fully human IgG1 backbones creating a human monoclonal IgG1 anti-PC (PC-mAbs) that specifically bind PC, apoptotic cells and oxLDL. Based on preventing macrophage oxLDL uptake and CCL2 production, four monoclonal PC-mAbs were selected, which to various extent reduced vascular inflammation and lesion development. Additional optimization and validation of two PC-mAb antibodies resulted in selection of PC-mAb X19-A05, which inhibited accelerated atherosclerosis. Clinical grade production of this antibody (ATH3G10) significantly attenuated vascular inflammation and accelerated atherosclerosis and was tolerated in safety studies in rats and cynomolgus monkeys. CONCLUSIONS: Chimeric anti-PCs can prevent accelerated atherosclerosis by inhibiting vascular inflammation directly and through reduced macrophage oxLDL uptake resulting in decreased lesions. PC-mAb represents a novel strategy for cardiovascular disease prevention.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/terapia , Imunoglobulina G/imunologia , Fosforilcolina/imunologia , Animais , Anticorpos Monoclonais/toxicidade , Aterosclerose/prevenção & controle , Quimera , LDL-Colesterol/antagonistas & inibidores , LDL-Colesterol/metabolismo , Colina/metabolismo , Modelos Animais de Doenças , Feminino , Macaca fascicularis , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Oxirredução , Ratos
6.
South Med J ; 114(9): 614-619, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34480197

RESUMO

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.


Assuntos
Inibidores de Checkpoint Imunológico/toxicidade , Pulmão/efeitos dos fármacos , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos
7.
Turk J Med Sci ; 50(4): 1111-1122, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32151118

RESUMO

Background/aim: To investigate the effect of intravitreal golimumab on rabbit retina histopathology. Materials and methods: Sixteen albino New Zealand rabbits were divided into three groups. The right eye of each rabbit in groups I, II, and III received a single intravitreal injection of 5 mg/0.05 mL (6 eyes), 10 mg/0.1 mL (6 eyes), or 20 mg/0.2 mL (4 eyes) golimumab, while left eyes served as controls with the same volume of a balanced salt solution injection. All animals were examined using slit-lamp biomicroscopy and indirect ophthalmoscopy before and after intravitreal injection and at days 1 and 7. Animals were euthanized on day 7 and the eyes were enucleated for immunohistochemistry evaluation and electron microscopic examination of the retinas. Results: For groups I, II, and III, the number of cells in the outer nuclear layer and the inner nuclear layer was decreased compared to those in the control groups. In group I, the percentage of caspase-3 staining of the outer nuclear layer was significantly higher than that in the control. For groups II and III, TUNEL and caspase-3 staining percentages in the outer and inner nuclear layers were found to be significantly higher than those for the control groups. In the ganglion cell layer, for groups I, II, and III, neither TUNEL nor caspase-3 staining percentages showed any significant difference between two groups. No significant dose-dependent relationship was found for increasing doses of golimumab in all layers. Myelin figures and karyorrhexis in the photoreceptor cells were prominent in electron microscopy of the golimumab-injected eyes. Conclusion: Golimumab caused apoptosis in both photoreceptors and bipolar cells of the rabbit retina. Potential retinal toxicity of intravitreal golimumab should be considered if an intravitreal administration is planned.


Assuntos
Anticorpos Monoclonais/toxicidade , Neuropatia Óptica Tóxica/etiologia , Animais , Anticorpos Monoclonais/administração & dosagem , Apoptose , Modelos Animais de Doenças , Injeções Intravítreas , Oftalmoscopia , Coelhos , Lâmpada de Fenda
8.
Pharm Res ; 36(12): 169, 2019 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-31654236

RESUMO

PURPOSE: The purpose of this study was to validate a ligand binding assay for the quantitation of a monoclonal antibody-based biotherapeutics (PF-57781346) in samples collected via capillary microsampling to support a regulated mouse toxicity study. METHOD: A quantitative ligand binding assay on the Gyrolab platform was developed to quantify PF-57781346 in blood samples derived from capillary mouse serial sampling. The method validation evaluated assay characteristics including accuracy and precision, influence of sample processing on drug quantitation, whole blood matrix selectivity, dilution linearity and the stability of the drug in the study sample matrix. RESULTS: The method validation demonstrated acceptable analytical characteristics. The whole blood selectivity testing demonstrated accuracy between -4.8% and 13.9% in 10 out of 10 individual whole blood samples, suggesting that drug quantitation from whole blood is not impacted by the serial sampling procedure. Short-term and long-term drug stability in study sample matrix were established to cover required stability for sample storage and analysis (accuracy between -7.3% and 6.1%). CONCLUSION: We reported a successful validation of a bioanalytical method that quantifies PF-55781346 in samples collected via capillary microsampling. The experience shared in this study could serve as a model process for bioanalytical method validation when capillary microsampling is used.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Imunoensaio/métodos , Animais , Coleta de Amostras Sanguíneas/métodos , Estabilidade de Medicamentos , Ligantes , Camundongos , Modelos Animais , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Toxicocinética
9.
Nature ; 501(7465): 102-6, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23903654

RESUMO

Prion infections cause lethal neurodegeneration. This process requires the cellular prion protein (PrP(C); ref. 1), which contains a globular domain hinged to a long amino-proximal flexible tail. Here we describe rapid neurotoxicity in mice and cerebellar organotypic cultured slices exposed to ligands targeting the α1 and α3 helices of the PrP(C) globular domain. Ligands included seven distinct monoclonal antibodies, monovalent Fab1 fragments and recombinant single-chain variable fragment miniantibodies. Similar to prion infections, the toxicity of globular domain ligands required neuronal PrP(C), was exacerbated by PrP(C) overexpression, was associated with calpain activation and was antagonized by calpain inhibitors. Neurodegeneration was accompanied by a burst of reactive oxygen species, and was suppressed by antioxidants. Furthermore, genetic ablation of the superoxide-producing enzyme NOX2 (also known as CYBB) protected mice from globular domain ligand toxicity. We also found that neurotoxicity was prevented by deletions of the octapeptide repeats within the flexible tail. These deletions did not appreciably compromise globular domain antibody binding, suggesting that the flexible tail is required to transmit toxic signals that originate from the globular domain and trigger oxidative stress and calpain activation. Supporting this view, various octapeptide ligands were not only innocuous to both cerebellar organotypic cultured slices and mice, but also prevented the toxicity of globular domain ligands while not interfering with their binding. We conclude that PrP(C) consists of two functionally distinct modules, with the globular domain and the flexible tail exerting regulatory and executive functions, respectively. Octapeptide ligands also prolonged the life of mice expressing the toxic PrP(C) mutant, PrP(Δ94-134), indicating that the flexible tail mediates toxicity in two distinct PrP(C)-related conditions. Flexible tail-mediated toxicity may conceivably play a role in further prion pathologies, such as familial Creutzfeldt-Jakob disease in humans bearing supernumerary octapeptides.


Assuntos
Anticorpos/imunologia , Anticorpos/toxicidade , Maleabilidade , Príons/química , Príons/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/toxicidade , Sítios de Ligação de Anticorpos , Calpaína/metabolismo , Cerebelo , Síndrome de Creutzfeldt-Jakob/metabolismo , Reagentes de Ligações Cruzadas , Mapeamento de Epitopos , Feminino , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/toxicidade , Técnicas In Vitro , Ligantes , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo , Proteínas PrPC/química , Proteínas PrPC/genética , Proteínas PrPC/imunologia , Príons/genética , Espécies Reativas de Oxigênio/metabolismo , Deleção de Sequência/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/toxicidade
11.
J Oncol Pharm Pract ; 25(1): 187-191, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29199505

RESUMO

Monoclonal antibodies have expanded as a novel class of therapeutic agents. In contrast to appropriate guidelines for safe handling of cytotoxics, there are no real standards for the safe handling of monoclonal antibodies. Many questions have arisen whether monoclonal antibodies have to be prepared under controlled circumstances or can be prepared on the ward. We developed a flowchart which provides recommendations for the classification of monoclonal antibodies according to their toxicity profile and takes practical and financial issues into account. It allows oncology pharmacists worldwide to define which monoclonal antibodies can/must be prepared in pharmacy aseptic facilities and which monoclonal antibodies can be prepared on the ward.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/toxicidade , Composição de Medicamentos/normas , Farmacêuticos/normas , Design de Software , Composição de Medicamentos/métodos , Humanos , Exposição Ocupacional/prevenção & controle , Exposição Ocupacional/normas , Alocação de Recursos , Medição de Risco
12.
Int J Toxicol ; 38(4): 319-325, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31220983

RESUMO

The pharmaceutical and biotechnology industries continually review the requirements for, and relevance of, safety assessment strategies. Various industry consortia are currently discussing and reviewing data on a range of topics with respect to regulatory toxicology programs. These consortia are charged with critical evaluation of data and the identification of opportunities to promote best practice and to introduce improved approaches to safety assessment. Such improvements may include enhanced predictivity, more efficient ways of working, and opportunities for promoting and implementing the 3Rs (replacement, refinement, or reduction). As each consortium is considering a distinct question, individual outputs and recommendations could be perceived to be conflicting. However, a common theme embraced by the consortia represented here is exploration of the most appropriate use of animals for the safety assessment of new medicinal products. This short review summarizes presentations and discussions from a symposium describing the work of four industry consortia and considers whether their recommendations can be aligned into realistic approaches to improve future toxicology testing strategies, highlighting justification for the appropriate use of different animal species and opportunities for reductions in animal use without compromising patient safety.


Assuntos
Alternativas aos Testes com Animais , Desenvolvimento de Medicamentos , Animais , Anticorpos Monoclonais/toxicidade , Bases de Dados Factuais , Humanos , Medição de Risco , Testes de Toxicidade
13.
Emerg Med J ; 36(5): 306-309, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30910911

RESUMO

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.


Assuntos
Antineoplásicos Imunológicos/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Neoplasias/tratamento farmacológico , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/toxicidade , Antineoplásicos Imunológicos/uso terapêutico , Serviço Hospitalar de Emergência/organização & administração , Serviço Hospitalar de Emergência/estatística & dados numéricos , Fadiga/etiologia , Feminino , Febre/etiologia , Humanos , Ipilimumab/uso terapêutico , Ipilimumab/toxicidade , Masculino , Pessoa de Meia-Idade , Neoplasias/complicações , Nivolumabe/uso terapêutico , Nivolumabe/toxicidade , Paris/epidemiologia , Prevalência , Estudos Retrospectivos , Vômito/etiologia
14.
PLoS Pathog ; 12(1): e1005401, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26821311

RESUMO

Antibodies against the prion protein PrPC can antagonize prion replication and neuroinvasion, and therefore hold promise as possible therapeutics against prion diseases. However, the safety profile of such antibodies is controversial. It was originally reported that the monoclonal antibody D13 exhibits strong target-related toxicity, yet a subsequent study contradicted these findings. We have reported that several antibodies against certain epitopes of PrPC, including antibody POM1, are profoundly neurotoxic, yet antibody ICSM18, with an epitope that overlaps with POM1, was reported to be innocuous when injected into mouse brains. In order to clarify this confusing situation, we assessed the neurotoxicity of antibodies D13 and ICSM18 with dose-escalation studies using diffusion-weighted magnetic resonance imaging and various histological techniques. We report that both D13 and ICSM18 induce rapid, dose-dependent, on-target neurotoxicity. We conclude that antibodies directed to this region may not be suitable as therapeutics. No such toxicity was found when antibodies against the flexible tail of PrPC were administered. Any attempt at immunotherapy or immunoprophylaxis of prion diseases should account for these potential untoward effects.


Assuntos
Anticorpos Monoclonais/toxicidade , Imunoterapia/métodos , Proteínas PrPC/imunologia , Doenças Priônicas/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epitopos de Linfócito B/imunologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Priônicas/patologia
15.
Toxicol Appl Pharmacol ; 355: 147-155, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30008375

RESUMO

Fibroblast Growth Factors (FGFs) and their receptors (FGFRs) have been proposed as potential drug targets for the treatment of obesity. The aim of this study was to assess the potential toxicity in rats of three anti-FGFR1c mAbs with differential binding activity prior to clinical development. Groups of male rats received weekly injections of either one of two FGFR1c-specific mAbs or an FGFR1c/FGFR4-specific mAb at 10 mg/kg for up to 4 weeks. All three mAbs caused significant reductions in food intake and weight loss leading to some animals being euthanized early for welfare reasons. In all three groups given these mAbs, microscopic changes were seen in the bones and heart valves. In the bones of the femoro-tibial joint, thickening of the diaphyseal cortex of long bones, due to deposition of well organized new lamellar bone, indicated that an osteogenic effect was observed. In the heart, valvulopathy described as an endocardial myxomatous change affecting the mitral, pulmonary, tricuspid and aortic valves was observed in all mAb-treated animals. The presence of FGFR1 mRNA expression in the heart valves was confirmed using in situ hybridization. Targeting the FGF-FGFR1c pathway with anti-FGFR1c mAbs leads to drug induced valvulopathy in rats. In effect, this precluded the development of these mAbs as potential anti-obesity drugs. The valvulopathy observed was similar to that described for fenfluramine and dexafenfluramine. The pathogenesis of the drug-induced valvulopathy is considered FGFR1c-mediated, based on the specificity of the mAbs and FGFR1 mRNA expression in the heart valves.


Assuntos
Fármacos Antiobesidade/toxicidade , Anticorpos Monoclonais/toxicidade , Doenças das Valvas Cardíacas/induzido quimicamente , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Animais , Fármacos Antiobesidade/farmacocinética , Anticorpos Monoclonais/farmacocinética , Osso e Ossos/patologia , Ingestão de Alimentos/efeitos dos fármacos , Doenças das Valvas Cardíacas/metabolismo , Doenças das Valvas Cardíacas/patologia , Valvas Cardíacas/metabolismo , Valvas Cardíacas/patologia , Masculino , Osteogênese/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Redução de Peso/efeitos dos fármacos
16.
Pharm Res ; 35(11): 222, 2018 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-30280329

RESUMO

PURPOSE: Biotherapeutics can be susceptible to oxidation during manufacturing and storage. Free L-methionine is known to protect methionine residues in proteins from oxidation. Similarly, free tryptophan and other indole derivatives have been shown to protect tryptophan residues from oxidation. N-acetyl-DL-tryptophan was previously identified as a potentially superior antioxidant to tryptophan as it has a lower oxidation potential and produces less peroxide upon light exposure. This study sought to confirm the antioxidant efficacy and safety of N-acetyl-DL-tryptophan and L-methionine as formulation components for biotherapeutic drugs. METHODS: Antibodies were subjected to AAPH and light exposure in the presence of N-acetyl-DL-tryptophan and L-methionine. Oxidation in relevant CDR and Fc residues was quantified by peptide map. In silico, in vitro, and in vivo studies were performed to evaluate the safety of N-acetyl-DL-tryptophan and L-methionine. RESULTS: Peptide mapping demonstrated that N-acetyl-DL-tryptophan was effective at protecting tryptophans from AAPH stress, and that the combination of N-acetyl-DL-tryptophan and L-methionine protected both tryptophan and methionine from AAPH stress. The safety assessment suggested an acceptable safety profile for both excipients. CONCLUSIONS: N-acetyl-tryptophan and L-methionine effectively reduce the oxidation of susceptible tryptophan and methionine residues in antibodies and are safe for use in parenteral biotherapeutic formulations.


Assuntos
Anticorpos Monoclonais/química , Antioxidantes/química , Metionina/química , Triptofano/análogos & derivados , Amidinas/química , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/toxicidade , Antioxidantes/administração & dosagem , Antioxidantes/toxicidade , Produtos Biológicos , Linhagem Celular Tumoral , Simulação por Computador , Composição de Medicamentos , Excipientes/química , Feminino , Humanos , Macaca fascicularis , Masculino , Metionina/administração & dosagem , Metionina/toxicidade , Testes de Mutagenicidade , Oxirredução , Conformação Proteica , Estabilidade Proteica , Coelhos , Triptofano/administração & dosagem , Triptofano/química , Triptofano/toxicidade , Raios Ultravioleta
17.
Arterioscler Thromb Vasc Biol ; 37(11): 2171-2181, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28912363

RESUMO

OBJECTIVE: Current experimental models of abdominal aortic aneurysm (AAA) do not accurately reproduce the major features of human AAA. We hypothesized that blockade of TGFß (transforming growth factor-ß) activity-a guardian of vascular integrity and immune homeostasis-would impair vascular healing in models of nondissecting AAA and would lead to sustained aneurysmal growth until rupture. APPROACH AND RESULTS: Here, we test this hypothesis in the elastase-induced AAA model in mice. We analyze AAA development and progression using ultrasound in vivo, synchrotron-based ultrahigh resolution imaging ex vivo, and a combination of biological, histological, and flow cytometry-based cellular and molecular approaches in vitro. Systemic blockade of TGFß using a monoclonal antibody induces a transition from a self-contained aortic dilatation to a model of sustained aneurysmal growth, associated with the formation of an intraluminal thrombus. AAA growth is associated with wall disruption but no medial dissection and culminates in fatal transmural aortic wall rupture. TGFß blockade enhances leukocyte infiltration both in the aortic wall and the intraluminal thrombus and aggravates extracellular matrix degradation. Early blockade of IL-1ß or monocyte-dependent responses substantially limits AAA severity. However, blockade of IL-1ß after disease initiation has no effect on AAA progression to rupture. CONCLUSIONS: Endogenous TGFß activity is required for the healing of AAA. TGFß blockade may be harnessed to generate new models of AAA with better relevance to the human disease. We expect that the new models will improve our understanding of the pathophysiology of AAA and will be useful in the identification of new therapeutic targets.


Assuntos
Anticorpos Monoclonais/toxicidade , Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/induzido quimicamente , Ruptura Aórtica/induzido quimicamente , Elastase Pancreática , Fator de Crescimento Transformador beta/antagonistas & inibidores , Remodelação Vascular/efeitos dos fármacos , Animais , Aorta Abdominal/imunologia , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Ruptura Aórtica/imunologia , Ruptura Aórtica/metabolismo , Ruptura Aórtica/patologia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Dilatação Patológica , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Interleucina-1beta/metabolismo , Cinética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síncrotrons , Trombose/induzido quimicamente , Trombose/metabolismo , Trombose/patologia , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo , Ultrassonografia , Cicatrização/efeitos dos fármacos
18.
Toxicol Pathol ; 46(4): 408-420, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29768985

RESUMO

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.


Assuntos
Anticorpos Monoclonais Humanizados/toxicidade , Osso e Ossos/efeitos dos fármacos , Articulações/efeitos dos fármacos , Fator de Crescimento Neural/antagonistas & inibidores , Animais , Anticorpos Monoclonais/toxicidade , Macaca fascicularis , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Tomografia Computadorizada por Raios X
19.
Regul Toxicol Pharmacol ; 98: 98-107, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30026135

RESUMO

Nonclinical safety testing of biopharmaceuticals can present significant challenges to human risk assessment with these innovative and often complex drugs. Emerging topics in this field were discussed recently at the 2016 Annual US BioSafe General Membership meeting. The presentations and subsequent discussions from the main sessions are summarized. The topics covered included: (i) specialty biologics (oncolytic virus, gene therapy, and gene editing-based technologies), (ii) the value of non-human primates (NHPs) for safety assessment, (iii) challenges in the safety assessment of immuno-oncology drugs (T cell-dependent bispecifics, checkpoint inhibitors, and costimulatory agonists), (iv) emerging therapeutic approaches and modalities focused on microbiome, oligonucleotide, messenger ribonucleic acid (mRNA) therapeutics, (v) first in human (FIH) dose selection and the minimum anticipated biological effect level (MABEL), (vi) an update on current regulatory guidelines, International Council for Harmonization (ICH) S1, S3a, S5, S9 and S11 and (vii) breakout sessions that focused on bioanalytical and PK/PD challenges with bispecific antibodies, cytokine release in nonclinical studies, determining adversity and NOAEL for biologics, the value of second species for toxicology assessment and what to do if there is no relevant toxicology species.


Assuntos
Produtos Biológicos/toxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Animais , Anticorpos Monoclonais/toxicidade , Terapia Baseada em Transplante de Células e Tecidos , Terapia Genética , Humanos , Proteínas Recombinantes/toxicidade , Medição de Risco
20.
Regul Toxicol Pharmacol ; 98: 69-79, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30009863

RESUMO

Toxicity studies in pregnant animals are not always necessary for assessing the human risk of developmental toxicity of biopharmaceuticals. The growing experience and information on target biology and molecule-specific pharmacokinetics present a powerful approach to accurately anticipate effects of target engagement by biopharmaceuticals using a weight of evidence approach. The weight of evidence assessment should include all available data including target biology, pharmacokinetics, class effects, genetically modified animals, human mutations, and a thorough literature review. When assimilated, this weight of evidence evaluation may be sufficient to inform risk for specific clinical indications and patient populations. While under current guidance this approach is only applicable for drugs and biologics for oncology, the authors would like to suggest that this approach may also be appropriate for other disease indications. When there is an unacceptable level of uncertainty and a toxicity study in pregnant animals could impact human risk assessment, then such studies should be considered. Determination of appropriate nonclinical species for developmental toxicity studies to inform human risk should consider species-specific limitations, reproductive physiology, and pharmacology of the biopharmaceutical. This paper will provide considerations and examples of the weight of evidence approach to evaluating the human risk of developmental toxicity of biopharmaceuticals.


Assuntos
Anticorpos Monoclonais/toxicidade , Produtos Biológicos/toxicidade , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Fetal/efeitos dos fármacos , Teratogênicos/toxicidade , Animais , Humanos , Medição de Risco , Testes de Toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA