Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Toxicol Appl Pharmacol ; 273(2): 298-313, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24035823

ABSTRACT

Trastuzumab emtansine (T-DM1) is the first antibody-drug conjugate (ADC) approved for patients with human epidermal growth factor receptor 2 (HER2)-positive metastatic breast cancer. The therapeutic premise of ADCs is based on the hypothesis that targeted delivery of potent cytotoxic drugs to tumors will provide better tolerability and efficacy compared with non-targeted delivery, where poor tolerability can limit efficacious doses. Here, we present results from preclinical studies characterizing the toxicity profile of T-DM1, including limited assessment of unconjugated DM1. T-DM1 binds primate ErbB2 and human HER2 but not the rodent homolog c-neu. Therefore, antigen-dependent and non-antigen-dependent toxicity was evaluated in monkeys and rats, respectively, in both single- and repeat-dose studies; toxicity of DM1 was assessed in rats only. T-DM1 was well tolerated at doses up to 40 mg/kg (~4400 µg DM1/m(2)) and 30 mg/kg (~ 6000 µg DM1/m(2)) in rats and monkeys, respectively. In contrast, DM1 was only tolerated up to 0.2mg/kg (1600 µg DM1/m(2)). This suggests that at least two-fold higher doses of the cytotoxic agent are tolerated in T-DM1, supporting the premise of ADCs to improve the therapeutic index. In addition, T-DM1 and DM1 safety profiles were similar and consistent with the mechanism of action of DM1 (i.e., microtubule disruption). Findings included hepatic, bone marrow/hematologic (primarily platelet), lymphoid organ, and neuronal toxicities, and increased numbers of cells of epithelial and phagocytic origin in metaphase arrest. These adverse effects did not worsen with chronic dosing in monkeys and are consistent with those reported in T-DM1-treated patients to date.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , Antineoplastic Agents/toxicity , Blood Platelets/drug effects , Cytotoxins/toxicity , Maytansine/analogs & derivatives , Ado-Trastuzumab Emtansine , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , Blood Platelets/metabolism , Blood Platelets/pathology , Body Weight/drug effects , Body Weight/physiology , Cytotoxins/adverse effects , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Male , Maytansine/adverse effects , Maytansine/toxicity , Random Allocation , Rats , Rats, Sprague-Dawley , Trastuzumab
2.
Int J Toxicol ; 28(3): 230-53, 2009.
Article in English | MEDLINE | ID: mdl-19546261

ABSTRACT

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


Subject(s)
Biopharmaceutics/methods , Drug Evaluation, Preclinical/methods , Drugs, Investigational/toxicity , Toxicity Tests/methods , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/toxicity , Biopharmaceutics/economics , Carcinogenicity Tests , Disease Models, Animal , Drug Evaluation, Preclinical/economics , Drug Evaluation, Preclinical/standards , Drug Labeling , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Transgenic , Pregnancy , Recombinant Fusion Proteins/toxicity , Species Specificity , Toxicity Tests/economics , Toxicity Tests/standards
3.
Regul Toxicol Pharmacol ; 40(3): 219-26, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15546677

ABSTRACT

Surrogate antibodies are a potential solution to the limited safety testing possible with humanized monoclonal antibodies with restricted species cross-reactivity. However, there are currently no defined criteria by which a potential surrogate antibody should be judged prior to its use in determining safety issues for the clinical agent. We propose that, potential surrogates should undergo rigorous evaluation to assess pharmacological and toxicological activities in comparison to the clinical agent. The current studies evaluated a chimeric mouse/rat anti-mouse CD11a monoclonal antibody (muM17) as a potential surrogate for efalizumab, a humanized anti-CD11a antibody in development for psoriasis. CD11a is a subunit of lymphocyte function associated antigen-1, an integrin involved in cell-cell interactions important to immune responses and inflammation. In vitro pharmacology studies included binding affinity to whole mouse blood and inhibitory activity of muM17 in a mixed lymphocyte response assay. In vivo pharmacology was examined using a delayed type hypersensitivity assay in female CD-1 mice. The toxicology evaluation included a murine tissue cross-reactivity study and in vivo multiple dose studies in female CD-1 mice which were administered muM17 (0.1-30 mg/kg) via subcutaneous injections once a week for 4 weeks. Clinical observations, body weight, clinical pathology, T cell CD11a expression, immunogenicity, toxicokinetics, and lymphoid organ histopathology were evaluated. Finally, since reproductive safety testing would be an important application of the proposed surrogate antibody, a pilot study in pregnant mice was conducted that demonstrated proportional transfer of muM17 into the fetus. These studies demonstrated that muM17 has pharmacological and toxicological activities similar to efalizumab. The selection of dose and regimen for GLP (Good Laboratory Practice) toxicology studies and extrapolation to clinical dose levels was based on pharmacodynamic activity (CD11a downmodulation on T cells).


Subject(s)
Antibodies, Monoclonal/toxicity , CD11a Antigen/immunology , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/toxicity , Cross Reactions , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Hypersensitivity, Delayed/pathology , Immunoglobulin G/immunology , In Vitro Techniques , Leukocyte Count , Lymphocyte Culture Test, Mixed , Male , Mice , Rats , Reproduction/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL