Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Microb Cell Fact ; 20(1): 67, 2021 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-33691697

RESUMO

BACKGROUND: Most patients with acute myeloid leukemia (AML) remain uncurable and require novel therapeutic methods. Gain-of-function FMS-like tyrosine kinase 3 (FLT3) mutations are present in 30-40% of AML patients and serve as an attractive therapeutic target. In addition, FLT3 is aberrantly expressed on blasts in > 90% of patients with AML, making the FLT3 ligand-based drug conjugate a promising therapeutic strategy for the treatment of patients with AML. Here, E. coli was used as a host to express recombinant human FLT3 ligand (rhFL), which was used as a specific vehicle to deliver cytotoxic drugs to FLT3 + AML cells. METHODS: Recombinant hFL was expressed and purified from induced recombinant BL21 (DE3) E. coli. Purified rhFL and emtansine (DM1) were conjugated by an N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP) linker. We evaluated the potency of the conjugation product FL-DM1 against FLT3-expressing AML cells by examining viability, apoptosis and the cell cycle. The activation of proteins related to the activation of FLT3 signaling and apoptosis pathways was detected by immunoblotting. The selectivity of FL-DM1 was assessed in our unique HCD-57 cell line, which was transformed with the FLT3 internal tandem duplication mutant (FLT3-ITD). RESULTS: Soluble rhFL was successfully expressed in the periplasm of recombinant E. coli. The purified rhFL was bioactive in stimulating FLT3 signaling in AML cells, and the drug conjugate FL-DM1 showed activity in cell signaling and internalization. FL-DM1 was effective in inhibiting the survival of FLT3-expressing THP-1 and MV-4-11 AML cells, with half maximal inhibitory concentration (IC50) of 12.9 nM and 1.1 nM. Additionally, FL-DM1 induced caspase-3-dependent apoptosis and arrested the cell cycle at the G2/M phase. Moreover, FL-DM1 selectively targeted HCD-57 cells transformed by FLT3-ITD but not parental HCD-57 cells without FLT3 expression. FL-DM1 can also induce obvious apoptosis in primary FLT3-positive AML cells ex vivo. CONCLUSIONS: Our data demonstrated that soluble rhFL can be produced in a bioactive form in the periplasm of recombinant E. coli. FL can be used as a specific vehicle to deliver DM1 into FLT3-expressing AML cells. FL-DM1 exhibited cytotoxicity in FLT3-expressing AML cell lines and primary AML cells. FL-DM1 may have potential clinical applications in treating patients with FLT3-positive AML.


Assuntos
Antineoplásicos/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Maitansina/farmacologia , Proteínas de Membrana/farmacologia , Animais , Antineoplásicos/metabolismo , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Maitansina/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Recombinantes/biossíntese , Transdução de Sinais/efeitos dos fármacos , Tirosina Quinase 3 Semelhante a fms/metabolismo
2.
Nat Prod Rep ; 37(5): 634-652, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31764930

RESUMO

Covering: 2014-2019We review recent progress on natural products that target cytoskeletal components, including microtubules, actin, intermediate filaments, and septins and highlight their demonstrated and potential utility in the treatment of human disease. The anticancer efficacy of microtubule targeted agents identified from plants, microbes, and marine organisms is well documented. We highlight new microtubule targeted agents currently in clinical evaluations for the treatment of drug resistant cancers and the accumulating evidence that the anticancer efficacy of these agents is not solely due to their antimitotic effects. Indeed, the effects of microtubule targeted agents on interphase microtubules are leading to their potential for more mechanistically guided use in cancers as well as neurological disease. The discussion of these agents as more targeted drugs also prompts a reevaluation of our thinking about natural products that target other components of the cytoskeleton. For instance, actin active natural products are largely considered chemical probes and non-selective toxins. However, studies utilizing these probes have uncovered aspects of actin biology that can be more specifically targeted to potentially treat cancer, neurological disorders, and infectious disease. Compounds that target intermediate filaments and septins are understudied, but their continued discovery and mechanistic evaluations have implications for numerous therapeutic indications.


Assuntos
Actinas/metabolismo , Produtos Biológicos/farmacologia , Citoesqueleto/efeitos dos fármacos , Microtúbulos/efeitos dos fármacos , Animais , Produtos Biológicos/química , Colchicina/química , Colchicina/metabolismo , Colchicina/farmacologia , Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Células Eucarióticas/citologia , Células Eucarióticas/efeitos dos fármacos , Genoma , Humanos , Maitansina/química , Maitansina/metabolismo , Maitansina/farmacologia , Microtúbulos/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/patologia , Taxoides/química , Taxoides/farmacologia
3.
Bioorg Med Chem ; 28(23): 115785, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33099182

RESUMO

ADCs based on the natural product maytansine have been successfully employed clinically. In a previous report, ADCs based on hydrophilic non-cell permeable maytansinoids was presented. The authors in this report further explore the maytansine scaffold to develop tubulin inhibitors capable of cell permeation. The research resulted in amino-benzoyl-maytansinoid payloads that were further elaborated with linkers for conjugating to antibodies. This approach was applied to MUC16 tumor targeting antibodies for ovarian cancers. A positive control ADC was evaluated alongside the amino-benzoyl-maytansinoid ADC and the efficacy observed was equivalent while the isotype control ADCs had no effect.


Assuntos
Imunoconjugados/metabolismo , Maitansina/química , Moduladores de Tubulina/química , Animais , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Maitansina/metabolismo , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Relação Estrutura-Atividade , Transplante Heterólogo , Moduladores de Tubulina/metabolismo
4.
Curr Microbiol ; 76(8): 954-958, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29858620

RESUMO

Ansamitocins are extraordinarily potent antitumor agents. Ansamitocin P-3 (AP-3), which is produced by Actinosynnema pretiosum, has been developed as a cytotoxic drug for breast cancer. Despite its importance, AP-3 is of limited applicability because of the low production yield. A. pretiosum strain X47 was developed from A. pretiosum ATCC 31565 by mutation breeding and shows a relatively high AP-3 yield. Here, we analyzed the A. pretiosum X47 genome, which is ~8.13 Mb in length with 6693 coding sequences, 58 tRNA genes, and 15 rRNA genes. The DNA sequence of the ansamitocin biosynthetic gene cluster is highly similar to that of the corresponding cluster in A. pretiosum ATCC 31565, with 99.9% identity. However, RT-qPCR analysis showed that the expression levels of ansamitocin biosynthetic genes were significantly increased in X47 compared with the levels in the wild-type strain, consistent with the higher yield of AP-3 in X47. The annotated complete genome sequence of this strain will facilitate understanding the molecular mechanisms of ansamitocin biosynthesis and regulation in A. pretiosum and help further genetic engineering studies to enhance the production of AP-3.


Assuntos
Actinobacteria/genética , Actinobacteria/metabolismo , Antibióticos Antineoplásicos/metabolismo , Genoma Bacteriano , Maitansina/análogos & derivados , Análise de Sequência de DNA , Vias Biossintéticas/genética , Perfilação da Expressão Gênica , Maitansina/metabolismo , Anotação de Sequência Molecular
5.
Cancer Sci ; 108(7): 1458-1468, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28388007

RESUMO

Trastuzumab emtansine (T-DM1), an antibody-drug conjugate (ADC) consisting of human epidermal growth factor receptor 2 (HER2)-targeted mAb trastuzumab linked to antimicrotubule agent mertansine (DM1), has been approved for the treatment of HER2-positive metastatic breast cancer. Acquired resistance has been a major obstacle to T-DM1 treatment, and mechanisms remain incompletely understood. In the present study, we established a T-DM1-resistant N87-KR cell line from HER2-positive N87 gastric cancer cells to investigate mechanisms of acquired resistance and develop strategies for overcoming it. Although the kinetics of binding, internalization, and externalization of T-DM1 were the same in N87-KR cells and N87 cells, N87-KR was strongly resistant to T-DM1, but remained sensitive to both trastuzumab and DM1. T-DM1 failed to inhibit microtubule polymerization in N87-KR cells. Consistently, lysine-MCC-DM1, the active T-DM1 metabolite that inhibits microtubule polymerization, accumulated much less in N87-KR cells than in N87 cells. Furthermore, lysosome acidification, achieved by vacuolar H+ -ATPase (V-ATPase), was much diminished in N87-KR cells. Notably, treatment of sensitive N87 cells with the V-ATPase selective inhibitor bafilomycin A1 induced T-DM1 resistance, suggesting that aberrant V-ATPase activity decreases T-DM1 metabolism, leading to T-DM1 resistance in N87-KR cells. Interestingly, HER2-targeted ADCs containing a protease-cleavable linker, such as hertuzumab-vc-monomethyl auristatin E, were capable of efficiently overcoming this resistance. Our results show for the first time that a decrease in T-DM1 metabolites induced by aberrant V-ATPase activity contributes to T-DM1 resistance, which could be overcome by HER2-targeted ADCs containing different linkers, including a protease-cleavable linker. Accordingly, we propose that V-ATPase activity in lysosomes is a novel biomarker for predicting T-DM1 resistance.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Maitansina/análogos & derivados , Neoplasias Gástricas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Ado-Trastuzumab Emtansina , Animais , Anticorpos Monoclonais Humanizados/metabolismo , Antineoplásicos/metabolismo , Western Blotting , Linhagem Celular Tumoral , Humanos , Imunoconjugados/metabolismo , Imunoconjugados/farmacologia , Maitansina/metabolismo , Maitansina/farmacologia , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Receptor ErbB-2/biossíntese , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Pharm ; 13(7): 2387-96, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27248573

RESUMO

Antibody-drug conjugates (ADC) rely on the target-binding specificity of an antibody to selectively deliver potent drugs to cancer cells. IgG antibody half-life is regulated by neonatal Fc receptor (FcRn) binding. Histidine 435 of human IgG was mutated to alanine (H435A) to explore the effect of FcRn binding on the pharmacokinetics, efficacy, and tolerability of two separate maytansine-based ADC pairs with noncleavable linkers, (c-DM1 and c-H435A-DM1) and (7v-Cys-may and 7v-H435A-Cys-may). The in vitro cell-killing potency of each pair of ADCs was similar, demonstrating that H435A showed no measurable impact on ADC bioactivity. The H435A mutant antibodies showed no detectable binding to human or mouse FcRn in vitro, whereas their counterpart wild-type IgG ADCs were found to bind to FcRn at pH = 6.0. In xenograft bearing SCID mice expressing mouse FcRn, the AUC of 7v-Cys-may was 1.6-fold higher than that of 7v-H435A-may, yet the observed efficacy was similar. More severe thrombocytopenia was observed with 7v-H435A-Cys-may as compared to 7v-Cys-may at multiple dose levels. The AUC of c-DM1 was approximately 3-fold higher than that of c-H435A-DM1 in 786-0 xenograft bearing SCID mice, which led to a 3-fold difference in efficacy by dose. Murine FcRn knockout, human FcRn transgenic line 32 SCID animals bearing 786-0 xenografts showed an amplified exposure difference between c-DM1 and c-H435A-DM1 as compared to murine FcRn expressing SCID mice, leading to a 10-fold higher dose required for efficacy despite a 6-fold higher AUC of the c-H435A-DM1. The accelerated clearance observed for the noncleavable maytansine ADCs with the H435A FcRn mutation led to reduced efficacy at equivalent doses and exacerbation of clinical pathology parameters (decreased tolerability) at equivalent doses. The results show that reduced ADC clearance mediated by FcRn modulation can improve therapeutic index.


Assuntos
Anticorpos/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoconjugados/farmacologia , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Animais , Anticorpos/genética , Ligante CD27/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunoconjugados/química , Maitansina/metabolismo , Camundongos , Camundongos SCID , Receptores Fc/genética
7.
Appl Microbiol Biotechnol ; 100(6): 2641-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26572523

RESUMO

Ansamitocins isolated from Actinosynnema pretiosum, potent antitumor compounds, belong to the family of maytansinoids, and the antibody-maytansinoid conjugates are currently under different phases of clinical trials. The clinical applications of ansamitocins have stimulated extensive studies to improve their production yields. In this study, we investigated the function of a pathway-specific S treptomyces antibiotic regulatory protein (SARP) family regulator, Asm18, and observed that ectopic overexpression of the asm18 gene increased the production of N-demethyl-4,5-desepoxy-maytansinol (2) to 50 mg/L in the HGF052 + pJTU824-asm18 strain, an increase by 4.7-fold compared to that of the control strain HGF052 + pJTU824. Real-time PCR analysis showed that the overexpression of the asm18 gene selectively increased the transcription levels of the genes involved in the biosynthesis of the starter unit (asm43), polyketide assembly (asmA), post-PKS modification (asm21), as well as the transcription levels of the regulatory gene (asm8), which is a specific LAL-type activator in ansamitocin biosynthesis. With the increase of fermentation titre, seven ansamitocin analogs (1-7) including three new ones (1, 5, and 6) and maytansinol (7) were isolated from the HGF052 + pJTU824-asm18 strain. Our results not only pave the way for further improving the production of ansamitocin analogs but also indicate that the post-PKS modifications of ansamitocin biosynthesis are flexible, which brings a potential of producing maytansinol, the most fascinating intermediate for the synthesis of antibody-maytansinoid conjugates, by optimizing the HGF052 and/or HGF052 + pJTU824-asm18 strains.


Assuntos
Actinobacteria/genética , Actinobacteria/metabolismo , Antineoplásicos/metabolismo , Maitansina/análogos & derivados , Engenharia Metabólica/métodos , Expressão Gênica , Perfilação da Expressão Gênica , Genes Bacterianos , Maitansina/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
8.
Appl Microbiol Biotechnol ; 100(6): 2651-62, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26585444

RESUMO

Ansamitocin P-3 (AP-3), an amacrocyclic lactam compound, is produced by Actinosynnema pretiosum. As a group of maytansinoid antibiotics, ansamitocins have an extraordinary antitumor activity by blocking the assembly of tubulin forming into functional microtubules. The biosynthesis of ansamitocins is initialized by the formation of UDP-glucose (UDPG) which is converted from glucose-1-phosphate (G1P). In this study, we focused on the influence of enhancement of UDPG biosynthesis on the production of ansamitocins in A. pretiosum. The homologous overexpressions of phosphoglucomutase, starch phosphorylase, and UTP-G1P uridylyltransferase, respectively, could largely increase the pool sizes of G1P and UDPG and result in improved AP-3 production. The elevated intracellular glucose-6-phosphate (G6P) level provided by the enhanced glyconeogenesis had, however, no significant effects on the biosynthesis of AP-3. The G6P-G1P-UDPG pathway was therefore systematically engineered by multiple genetic modifications, and a significant increase in AP-3 production was achieved (168 mg/L of AP-3 in flask culture, 40 % higher than the control strain). We also found that the enhancement of starch assimilation pathway could also improve the assembly of AP-3 to some extent. In addition, heterologous gene overexpression from Actinosynnema mirum could result in more AP-3 biosynthesis in comparison to the corresponding homologous overexpression, suggesting an alternative and promising avenue of metabolic engineering strategy for improving AP-3 production.


Assuntos
Actinobacteria/genética , Actinobacteria/metabolismo , Vias Biossintéticas/genética , Maitansina/análogos & derivados , Engenharia Metabólica/métodos , Moduladores de Tubulina/metabolismo , Uridina Difosfato Glucose/biossíntese , Gluconeogênese , Maitansina/metabolismo , Amido/metabolismo
9.
Drug Metab Dispos ; 40(10): 1927-34, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22752008

RESUMO

The in vitro characterization of the inhibition potential of four representative maytansinoid species observed upon hepatic and/or tumor in vivo processing of antibody-maytansine conjugates (AMCs) with cleavable and noncleavable linkers is reported. We investigated the free maytansinoid species N(2')-deacetyl-N(2')-(3-mercapto-1-oxopropyl)-maytansine (DM1), (S)-methyl-DM1, and N(2')-deacetyl-N(2')-(4-mercapto-4-methyl-1-oxopentyl)-maytansine (DM4) as representative cleavable linker catabolites and Lysine-N(ε)-N-succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate-DM1 (Lys-MCC-DM1) as the representative noncleavable linker catabolite. Studies with recombinant human cytochromes P450 (P450s) indicate CYP2D6, CYP3A4, and CYP3A5 are the primary isoforms responsible for the oxidative metabolism of DM1, (S)-methyl-DM1, and DM4. Lys-MCC-DM1 was not metabolized by any of the P450 isoforms studied. DM1 was shown to be a reversible inhibitor of CYP2C8 (K(i) = 11 ± 3 µM) and CYP2D6 (K(i) = 14 ± 2 µM). Lys-MCC-DM1 and (S)-methyl-DM1 showed no reversible or time-dependent inactivation of any of the P450s studied. DM1 and DM4 inactivated CYP3A from human liver microsomes with K(i)/k(inact) values of 4.8 ± 0.9 µM/0.035 ± 0.002 min(-1) and 3.3 ± 0.2 µM/0.114 ± 0.002 min(-1), respectively. DM1 and DM4 inactivated recombinant CYP3A4 with K(i)/k(inact) values of 3.4 ± 1.0 µM/0.058 ± 0.005 min(-1) and 1.4 ± 0.3 µM/0.117 ± 0.006 min(-1), respectively. Because of instability in plasma, further characterization of the DM1 and DM4 intramolecular and intermolecular disulfide conjugates observed in vivo is required before an accurate drug-drug interaction (DDI) prediction can be made. AMCs with noncleavable thioether-linked DM1 as the cytotoxic agent are predicted to have no potential for a DDI with any of the major human P450s studied.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Inibidores das Enzimas do Citocromo P-450 , Inibidores Enzimáticos/farmacologia , Imunoconjugados/farmacologia , Maitansina/farmacologia , Anticorpos Monoclonais/metabolismo , Antineoplásicos/metabolismo , Biotransformação , Citocromo P-450 CYP2D6/metabolismo , Inibidores do Citocromo P-450 CYP2D6 , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/metabolismo , Dextrometorfano/metabolismo , Dextrorfano/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/metabolismo , Humanos , Imunoconjugados/metabolismo , Cinética , Maitansina/análogos & derivados , Maitansina/metabolismo , Microssomos Hepáticos/enzimologia , Paclitaxel/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Medição de Risco
10.
Bioanalysis ; 14(6): 357-368, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35234045

RESUMO

Aim: To report the development and validation of an LC-MS/MS method for the simultaneous determination of unconjugated payload DM4 and its metabolite S-methyl-DM4 in human plasma. Methodology: A workflow of protein precipitation followed by reduction and solid phase extraction was employed to remove antibody-maytansinoid conjugates from plasma matrix, release DM4 from endogenous conjugates, and generate a clean sample extract for analysis, respectively. Sodium adduct species of both analytes were selected for multiple reaction monitoring to meet the assay sensitivity requirement in liquid chromatography with tandem mass spectrometry. Conclusion: The method was fully validated for a dynamic range of 0.100-50.0 ng/ml for both analytes along with desired stability and acceptable incurred sample reanalysis.


Assuntos
Imunoconjugados/sangue , Maitansina/sangue , Cromatografia Líquida , Humanos , Imunoconjugados/química , Imunoconjugados/metabolismo , Maitansina/análogos & derivados , Maitansina/metabolismo , Espectrometria de Massas em Tandem
11.
Bioconjug Chem ; 22(4): 728-35, 2011 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-21391620

RESUMO

Antibody-maytansinoid conjugates (AMCs) are targeted chemotherapeutic agents consisting of a potent microtubule-depolymerizing maytansinoid (DM1 or DM4) attached to lysine residues of a monoclonal antibody (mAb) using an uncleavable thioether linker or a stable disulfide linker. Most of the administered dose of an antibody-based therapeutic is slowly catabolized by the liver and other tissues of the reticuloendothelial system. Maytansinoids released from an AMC during this catabolic process could potentially be a source of toxicity. To investigate this, we isolated and identified liver metabolites in mice for three different [(3)H]AMCs with structures similar to those currently undergoing evaluation in the clinic. We then synthesized each metabolite to confirm the identification and assessed their cytotoxic potencies when added extracellularly. We found that the uncleavable mAb-SMCC-[(3)H]DM1 conjugate was degraded to a single major maytansinoid metabolite, lysine-SMCC-[(3)H]DM1, that was nearly 50-fold less cytotoxic than maytansine. The two disulfide-linked conjugates, mAb-SPP-[(3)H]DM1 and mAb-SPDB-[(3)H]DM4, were also found to be catabolized to the analogous lysine-linked maytansinoid metabolites. However, subsequent reduction, S-methylation, and NADPH-dependent oxidation steps in the liver yielded the corresponding S-methyl sulfoxide and S-methyl sulfone derivatives. The cytotoxic potencies of the oxidized maytansinoids toward several human carcinoma cell lines were found to be 5- to 50-fold less potent than maytansine. Our results suggest that liver plays an important role in the detoxification of both cleavable and uncleavable AMCs.


Assuntos
Anticorpos Monoclonais/metabolismo , Desenho de Fármacos , Fígado/metabolismo , Maitansina/metabolismo , Animais , Anticorpos Monoclonais/química , Feminino , Fígado/química , Maitansina/análogos & derivados , Maitansina/química , Camundongos , Camundongos Endogâmicos , Estrutura Molecular
12.
Phys Biol ; 8(5): 056004, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21836336

RESUMO

We propose a stochastic model that accounts for the growth, catastrophe and rescue processes of steady-state microtubules assembled from MAP-free tubulin in the possible presence of a microtubule-associated drug. As an example of the latter, we both experimentally and theoretically study the perturbation of microtubule dynamic instability by S-methyl-D-DM1, a synthetic derivative of the microtubule-targeted agent maytansine and a potential anticancer agent. Our model predicts that among the drugs that act locally at the microtubule tip, primary inhibition of the loss of GDP tubulin results in stronger damping of microtubule dynamics than inhibition of GTP tubulin addition. On the other hand, drugs whose action occurs in the interior of the microtubule need to be present in much higher concentrations to have visible effects.


Assuntos
Antineoplásicos/farmacologia , Maitansina/análogos & derivados , Microtúbulos/efeitos dos fármacos , Animais , Antineoplásicos/metabolismo , Sítios de Ligação , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Cinética , Maitansina/metabolismo , Maitansina/farmacologia , Microtúbulos/metabolismo , Ligação Proteica , Ouriços-do-Mar , Processos Estocásticos , Tubulina (Proteína)/metabolismo
13.
Bioconjug Chem ; 21(9): 1588-95, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20698491

RESUMO

In the manufacture of the antibody-drug conjugate Trastuzumab-DM1 (T-DM1), the lysine residues on the antibody trastuzumab (Tmab) are modified to form the intermediate Tmab-MCC (T-MCC) and then conjugated with the drug DM1. Our goal is to understand the effects of modification and conjugation steps on the physicochemical stability of the antibody. The structural stability of Tmab relative to its modified and conjugated forms was assessed, employing thermally induced stress conditions to formulations containing Tmab, T-MCC, and T-DM1. DSC, SEC, CE-SDS, and LC-MS were used to study the stability of Tmab, T-MCC, and T-DM1 to thermal stress. The DSC thermograms show a decrease in melting temperature for the CH2 transition, in the order Tmab > T-MCC > T-DM1. As per SEC analysis, a significant increase in level of aggregation was detected in T-MCC (∼32%) and T-DM1 (∼5%) after 14 days at 40 °C. Tmab did not show significant aggregate formation. CE-SDS and LC-MS data demonstrate that the aggregation in the case of T-MCC is largely covalent and involves mechanisms other than formation of intermolecular disulfide cross-links. The aggregation observed for T-MCC was significantly inhibited upon addition of amino acids with nucleophilic side chains containing thiol (Cys) and hydroxyl moieties (Ser, Tyr). The covalent aggregation observed for T-MCC and the ability of nucleophilic amino acids, particularly Cys, to inhibit it indicate that the maleimide moiety in the MCC linker may react to form intermolecular covalent cross-links between T-MCC molecules, possibly through a Michael addition mechanism. In addition, DSC results demonstrate that the conjugation of the drug moiety DM1 to Tmab results in destabilization of the CH2 domain of the antibody.


Assuntos
Anticorpos Monoclonais/química , Antineoplásicos Fitogênicos/química , Maitansina/química , Ado-Trastuzumab Emtansina , Aminoácidos/química , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos Fitogênicos/metabolismo , Química Farmacêutica , Cromatografia Líquida , Reagentes de Ligações Cruzadas/química , Estabilidade de Medicamentos , Eletroforese Capilar , Imunoconjugados/química , Imunoconjugados/metabolismo , Maleimidas/química , Maitansina/análogos & derivados , Maitansina/metabolismo , Compostos de Sulfidrila/química , Espectrometria de Massas em Tandem , Temperatura , Fatores de Tempo , Trastuzumab
14.
Bioconjug Chem ; 21(1): 84-92, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19891424

RESUMO

Antibody-drug conjugates (ADCs) are designed to eradicate cancer cells that express the target antigen on their cell surface. A key component of an ADC is the linker that covalently connects the cytotoxic agent to the antibody. Several antibody-maytansinoid conjugates prepared with disulfide-based linkers such as those targeting the CanAg antigen have been shown to display more activity in preclinical mouse xenograft models than corresponding conjugates prepared with uncleavable thioether-based linkers. To investigate how the linker influences delivery and activation of antibody-maytansinoid conjugates, we isolated and characterized the [(3)H]maytansinoids from CanAg-positive tumor tissues following a single intravenous administration of 300 microg/kg (based on maytansinoid dose) of anti-CanAg antibody (huC242)-(3)H-maytansinoid conjugates prepared with cleavable disulfide linkers and an uncleavable thioether linker. We identified three target-dependent tumor metabolites of the disulfide-linked huC242-SPDB-DM4, namely, lysine-N(epsilon)-SPDB-DM4, DM4, and S-methyl-DM4. We found similar metabolites for the less hindered disulfide-linked huC242-SPP-DM1 conjugate with the exception that no S-methyl-DM1 was detected. The sole metabolite of the uncleavable thioether-linked huC242-SMCC-DM1 was lysine-N(epsilon)-SMCC-DM1. The AUC for the metabolites of huC242-SMCC-DM1 at the tumor over 7 d was about 2-fold greater than the corresponding AUC for the metabolites of the disulfide-linked conjugates. The lipophilic metabolites of the disulfide-linked conjugates were found to be nearly 1000 times more cytotoxic than the more hydrophilic lysine-N(epsilon)-linker-maytansinoids in cell-based viability assays when added extracellularly. The cell killing properties associated with the lipophilic metabolites of the disulfide-linked conjugates (DM4 and S-methyl-DM4, and DM1) provide an explanation for the superior in vivo efficacy that is often observed with antibody-maytansinoid conjugates prepared with disulfide-based linkers in xenograft mouse models.


Assuntos
Anticorpos/metabolismo , Dissulfetos/química , Imunoconjugados/metabolismo , Imunoconjugados/uso terapêutico , Maitansina/metabolismo , Neoplasias/metabolismo , Sulfetos/química , Animais , Anticorpos/química , Anticorpos/imunologia , Anticorpos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Maitansina/química , Maitansina/imunologia , Maitansina/uso terapêutico , Camundongos , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Int J Pharm ; 582: 119331, 2020 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-32289484

RESUMO

The major drawbacks of the cytotoxin like DM1 are the off-target effects. Here, the targeting nanovesicles were developed by synthesizing tocopherol-SS-DM1 and conjugating a pH low insertion peptide (pHLIP) to PEGylated phospholipids, in which tocopherol-SS-DM1 improves the drug loading and is glutathione responsive in the cytoplasm, meanwhile, the pH insertion peptide targets the acidic microenvironment of cancer cells. Besides, these nanovesicles can accumulate at the endoplasmic reticulum and show increased cancer therapeutic effects both in vitro and in vivo. These targeting nanovesicles provide a novel formulation for subcellular organelle targeting, a platform for precisely delivery of cytotoxic DM1 to cancer cells, and an alternative strategy for antibody-drug conjugates (ADCs).


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Portadores de Fármacos , Retículo Endoplasmático/metabolismo , Glutationa/metabolismo , Lipídeos/química , Maitansina/farmacologia , Proteínas de Membrana/metabolismo , Nanopartículas , Neoplasias/tratamento farmacológico , Pró-Fármacos/farmacologia , Tocoferóis/farmacologia , Células A549 , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/metabolismo , Composição de Medicamentos , Liberação Controlada de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Células MCF-7 , Maitansina/química , Maitansina/metabolismo , Proteínas de Membrana/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Neoplasias/patologia , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Tocoferóis/química , Tocoferóis/metabolismo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Chem Biol ; 15(8): 863-74, 2008 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-18721757

RESUMO

Ansamitocins are potent antitumor maytansinoids produced by Actinosynnema pretiosum. Their biosynthesis involves the initial assembly of a macrolactam polyketide, followed by a series of postpolyketide synthase (PKS) modifications. Three ansamitocin glycosides were isolated from A. pretiosum and fully characterized structurally as novel ansamitocin derivatives, carrying a beta-D-glucosyl group attached to the macrolactam amide nitrogen in place of the N-methyl group. By gene inactivation and complementation, asm25 was identified as the N-glycosyltransferase gene responsible for the macrolactam amide N-glycosylation of ansamitocins. Soluble, enzymatically active Asm25 protein was obtained from asm25-expressing E. coli by solubilization from inclusion bodies. Its optimal reaction conditions, including temperature, pH, metal ion requirement, and Km/Kcat, were determined. Asm25 also showed broad substrate specificity toward other ansamycins and synthetic indolin-2-ones. To the best of our knowledge, this represents the first in vitro characterization of a purified antibiotic N-glycosyltransferase.


Assuntos
Amidas/metabolismo , Antineoplásicos/química , Antineoplásicos/metabolismo , Proteínas de Bactérias/metabolismo , Glucosiltransferases/metabolismo , Maitansina/análogos & derivados , Actinomycetales/enzimologia , Antifúngicos/farmacologia , Antineoplásicos/isolamento & purificação , Antineoplásicos/farmacologia , Basidiomycota/efeitos dos fármacos , Linhagem Celular Tumoral , Glicosídeos/química , Glicosídeos/metabolismo , Glicosilação , Humanos , Cinética , Lactamas/química , Lactamas/metabolismo , Maitansina/química , Maitansina/isolamento & purificação , Maitansina/metabolismo , Maitansina/farmacologia , Renaturação Proteica , Especificidade por Substrato , Uridina Difosfato Glucose/metabolismo
17.
Bioconjug Chem ; 19(8): 1673-83, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18637680

RESUMO

CD22 represents a promising target for antibody-drug conjugate therapy in the context of B cell malignancies since it rapidly internalizes, importing specifically bound antibodies with it. To determine the pharmacokinetic parameters of anti-CD22-MCC-DM1 and MC-MMAF conjugates, various approaches to quantifying total and conjugated antibody were investigated. Although the total antibody assay formats gave similar results for both conjugates, the mouse pharmacokinetic profile for the anti-CD22-MCC-DM1 and MC-MMAF appeared significantly different depending on the conjugated antibody assay format. Since these differences significantly impacted the PK parameters determination, we investigated the effect of the drug/antibody ratio on the total and conjugated antibody quantification using multiple assay formats. Our investigations revealed the limitations of some assay formats to quantify anti-CD22-MCC-DM1 and MC-MMAF with different drug load and in the context of a heterogeneous ADC population highlight the need to carefully plan the assay strategy for the total and conjugated antibody quantification in order to accurately determine the ADC PK parameters.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacocinética , Imunoconjugados/metabolismo , Imunoconjugados/farmacocinética , Maleimidas/metabolismo , Maitansina/análogos & derivados , Maitansina/metabolismo , Oligopeptídeos/metabolismo , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Animais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/imunologia , Humanos , Imunoconjugados/análise , Imunoconjugados/imunologia , Camundongos , Sensibilidade e Especificidade
18.
Nat Commun ; 9(1): 2106, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29844393

RESUMO

Microtubule-targeting agents (MTAs) like taxol and vinblastine are among the most successful chemotherapeutic drugs against cancer. Here, we describe a fluorescence anisotropy-based assay that specifically probes for ligands targeting the recently discovered maytansine site of tubulin. Using this assay, we have determined the dissociation constants of known maytansine site ligands, including the pharmacologically active degradation product of the clinical antibody-drug conjugate trastuzumab emtansine. In addition, we discovered that the two natural products spongistatin-1 and disorazole Z with established cellular potency bind to the maytansine site on ß-tubulin. The high-resolution crystal structures of spongistatin-1 and disorazole Z in complex with tubulin allowed the definition of an additional sub-site adjacent to the pocket shared by all maytansine-site ligands, which could be exploitable as a distinct, separate target site for small molecules. Our study provides a basis for the discovery and development of next-generation MTAs for the treatment of cancer.


Assuntos
Polarização de Fluorescência/métodos , Maitansina/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Ado-Trastuzumab Emtansina , Animais , Antineoplásicos/metabolismo , Sítios de Ligação , Humanos , Ligantes , Macrolídeos/metabolismo , Maitansina/análogos & derivados , Oxazóis/metabolismo , Trastuzumab/metabolismo
19.
Nucl Med Biol ; 67: 43-51, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30390575

RESUMO

INTRODUCTION: Our objective was to determine correlations between the tumor uptake and T/B ratios for 89Zr-labeled T-DM1 (89Zr-DFO-T-DM1) in mice with human BC xenografts by microPET/CT and biodistribution studies with HER2 expression and response to treatment with trastuzumab-DM1 (T-DM1). METHODS: The tumor and normal tissue uptake and T/B ratios for 89Zr-DFO-T-DM1 (10 µg; 7.0 MBq) incorporated into a therapeutic dose (60 µg) were determined by microPET/CT and biodistribution studies at 96 h p.i. in NOD/SCID mice with s.c. MDA-MB-231 (5 × 104 HER2/cell), MDA-MB-361 (5 × 105 HER2/cell) and BT-474 (2 × 106 HER2/cell) human BC xenografts. Mice bearing these tumors were treated with T-DM1 (3.6 mg/kg every 3 weeks) and the tumor doubling time estimated by fitting of tumor volume vs. time curves. A tumor doubling time ratio (TDR) was calculated by dividing the doubling time for T-DM1 and normal saline treated control mice. The clonogenic survival (CS) of BC cells with increasing HER2 expression treated for 72 h in vitro with T-DM1 or trastuzumab (0-100 µg/mL) was compared. Correlations were determined between the T/B ratios for 89Zr-DFO-T-DM1 and HER2 expression, TDR and CS, and between CS and TDR. RESULTS: Uptake of 89Zr-DFO-T-DM1 in MDA-MB-231, MDA-MB-361 and BT-474 tumors was 2.4 ±â€¯0.4%ID/g, 6.9 ±â€¯2.2%ID/g and 9.8 ±â€¯1.1%ID/g, respectively. There was a non-linear but direct correlation between the T/B ratios for 89Zr-DFO-T-DM1 and HER2 expression with the T/B ratio ranging from 4.5 ±â€¯0.7 for MDA-MB-231 to 18.2 ±â€¯1.8 for MDA-MB-361 and 35.9 ±â€¯5.1 for BT-474 xenografts. Tumor intensity on microPET/CT images was proportional to HER2 expression. The standard uptake value (SUV) for the tumors on the images was strongly correlated with the T/B ratio in biodistribution studies. There was a direct linear correlation between the T/B ratio for 89Zr-DFO-T-DM1 and TDR, with TDR ranging from 0.9 for MDA-MB-231 to 1.6 for MDA-MB-361 and 2.1 for BT-474 tumors. The cytotoxicity of T-DM1 in vitro on BC cells was dependent on HER2 expression but T-DM1 was more potent than trastuzumab. There was an inverse correlation between the TDR for mice treated with T-DM1 and CS of BC cells exposed in vitro to T-DM1. CONCLUSIONS: Based on the direct correlations between the T/B ratio for 89Zr-DFO-T-DM1 by PET and HER2 expression and response to T-DM1, our results suggest that PET with 89Zr-DFO-T-DM1 may predict response of HER2-positive BC to treatment with T-DM1. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE: Our results suggest that PET with 89Zr-DFO-T-DM1 may predict response to treatment with T-DM1 in HER-positive BC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Maitansina/análogos & derivados , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Radioisótopos , Receptor ErbB-2/metabolismo , Trastuzumab/metabolismo , Trastuzumab/uso terapêutico , Zircônio , Ado-Trastuzumab Emtansina , Animais , Transporte Biológico , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/terapia , Maitansina/sangue , Maitansina/metabolismo , Maitansina/farmacocinética , Maitansina/uso terapêutico , Camundongos , Distribuição Tecidual , Trastuzumab/sangue , Trastuzumab/farmacocinética , Resultado do Tratamento
20.
Trends Cancer ; 4(10): 662-670, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30292350

RESUMO

For millenia, plants have been a major source of medications against human and animal diseases. In the case of anticancer agents, a significant number of current agents can trace their source back to nominally plant secondary metabolites, with examples being taxol, vinca alkaloids, camptothecin (CPT), and their modified derivatives. However, it is now becoming apparent that these and other plant-derived materials, plus similar agents from marine sources may well have a microbe in their background. In this short Opinion, evidence for such claims are presented for some of the agents currently in use or in preclinical and clinical trials against cancer.


Assuntos
Antineoplásicos/metabolismo , Produtos Biológicos/metabolismo , Endófitos/metabolismo , Neoplasias/tratamento farmacológico , Plantas/microbiologia , Animais , Antineoplásicos/isolamento & purificação , Antineoplásicos/uso terapêutico , Bactérias/metabolismo , Produtos Biológicos/isolamento & purificação , Produtos Biológicos/uso terapêutico , Fungos/metabolismo , Humanos , Maitansina/isolamento & purificação , Maitansina/metabolismo , Maitansina/uso terapêutico , Paclitaxel/isolamento & purificação , Paclitaxel/metabolismo , Paclitaxel/uso terapêutico , Poríferos/microbiologia , Rizosfera , Urocordados/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA