Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Métodos Terapêuticos e Terapias MTCI
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
MAbs ; 12(1): 1763727, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32449878

RESUMO

The importance of speed to clinic for medicines that may address unmet medical needs puts pressure on product development timelines. Historically, both toxicology and first-in-human clinical materials are generated using the same clonal-derived cells to ensure safety and minimize any development risks. However, cell line development with single cell cloning is time consuming, and aggravated by the time needed to screen for a lead clone based on cell line stability and manufacturability. In order to achieve faster timelines, we have used pools of up to six clones for earlier production of drug substance for regulatory filing-enabling toxicology studies, and then the final single clone was selected for production of clinical materials. This approach was enabled by using platform processes across all stages of early development, including expression vectors, host cell lines, media, and production processes. Through comprehensive cell culture and product quality analysis, we demonstrated that the toxicology material was representative of the clinical material for all six monoclonal antibody programs evaluated. Our extensive development experience further confirmed that using a pool of clones for toxicology material generation is a reliable approach to shorten the early development timeline.


Assuntos
Anticorpos Monoclonais/imunologia , Células Clonais/imunologia , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/metabolismo , Proteínas Recombinantes/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/toxicidade , Células CHO , Células Clonais/efeitos dos fármacos , Cricetinae , Cricetulus , Drogas em Investigação/uso terapêutico , Drogas em Investigação/toxicidade , Humanos , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Testes de Toxicidade/métodos
2.
Mol Cancer Ther ; 18(10): 1832-1843, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31350344

RESUMO

The FGFR4/FGF19 signaling axis is overactivated in 20% of liver tumors and currently represents a promising targetable signaling mechanism in this cancer type. However, blocking FGFR4 or FGF19 has proven challenging due to its physiological role in suppressing bile acid synthesis which leads to increased toxic bile acid plasma levels upon FGFR4 inhibition. An FGFR4-targeting antibody, U3-1784, was generated in order to investigate its suitability as a cancer treatment without major side effects.U3-1784 is a high-affinity fully human antibody that was obtained by phage display technology and specifically binds to FGFR4. The antibody inhibits cell signaling by competing with various FGFs for their FGFR4 binding site thereby inhibiting receptor activation and downstream signaling via FRS2 and Erk. The inhibitory effect on tumor growth was investigated in 10 different liver cancer models in vivo The antibody specifically slowed tumor growth of models overexpressing FGF19 by up to 90% whereas tumor growth of models not expressing FGF19 was unaffected. In cynomolgus monkeys, intravenous injection of U3-1784 caused elevated serum bile acid and liver enzyme levels indicating potential liver damage. These effects could be completely prevented by the concomitant oral treatment with the bile acid sequestrant colestyramine, which binds and eliminates bile acids in the gut. These results offer a new biomarker-driven treatment modality in liver cancer without toxicity and they suggest a general strategy for avoiding adverse events with FGFR4 inhibitors.


Assuntos
Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais/uso terapêutico , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/imunologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Resina de Colestiramina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos , Sorafenibe/farmacologia
3.
Int J Mol Sci ; 19(11)2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30469350

RESUMO

Biotherapeutics are a rapidly growing portion of the total pharmaceutical market accounting for almost one-half of recent new drug approvals. A major portion of these approvals each year are monoclonal antibodies (mAbs). During development, non-clinical pharmacology and toxicology testing of mAbs differs from that done with chemical entities since these biotherapeutics are derived from a biological source and therefore the animal models must share the same epitopes (targets) as humans to elicit a pharmacological response. Mechanisms of toxicity of mAbs are both pharmacological and non-pharmacological in nature; however, standard in silico predictive toxicological methods used in research and development of chemical entities currently do not apply to these biotherapeutics. Challenges and potential opportunities exist for new methodologies to provide a more predictive program to assess and monitor potential adverse drug reactions of mAbs for specific patients before and during clinical trials and after market approval.


Assuntos
Anticorpos Monoclonais/toxicidade , Terapia Biológica/métodos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Terapia Biológica/tendências , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Humanos
4.
Toxicol Sci ; 166(1): 192-202, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30099540

RESUMO

CFZ533 is a pathway blocking, nondepleting anti-CD40 antibody that is in clinical development for inhibition of transplant organ rejection and therapy for autoimmune diseases. A 26-week GLP toxicity study in sexually mature Cynomolgus monkeys was conducted in order to support chronic application of CFZ533. CFZ533 was subcutaneously administered at doses up to 150 mg/kg/week and was safe and generally well tolerated. CFZ533 showed no adverse effects for cardiovascular, respiratory, and neurobehavioral endpoints, and no changes were observed for blood lymphocyte and platelet counts or blood coagulation markers. In line with the nondepleting nature of CFZ533, CD20+ B cells in the blood were only marginally reduced. A complete suppression of germinal center (GC) development in lymph nodes and spleen was the most prominent result of post-mortem histological investigations. This was corroborated by an abrogated T-dependent antibody response (TDAR) to the antigen Keyhole Limpet Hemocyanin (KLH) as well as an absence of anti-drug antibodies (ADAs) in the absence of B cell depletion as seen with immunophenotyping and histology. When serum levels of CFZ533 in recovery animals dropped levels necessary for full CD40 occupancy on B cells, all animals were able to mount a TDAR to KLH. All histological changes also reverted to normal appearance after recovery. In summary, CFZ533 was shown to be well tolerated and safe in the 26-week toxicity study with a distinct pharmacodynamic profile in histology and immune function.


Assuntos
Anticorpos Monoclonais/toxicidade , Linfócitos B/efeitos dos fármacos , Antígenos CD40/imunologia , Animais , Anticorpos Monoclonais/sangue , Linfócitos B/citologia , Linfócitos B/imunologia , Reações Cruzadas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Hemocianinas/imunologia , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Injeções Intravenosas , Macaca fascicularis , Masculino , Testes de Toxicidade , Toxicocinética
5.
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
6.
Toxicol In Vitro ; 45(Pt 3): 296-308, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28263892

RESUMO

Many monoclonal antibodies (mAbs) licensed for human use or in clinical development for cancer and autoimmune disease directly interact with the immune system. These immunomodulatory mAbs have an inherent risk of adverse immune-mediated drug reactions, including infusion reactions, cytokine storms, immunosuppression and autoimmunity. A thorough understanding of the potential for immunotoxicity of a mAb is required to support administration to humans. This review will highlight the key role of in vitro assays in defining the immunopharmacology, immunotoxicity and immunogenicity of mAbs. A wide range of in vitro tests with multiple formats of different complexity can be utilized to characterize i) the antibody-binding domains of the mAb, such as on-target binding and downstream pharmacological effects (e.g. immunosuppression, immune activation, cytokine release) in both humans and animal species used for toxicology studies and off-target binding; ii) Fc-dependent effects such as Fc-mediated cellular activation (e.g. of leukocytes, platelets) and cytokine release, complement activation; and iii) product-related factors (sequence, physical-chemical properties and impurities) that can impact both pharmacological activity and immunogenicity potential of a mAb. These assays can be crucial to the selection of mAbs with an optimum balance of safety and efficacy, in defining whether a mAb is a high risk molecule, and together with animal data, can inform human safe starting doses and escalation schemes.


Assuntos
Anticorpos Monoclonais/toxicidade , Fatores Imunológicos/toxicidade , Animais , Anticorpos Monoclonais/efeitos adversos , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Humanos , Fatores Imunológicos/efeitos adversos , Técnicas In Vitro , Medição de Risco , Segurança , Especificidade da Espécie
7.
Drug Discov Today Technol ; 21-22: 85-93, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27978992

RESUMO

While immunomodulatory monoclonal antibodies (mAbs) have a wide therapeutic potential, exaggerated immunopharmacology may drive both acute and delayed immunotoxicity. The existing tools for immunotoxicity assessment do not accurately predict the full range of immunotoxicities observed in humans. New and optimized models, assays, endpoints and biomarkers in animals and humans are required to safeguard patients and allow them access to these often transformational therapies.


Assuntos
Anticorpos Monoclonais/toxicidade , Fatores Imunológicos/toxicidade , Animais , Avaliação Pré-Clínica de Medicamentos , Humanos , Testes de Toxicidade , Pesquisa Translacional Biomédica
8.
J Pharm Biomed Anal ; 131: 309-315, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27619177

RESUMO

Unwanted immunogenicity of protein therapeutics can result in severe side effects and should be assessed in animals before applying the treatment to humans. Monkeys are the most relevant choice for pre-clinical toxicity testing of antibody-based therapeutics. To assess the immunogenicity of HD105, a novel antibody therapeutic that targets both vascular endothelial growth factor and Delta-like-ligand 4, a bridging enzyme-linked immunosorbent assay was developed as an anti-drug antibody (ADA) assay and validated for use in pre-clinical studies using non-human primates. This method was found to have suitable assay sensitivity, intra- and inter-assay precision, confirmation, drug tolerance, recovery, and sample stability for measuring ADA in monkey serum samples. The results showed that ADA elevation occurred following repeated doses of HD105, and that ADA production was negatively associated with serum HD105 concentration. These results suggest that intravenous administration of HD105 induces production of ADA in monkeys and that the detection of ADA may be negatively influenced by free HD105 in serum.


Assuntos
Anticorpos Monoclonais/sangue , Autoanticorpos/sangue , Química Farmacêutica/normas , Animais , Anticorpos Monoclonais/toxicidade , Autoanticorpos/efeitos dos fármacos , Química Farmacêutica/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Ensaio de Imunoadsorção Enzimática/normas , Feminino , Humanos , Macaca fascicularis , Masculino , Reprodutibilidade dos Testes
9.
Reprod Toxicol ; 64: 98-104, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27112525

RESUMO

Details of embryo-fetal development (EFD) studies were compiled from published FDA approval documents for 43 small molecule drugs (2014-2015) and 37 monoclonal antibodies (mAbs, 2002-2015). Anti-cancer agents were analyzed separately. Rats and rabbits were the species used for EFD studies on 93% of small molecule drugs. Overall, the rat and rabbit were equally sensitive to maternal and fetal toxicity (including teratogenicity). Dosages equivalent to more than 50-times the human exposure (or 10-times for mAbs) were frequently used, but were unnecessary for 90% of drugs. EFD studies were not required for several recently approved mAbs owing to pre-existing scientific knowledge. The cynomolgus monkey was used for developmental toxicity testing of 75% of mAbs, frequently using an ePPND study design. Studies in pregnant rodents using homologous murine antibodies supplemented or replaced monkey studies under some circumstances. Most anti-cancer small molecules and mAbs were tested for developmental toxicity in at least one species.


Assuntos
Anticorpos Monoclonais/toxicidade , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Fetal/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/toxicidade , Animais , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Feminino , Gravidez , Coelhos , Ratos , Especificidade da Espécie , Estados Unidos , United States Food and Drug Administration
10.
Ann Rheum Dis ; 75(1): 253-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25165034

RESUMO

OBJECTIVE: Biological disease-modifying antirheumatic drugs (DMARDs) that inhibit aberrant immune reactions in rheumatoid arthritis (RA) cannot induce complete remission in all patients. Combination therapies using two biological DMARDs have failed to exert additive effects and increased serious infections. We have found that cell cycle inhibition of synovial fibroblasts with cyclin-dependent kinase (CDK) inhibitors ameliorated the disease in animal models of RA without attenuating acquired immunity. The objective of this study was to determine whether a clinically well-tolerated selective CDK 4/6 inhibitor (CDKI), palbociclib, is effective and whether combination with cytokine blockers acts additively without enhancing immune suppression. METHODS: The effects of CDKI on haematopoiesis and physical and behavioural findings in mice were evaluated. Mice with collagen-induced arthritis (CIA) were treated with CDKI, etanercept or anti-interleukin (IL)-6 receptor antibody (MR16-1) alone or with a combination of CDKI with etanercept or MR16-1. Their clinical, histological and radiographic scores, serum anti-(type II collagen (CII)) antibody levels and proliferative responses of lymph node cells to CII were determined. RESULTS: Although CDKI induced marginal myelosuppression, it was well tolerated and ameliorated CIA dose-dependently. The combinations of low-dose CDKI and either tumour necrosis factor-α or IL-6 blocker enhanced the antiarthritic effects additively. The addition of CDKI to either cytokine blocker did not affect the levels of anti-CII antibodies and proliferative responses of lymphocytes to CII. CONCLUSIONS: A clinically well-tolerated CDK4/6 inhibitor exerted antiarthritic effects in this mouse model. By combining therapeutic agents targeting immune reaction and synovial proliferation, we have demonstrated for the first time that two molecular targeting treatments act additively and may not increase immune suppression.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Produtos Biológicos/uso terapêutico , Piperazinas/uso terapêutico , Piridinas/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/toxicidade , Formação de Anticorpos/efeitos dos fármacos , Antirreumáticos/administração & dosagem , Antirreumáticos/toxicidade , Artrite Experimental/imunologia , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Produtos Biológicos/administração & dosagem , Produtos Biológicos/toxicidade , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Quimioterapia Combinada , Etanercepte/administração & dosagem , Etanercepte/uso terapêutico , Etanercepte/toxicidade , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos Endogâmicos DBA , Terapia de Alvo Molecular/métodos , Piperazinas/administração & dosagem , Piperazinas/toxicidade , Piridinas/administração & dosagem , Piridinas/toxicidade , Receptores de Interleucina-6/antagonistas & inibidores
11.
Pharm Res ; 32(11): 3584-92, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26108879

RESUMO

Antibody drug conjugates (ADCs) are promising therapies currently in development for oncology with unique and challenging regulatory and scientific considerations. While there are currently no regulatory guidelines specific for the nonclinical development of ADCs, there are harmonized international guidelines (e.g., ICHS6(R1), ICHM3(R2), ICHS9) that apply to ADCs and provide a framework for their complex development with issues that apply to both small and large molecules. The regulatory and scientific perspectives on ADCs are evolving due to both the advances in ADC technology and a better understanding of the safety and efficacy of ADCs in clinical development. This paper introduces the key scientific and regulatory aspects of the nonclinical development of ADCs, discusses important regulatory and scientific issues in the nonclinical to clinical dose translation of ADCs, and introduces new concepts in the areas of pharmacokinetic/pharmacodynamic (PK/PD) modeling and simulation.


Assuntos
Anticorpos Monoclonais/toxicidade , Descoberta de Drogas/métodos , Imunoconjugados/toxicidade , Testes de Toxicidade/métodos , Animais , Anticorpos Monoclonais/química , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Guias como Assunto , Humanos , Imunoconjugados/química , Legislação de Medicamentos
12.
AAPS J ; 17(5): 1055-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26024656

RESUMO

Antibody drug conjugates (ADCs) are biopharmaceutical molecules consisting of a cytotoxic small molecule covalently linked to a targeted protein carrier via a stable cleavable or noncleavable linker. The process of conjugation yields a highly complex molecule with biochemical properties that are distinct from those of the unconjugated components. The impact of these biochemical differences on the safety and pharmacokinetic (PK) profile of the conjugate must be considered when determining the types of nonclinical safety studies required to support clinical development of ADCs. The hybrid nature of ADCs highlights the need for a science-based approach to safety assessment that incorporates relevant aspects of small and large molecule testing paradigms. This thinking is reflected in current regulatory guidelines, where sections pertaining to conjugates allow for a flexible approach to nonclinical safety testing. The aim of this article is to review regulatory expectations regarding early assessment of nonclinical safety considerations and discuss how recent advances in our understanding of ADC-mediated toxicity can be used to guide the types of nonclinical safety studies needed to support ADC clinical development. The review will also explore nonclinical testing strategies that can be used to streamline ADC development by assessing the safety and efficacy of next generation ADC constructs using a rodent screen approach.


Assuntos
Anticorpos Monoclonais/toxicidade , Imunoconjugados/toxicidade , Testes de Toxicidade/métodos , Animais , Anticorpos Monoclonais/química , Antineoplásicos/química , Antineoplásicos/toxicidade , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Imunoconjugados/química , Legislação de Medicamentos
13.
MAbs ; 6(3): 697-707, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24598534

RESUMO

Antagonist anti-CD28 antibodies prevent T-cell costimulation and are functionally different from CTLA4Ig since they cannot block CTLA-4 and PDL-1 co-inhibitory signals. They demonstrated preclinical efficacy in suppressing effector T cells while enhancing immunoregulatory mechanisms. Because a severe cytokine release syndrome was observed during the Phase 1 study with the superagonist anti-CD28 TGN1412, development of other anti-CD28 antibodies requires careful preclinical evaluation to exclude any potential immunotoxicity side-effects. The failure to identify immunological toxicity of TGN1412 using macaques led us to investigate more relevant preclinical models. We report here that contrary to macaques, and like in man, all baboon CD4-positive T lymphocytes express CD28 in their effector memory cells compartment, a lymphocyte subtype that is the most prone to releasing cytokines after reactivation. Baboon lymphocytes are able to release pro-inflammatory cytokines in vitro in response to agonist or superagonist anti-CD28 antibodies. Furthermore, we compared the reactivity of human and baboon lymphocytes after transfer into non obese diabetic/severe combined immunodeficiency (NOD/SCID) interleukin-2rγ knockout mice and confirmed that both cell types could release inflammatory cytokines in situ after injection of agonistic anti-CD28 antibodies. In contrast, FR104, a monovalent antagonistic anti-CD28 antibody, did not elicit T cell activation in these assays, even in the presence of anti-drug antibodies. Infusion to baboons also resulted in an absence of cytokine release. In conclusion, the baboon represents a suitable species for preclinical immunotoxicity evaluation of anti-CD28 antibodies because their effector memory T cells do express CD28 and because cytokine release can be assessed in vitro and trans vivo.


Assuntos
Anticorpos Bloqueadores/toxicidade , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/toxicidade , Antígenos CD28/antagonistas & inibidores , Papio anubis/imunologia , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/toxicidade , Antígenos CD28/imunologia , Citocinas/biossíntese , Avaliação Pré-Clínica de Medicamentos , Humanos , Memória Imunológica , Ativação Linfocitária , Macaca fascicularis , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Modelos Animais , Especificidade da Espécie , Linfócitos T/imunologia
14.
Regul Toxicol Pharmacol ; 67(3): 382-91, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24012707

RESUMO

Antibody drug conjugates (ADCs) include monoclonal antibodies that are linked to cytotoxic small molecules. A number of these agents are currently being developed as anti-cancer agents designed to improve the therapeutic index of the cytotoxin (i.e., cytotoxic small molecule or cytotoxic agent) by specifically delivering it to tumor cells. This paper presents primary considerations for the nonclinical safety evaluation of ADCs and includes strategies for the evaluation of the entire ADC or the various individual components (i.e., antibody, linker or the cytotoxin). Considerations are presented on how to design a nonclinical safety assessment program to identify the on- and off-target toxicities to enable first-in-human (FIH) studies. Specific discussions are also included that provide details as to the need and how to conduct the studies for evaluating ADCs in genetic toxicology, tissue cross-reactivity, safety pharmacology, carcinogenicity, developmental and reproductive toxicology, biotransformation, toxicokinetic monitoring, bioanalytical assays, immunogenicity testing, test article stability and the selection of the FIH dose. Given the complexity of these molecules and our evolving understanding of their properties, there is no single all-encompassing nonclinical strategy. Instead, each ADC should be evaluated on a case-by-case scientifically-based approach that is consistent with ICH and animal research guidelines.


Assuntos
Anticorpos Monoclonais/toxicidade , Antineoplásicos/toxicidade , Imunoconjugados/toxicidade , Testes de Toxicidade , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Antineoplásicos/química , Antineoplásicos/imunologia , Antineoplásicos/farmacocinética , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Guias como Assunto , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Projetos de Pesquisa , Testes de Toxicidade/métodos , Testes de Toxicidade/normas
15.
Int J Toxicol ; 32(2): 100-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23559642

RESUMO

ALXN4100TPO, a thrombopoietin (TPO) receptor agonist, increases platelets, abrogates radiation-induced thrombocytopenia and affords significant survival benefit to lethally irradiated mice. This preliminary nonclinical safety study assessed effects of a single subcutaneous (sc) administration of ALXN4100TPO in CD2F1 mice randomized into naïve, control antibody (ALXN4200, 100 mg/kg), low (1 mg/kg), medium (10 mg/kg), or high (100 mg/kg) ALXN4100TPO doses. End points included clinical observations, body weight changes, hematology, histopathology, pharmacokinetics, pharmacodynamics by measuring platelet counts, and endogenous TPO (eTPO) levels. Salient findings were prominent increase in platelet counts and end cells of myeloid and lymphoid lineages; elevated megakaryopoiesis in bone marrow; and extramedullary hematopoiesis in spleen and liver. Serum ALXN4100TPO levels were maximum 24 hours after administration, with a half-life of 13 days. Endogenous TPO levels were elevated in 10 and 100 mg/kg ALXN4100TPO-treated groups. In conclusion, ALXN4100TPO (1-100 mg/kg, sc) treatment in CD2F1 mice resulted in profound pharmacological changes in the hematopoietic tissue; however, no life-threatening adverse events were observed.


Assuntos
Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Receptores de Trombopoetina/agonistas , Trombopoetina/farmacocinética , Trombopoetina/toxicidade , Animais , Anticorpos Monoclonais Humanizados , Disponibilidade Biológica , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos , Contagem de Plaquetas , Baço/efeitos dos fármacos , Baço/patologia , Trombocitopenia/tratamento farmacológico , Trombocitopenia/patologia , Trombopoese/efeitos dos fármacos
16.
Toxicol Pathol ; 41(2): 280-302, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23471185

RESUMO

Biotherapeutics are expanding the arsenal of therapeutics available for treating and preventing disease. Although initially thought to have limited side effects due to the specificity of their binding, these drugs have now been shown to have potential for adverse drug reactions including effects on peripheral blood cell counts or function. Hematotoxicity caused by a biotherapeutic can be directly related to the activity of the biotherapeutic or can be indirect and due to autoimmunity, biological cascades, antidrug antibodies, or other immune system responses. Biotherapeutics can cause hematotoxicity primarily as a result of cellular activation, cytotoxicity, drug-dependent and independent immune responses, and sequelae from initiating cytokine and complement cascades.  The underlying pathogenesis of biotherapeutic-induced hematotoxicity often is poorly understood. Nonclinical studies have generally predicted clinical hematotoxicity for recombinant cytokines and growth factors.  However, most hematologic liabilities of biotherapeutics are not based on drug class but are species specific, immune-mediated, and of low incidence. Despite the potential for unexpected hematologic toxicity, the risk-benefit profile of most biotherapeutics is favorable; hematologic effects are readily monitorable and managed by dose modification, drug withdrawal, and/or therapeutic intervention.  This article reviews examples of biotherapeutics that have unexpected hematotoxicity in nonclinical or clinical studies. 


Assuntos
Anticorpos Monoclonais/toxicidade , Produtos Biológicos/toxicidade , Terapia Biológica/efeitos adversos , Doenças Hematológicas/induzido quimicamente , Animais , Anticorpos Monoclonais/efeitos adversos , Produtos Biológicos/efeitos adversos , Humanos
17.
Br J Pharmacol ; 166(3): 823-46, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22168282

RESUMO

Monoclonal antibodies (mAbs) and fusion proteins directed towards cell surface targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 15 currently approved mAbs and fusion proteins targeted to the cell surface. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions'; and the US Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the 15 approved biopharmaceuticals were included: abatacept; abciximab; alefacept; alemtuzumab; basiliximab; cetuximab; daclizumab; efalizumab; ipilimumab; muromonab; natalizumab; panitumumab; rituximab; tocilizumab; and trastuzumab. For statistical analysis of concordance, data from these 15 were combined with data on the approved mAbs and fusion proteins directed towards soluble targets. Good concordance with human pharmacodynamics was found for mice receiving surrogates or non-human primates (NHPs) receiving the human pharmaceutical. In contrast, there was poor concordance for human pharmacodynamics in genetically deficient mice and for human adverse effects in all three test systems. No evidence that NHPs have superior predictive value was found.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/toxicidade , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/toxicidade , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Humanos , Dose Máxima Tolerável , Nível de Efeito Adverso não Observado , Farmacologia Clínica , Valor Preditivo dos Testes , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Solubilidade , Especificidade da Espécie
18.
Br J Pharmacol ; 166(3): 806-22, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22168335

RESUMO

Monoclonal antibodies (mAbs) and fusion proteins directed towards soluble targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 14 currently approved mAbs and fusion proteins targeted to soluble targets. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions' and United States Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the following approved biopharmaceuticals were included: adalimumab, anakinra, bevacizumab, canakinumab, certolizumab pegol, denosumab, eculizumab, etanercept, golimumab, infliximab, omalizumab, ranibizumab, rilonacept and ustekinumab. Some related biopharmaceuticals in late-stage development were also included for comparison. Good concordance with human pharmacodynamics was found for both non-human primates (NHPs) receiving the human biopharmaceutical and mice receiving rodent homologues (surrogates). In contrast, there was limited concordance for human adverse effects in genetically deficient mice, mice receiving surrogates or NHPs receiving the human pharmaceutical. In summary, the results of this survey show that although both mice and NHPs have good predictive value for human pharmacodynamics, neither species have good predictive value for human adverse effects. No evidence that NHPs have superior predictive value was found.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/toxicidade , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/toxicidade , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/farmacocinética , Avaliação Pré-Clínica de Medicamentos , Humanos , Dose Máxima Tolerável , Nível de Efeito Adverso não Observado , Farmacologia Clínica , Valor Preditivo dos Testes , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/farmacocinética , Solubilidade , Especificidade da Espécie
19.
Am J Cardiol ; 107(9): 1375-80, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21371685

RESUMO

As breast cancer survival increases, cardiotoxicity associated with chemotherapeutic regimens such as anthracyclines and trastuzumab becomes a more significant issue. Assessment of the left ventricular (LV) ejection fraction fails to detect subtle alterations in LV function. The objective of this study was to evaluate whether more sensitive echocardiographic measurements and biomarkers could predict future cardiac dysfunction in chemotherapy-treated patients. Forty-three patients diagnosed with breast cancer who received anthracyclines and trastuzumab therapy underwent echocardiography and blood sampling at 3 time points (baseline and 3 and 6 months during the course of chemotherapy). The LV ejection fraction; peak systolic myocardial longitudinal, radial, and circumferential strain; echocardiographic markers of diastolic function; N-terminal pro-B-type natriuretic peptide; and high-sensitivity cardiac troponin I were measured. Nine patients (21%) developed cardiotoxicity (1 at 3 months and 8 at 6 months) as defined by the Cardiac Review and Evaluation Committee reviewing trastuzumab. A decrease in longitudinal strain from baseline to 3 months and detectable high-sensitivity cardiac troponin I at 3 months were independent predictors of the development of cardiotoxicity at 6 months. The LV ejection fraction, parameters of diastolic function, and N-terminal pro-B-type natriuretic peptide did not predict cardiotoxicity. In conclusion, cardiac troponin plasma concentrations and longitudinal strain predict the development of cardiotoxicity in patients treated with anthracyclines and trastuzumab. The 2 parameters may be useful to detect chemotherapy-treated patients who may benefit from alternative therapies, potentially decreasing the incidence of cardiotoxicity and its associated morbidity and mortality.


Assuntos
Antraciclinas/toxicidade , Anticorpos Monoclonais/toxicidade , Antineoplásicos/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Disfunção Ventricular Esquerda/induzido quimicamente , Adulto , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/tratamento farmacológico , Ecocardiografia , Feminino , Humanos , Peptídeo Natriurético Encefálico/sangue , Volume Sistólico , Trastuzumab , Resultado do Tratamento , Troponina C/sangue , Disfunção Ventricular Esquerda/sangue
20.
Regul Toxicol Pharmacol ; 59(2): 227-36, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20937341

RESUMO

A total of 26 toxicology studies performed with biological drugs (monoclonal antibodies and related immunoglobulins as well as recombinant human proteins) in the primate have been evaluated to give an insight into the types of study designs used and results. The evaluation involved examination of pivotal repeat dose studies which ranged from 2 to 13 weeks in duration, used to support early clinical investigation. Either no findings were seen or restricted to those which could largely be explained by the drug's pharmacology. Based on these results and supporting findings from a literature review of other similar drugs in development or approved for marketed use, a case has been made to revisit aspects of the standard design approach of toxicology studies for biological drugs. Thus, consideration should be given to a move away from the use of three drug-treated and numerous non-dose recovery groups currently used to support initial clinical entry to a robust toxicological assessment that could be achieved in many cases with one control and one or two drug-treated groups and with non-dose recovery groups only for the control and highest level used. An argument for not routinely measuring for anti-drug antibodies unless there is a specific drug-related reason is also made.


Assuntos
Anticorpos Monoclonais/toxicidade , Avaliação Pré-Clínica de Medicamentos/métodos , Imunoglobulinas/toxicidade , Proteínas Recombinantes/toxicidade , Testes de Toxicidade/métodos , Animais , Feminino , Macaca fascicularis , Macaca mulatta , Masculino , Dose Máxima Tolerável , Modelos Animais , Projetos de Pesquisa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA