Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Chem Soc Rev ; 41(9): 3571-83, 2012 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-22349541

RESUMO

The use of Carbon Monoxide (CO) as a therapeutic agent has already been tested in human clinical trials. Pre-clinically, CO gas administration proved beneficial in animal models of various human diseases. However, the use of gaseous CO faces serious obstacles not the least being its well-known toxicity. To fully realise the promise of CO as a therapeutic agent, it is key to find novel avenues for CO delivery to diseased tissues in need of treatment, without concomitant formation of elevated, toxic blood levels of carboxyhemoglobin (COHb). CO-releasing molecules (CO-RMs) have the potential to constitute safe treatments if CO release in vivo can be controlled in a spatial and temporal manner. It has already been demonstrated in animals that CO-RMs can release CO and mimic the therapeutic effects of gaseous CO. While demonstrating the principle of treatment with CO-RMs, these first generation compounds are not suitable for human use. This tutorial review summarises the biological and chemical behaviour of CO, the current status of CO-RM development, and derives principles for the creation of the next generation of CO-RMs for clinical applications in humans.


Assuntos
Monóxido de Carbono/química , Monóxido de Carbono/farmacologia , Monóxido de Carbono/uso terapêutico , Carboxihemoglobina/química , Carboxihemoglobina/metabolismo , Doenças Cardiovasculares/tratamento farmacológico , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/uso terapêutico , Humanos , Fígado/efeitos dos fármacos , Compostos Organometálicos/química , Teoria Quântica
2.
Cancer Res ; 68(22): 9280-90, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19010901

RESUMO

HER2 is a validated target in breast cancer therapy. Two drugs are currently approved for HER2-positive breast cancer: trastuzumab (Herceptin), introduced in 1998, and lapatinib (Tykerb), in 2007. Despite these advances, some patients progress through therapy and succumb to their disease. A variation on antibody-targeted therapy is utilization of antibodies to deliver cytotoxic agents specifically to antigen-expressing tumors. We determined in vitro and in vivo efficacy, pharmacokinetics, and toxicity of trastuzumab-maytansinoid (microtubule-depolymerizing agents) conjugates using disulfide and thioether linkers. Antiproliferative effects of trastuzumab-maytansinoid conjugates were evaluated on cultured normal and tumor cells. In vivo activity was determined in mouse breast cancer models, and toxicity was assessed in rats as measured by body weight loss. Surprisingly, trastuzumab linked to DM1 through a nonreducible thioether linkage (SMCC), displayed superior activity compared with unconjugated trastuzumab or trastuzumab linked to other maytansinoids through disulfide linkers. Serum concentrations of trastuzumab-MCC-DM1 remained elevated compared with other conjugates, and toxicity in rats was negligible compared with free DM1 or trastuzumab linked to DM1 through a reducible linker. Potent activity was observed on all HER2-overexpressing tumor cells, whereas nontransformed cells and tumor cell lines with normal HER2 expression were unaffected. In addition, trastuzumab-DM1 was active on HER2-overexpressing, trastuzumab-refractory tumors. In summary, trastuzumab-DM1 shows greater activity compared with nonconjugated trastuzumab while maintaining selectivity for HER2-overexpressing tumor cells. Because trastuzumab linked to DM1 through a nonreducible linker offers improved efficacy and pharmacokinetics and reduced toxicity over the reducible disulfide linkers evaluated, trastuzumab-MCC-DM1 was selected for clinical development.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunotoxinas/uso terapêutico , Maitansina/análogos & derivados , Receptor ErbB-2/análise , Ado-Trastuzumab Emtansina , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais Humanizados , Apoptose/efeitos dos fármacos , Neoplasias da Mama/química , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Imunotoxinas/farmacocinética , Imunotoxinas/toxicidade , Maitansina/farmacocinética , Maitansina/uso terapêutico , Maitansina/toxicidade , Camundongos , Ratos , Ratos Sprague-Dawley , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Med Chem ; 49(14): 4392-408, 2006 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-16821799

RESUMO

Maytansine, a highly cytotoxic natural product, failed as an anticancer agent in human clinical trials because of unacceptable systemic toxicity. The potent cell killing ability of maytansine can be used in a targeted delivery approach for the selective destruction of cancer cells. A series of new maytansinoids, bearing a disulfide or thiol substituent were synthesized. The chain length of the ester side chain and the degree of steric hindrance on the carbon atom bearing the thiol substituent were varied. Several of these maytansinoids were found to be even more potent in vitro than maytansine. The targeted delivery of these maytansinoids, using monoclonal antibodies, resulted in a high, specific killing of the targeted cells in vitro and remarkable antitumor activity in vivo.


Assuntos
Antineoplásicos/síntese química , Maitansina/análogos & derivados , Maitansina/síntese química , Animais , Anticorpos Monoclonais/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Dissulfetos/síntese química , Dissulfetos/química , Dissulfetos/farmacologia , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Maitansina/química , Maitansina/farmacologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Relação Estrutura-Atividade , Compostos de Sulfidrila/síntese química , Compostos de Sulfidrila/química , Compostos de Sulfidrila/farmacologia , Transplante Heterólogo
4.
Cancer Res ; 66(8): 4426-33, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16618769

RESUMO

Antibody-drug conjugates are targeted anticancer agents consisting of a cytotoxic drug covalently linked to a monoclonal antibody for tumor antigen-specific activity. Once bound to the target cell-surface antigen, the conjugate must be processed to release an active form of the drug, which can reach its intracellular target. Here, we used both biological and biochemical methods to better define this process for antibody-maytansinoid conjugates. In particular, we examined the metabolic fate in cells of huC242-maytansinoid conjugates containing either a disulfide linker (huC242-SPDB-DM4) or a thioether linker (huC242-SMCC-DM1). Using cell cycle analysis combined with lysosomal inhibitors, we showed that lysosomal processing is required for the activity of antibody-maytansinoid conjugates, irrespective of the linker. We also identified and characterized the released maytansinoid molecules from these conjugates, and measured their rate of release compared with the kinetics of cell cycle arrest. Both conjugates are efficiently degraded in lysosomes to yield metabolites consisting of the intact maytansinoid drug and linker attached to lysine. The lysine adduct is the sole metabolite from the thioether-linked conjugate. However, the lysine metabolite generated from the disulfide-linked conjugate is reduced and S-methylated to yield the lipophilic and potently cytotoxic metabolite, S-methyl-DM4. These findings provide insight into the mechanism of action of antibody-maytansinoid conjugates in general, and more specifically, identify a biochemical mechanism that may account for the significantly enhanced antitumor efficacy observed with disulfide-linked conjugates.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Neoplasias do Colo/tratamento farmacológico , Imunotoxinas/farmacocinética , Maitansina/análogos & derivados , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacocinética , Reagentes de Ligações Cruzadas/farmacologia , Dissulfetos/química , Dissulfetos/farmacocinética , Dissulfetos/farmacologia , Células HT29 , Humanos , Imunotoxinas/química , Imunotoxinas/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Maitansina/química , Maitansina/farmacocinética , Camundongos , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Expert Opin Biol Ther ; 6(3): 281-91, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16503736

RESUMO

It is a commonly held belief that most treatments for disseminated cancers are only moderately effective because the agents lack cell-killing mechanisms that act specifically on cancer cells. In antibody-drug conjugates, such nonspecific cytotoxic agents are combined with exquisitely specific monoclonal antibodies that bind to tumour-associated antigens and, thus, get endowed with new pharmacological characteristics. Not only is their activity newly targeted towards tumours and tumour cells, which hopefully renders them more tumour-specific, but they also acquire much of the pharmacokinetic behaviour of the monoclonal antibody component. With the structural composition of a macromolecular protein (the antibody), a small chemical cytotoxic agent and a linker to chemically connect these two molecules, antibody-drug conjugates are some of the most complex pharmacological agents ever developed. Their development over the last 20 years or so owes much to sophisticated in vitro and in vivo preclinical testing. This review attempts to summarise and exemplify many of the factors that had to be considered during the development, with special emphasis on the in vivo pharmacology of these agents.


Assuntos
Anticorpos Antineoplásicos/administração & dosagem , Anticorpos Antineoplásicos/metabolismo , Antineoplásicos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Imunoconjugados/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Animais , Humanos , Neoplasias/metabolismo
6.
Cancer Res ; 66(6): 3214-21, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540673

RESUMO

Conjugates of the anti-CanAg humanized monoclonal antibody huC242 with the microtubule-formation inhibitor DM1 (a maytansinoid), or with the DNA alkylator DC1 (a CC1065 analogue), have been evaluated for their ability to eradicate mixed cell populations formed from CanAg-positive and CanAg-negative cells in culture and in xenograft tumors in mice. We found that in culture, conjugates of either drug killed not only the target antigen-positive cells but also the neighboring antigen-negative cells. Furthermore, we showed that, in vivo, these conjugates were effective in eradicating tumors containing both antigen-positive and antigen-negative cells. The presence of antigen-positive cells was required for this killing of bystander cells. This target cell-activated killing of bystander cells was dependent on the nature of the linker between the antibody and the drug. Conjugates linked via a reducible disulfide bond were capable of exerting the bystander effect whereas equally potent conjugates linked via a nonreducible thioether bond were not. Our data offer a rationale for developing optimally constructed antibody-drug conjugates for treating tumors that express the target antigen either in a homogeneous or heterogeneous manner.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Imunoconjugados/farmacologia , Maitansina/análogos & derivados , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Reagentes de Ligações Cruzadas/química , Feminino , Células HT29 , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Maitansina/química , Maitansina/farmacologia , Camundongos , Camundongos SCID , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Protein Sci ; 14(9): 2436-46, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16081651

RESUMO

Immunoconjugates are being explored as novel cancer therapies with the promise of target-specific drug delivery. The immunoconjugate, huN901-DM1, composed of the humanized monoclonal IgG1 antibody, huN901, and the maytansinoid drug, DM1, is being tested in clinical trials to treat small cell lung carcinoma (SCLC). huN901-DM1 contains an average of three to four DM1 drug molecules per huN901 antibody molecule. The drug molecules are linked to huN901 through random modification of huN901 at epsilon-amino groups of lysine residues, thus yielding a heterogeneous population of conjugate species. We studied the drug distribution profile of huN901-DM1 by electrospray time-of-flight mass spectrometry(ESI-TOFMS), which showed that one to six DM1 drug molecules were attached to an antibody molecule. Both light and heavy chains contained linked drugs. The conjugation sites in both chains were determined by peptide mapping using trypsin and Asp-N protease digestion. Trypsin digestion identified modified lysine residues, since these residues were no longer susceptible to enzymatic cleavage after conjugation with the drug. With respect to Asp-N digestion, modified peptides were identified by observing a mass increase corresponding to the modification. The two digestion methods provided consistent results, leading to the identification of 20 modified lysine residues in both light and heavy chains. Each lysine residue was only partially modified. No conjugation sites were found in complementarity determining regions (CDRs). Using structural models of human IgG1, it was found that modified lysine residues were on the surface in areas of structural flexibility and had large solvent accessibility.


Assuntos
Anticorpos Monoclonais/química , Imunoconjugados/química , Maitansina/análogos & derivados , Espectrometria de Massas por Ionização por Electrospray/métodos , Sequência de Aminoácidos , Glicosilação , Imunoconjugados/metabolismo , Lisina/química , Maitansina/química , Metaloendopeptidases/metabolismo , Dados de Sequência Molecular , Mapeamento de Peptídeos , Homologia de Sequência de Aminoácidos , Tripsina/metabolismo
8.
Rapid Commun Mass Spectrom ; 19(13): 1806-14, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15945030

RESUMO

Recombinant monoclonal antibody drug products play an increasingly important role in the treatment of various diseases. Antibodies are large, multi-chain proteins and antibody preparations often contain several molecular variants, which renders them heterogeneous. The heterogeneity is further increased in immunoconjugates prepared by covalently linking several drug molecules per antibody molecule. As part of the product characterization, the molecular weights of the antibodies or their drug conjugates need to be measured. Electrospray ionization mass spectrometry (ESI-MS) is well suited for the analysis of recombinant antibodies and immunoconjugates. Sample preparation is an important element of ESI-MS analysis, in particular samples need to be freed of interfering charged species, such as salts and buffer components. In this paper, Amicon centrifugal filters, reversed-phase high-performance liquid chromatography (HPLC), and size-exclusion HPLC were evaluated for sample desalting. Size-exclusion HPLC, using aqueous acetonitrile as the mobile phase, directly coupled to ESI-MS provided the best performance and was optimized for the study of immunoconjugates. The results showed that antibodies carrying covalently linked maytansinoid molecules generated charge envelope profiles that differ from those of the non-conjugated antibody. For the determination of the distribution of the various conjugate species in an immunoconjugate sample prepared by randomly linking in the average 3.6 drug molecules per antibody molecule, the experimental conditions needed to be carefully selected to allow acquisition of the whole spectrum containing the charge envelopes of all species.


Assuntos
Imunoconjugados/análise , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Espectrometria de Massas , Maitansina/química , Peso Molecular , Preparações Farmacêuticas/química , Proteínas/química , Espectrometria de Massas por Ionização por Electrospray
9.
J Pharmacol Exp Ther ; 308(3): 1073-82, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14634038

RESUMO

The humanized monoclonal antibody maytansinoid conjugate, cantuzumab mertansine (huC242-DM1) that contains on average three to four linked drug molecules per antibody molecule was evaluated in CD-1 mice for its pharmacokinetic behavior and tissue distribution, and the results were compared with those of the free antibody huC242. The pharmacokinetics in blood were similar for (125)I-labeled conjugate and antibody with terminal half-lives of 154 and 156 h, respectively. Pharmacokinetic analysis using an enzyme-linked immunosorbent assay (ELISA) method, which measures intact conjugate in plasma samples revealed a faster clearance for the conjugate corresponding to a half-life of 42.2 h. This faster clearance is explained as the result of clearance from circulation and concomitant clearance of drug from circulating conjugate through linker cleavage. An antibody-specific ELISA allowed the determination of the clearance rate of the antibody component from circulation. The drug clearance rate from circulating conjugate was then calculated as the difference between the clearance of the conjugate and the clearance of the antibody component and found to be about three times that of the antibody component. The above results were confirmed with a conjugate, huC242-[(3)H]DM1, where the linked DM1 drugs carried a stable tritium label. Tissue distribution studies with (125)I-labeled conjugate and antibody showed antibody-like behavior for the conjugate; the antibody of the conjugate did not distribute or bind significantly to any solid tissue.


Assuntos
Antineoplásicos/farmacocinética , Imunoconjugados/farmacocinética , Maitansina/farmacocinética , Animais , Anticorpos/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos Fitogênicos/farmacocinética , Ensaio de Imunoadsorção Enzimática , Feminino , Radioisótopos do Iodo , Maitansina/análogos & derivados , Camundongos , Distribuição Tecidual , Trítio
10.
Cancer Res ; 63(16): 5073-83, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12941837

RESUMO

An antagonistic monoclonal antibody, designated EM164, has been developed which binds specifically to the human insulin-like growth factor I receptor (IGF-IR) and inhibits the proliferation and survival functions of the receptor in cancer cells. EM164 was initially selected by a rapid cell-based screen of hybridoma supernatants to identify antibodies that bind to IGF-IR but not to the homologous insulin receptor and that show maximal inhibition of IGF-I-stimulated autophosphorylation of IGF-IR. EM164 binds tightly to IGF-IR with a dissociation constant K(d) of 0.1 nM, inhibits binding of IGF-I and antagonizes its effects on cells completely, and has no agonistic activity on its own. EM164 inhibits IGF-I-, IGF-II-, and serum-stimulated proliferation and survival of diverse human cancer cell lines in vitro, including breast, lung, colon, cervical, ovarian, pancreatic, melanoma, prostate, neuroblastoma, rhabdomyosarcoma, and osteosarcoma cancer lines. It also suppresses the autocrine or paracrine proliferation of several cancer cell lines. EM164 was the most potent antagonistic anti-IGF-IR antibody tested when compared with several commercially available antibodies. The in vitro inhibitory effect could be extended to in vivo tumor models, where EM164 caused regression of established BxPC-3 human pancreatic tumor xenografts in SCID mice. The antitumor effect of treatment with EM164 could be enhanced by combining it with the cytotoxic agent gemcitabine. These data support the development of EM164 as a candidate therapeutic agent that targets IGF-IR function in cancer cells.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias Experimentais/terapia , Receptor IGF Tipo 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Ciclo Celular , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Camundongos SCID , Neoplasias Experimentais/patologia , Fosforilação , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA