Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 110(14): 5404-9, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23493549

RESUMO

The key role played by fucose in glycoprotein and cellular function has prompted significant research toward identifying recombinant and biochemical strategies for blocking its incorporation into proteins and membrane structures. Technologies surrounding engineered cell lines have evolved for the inhibition of in vitro fucosylation, but they are not applicable for in vivo use and drug development. To address this, we screened a panel of fucose analogues and identified 2-fluorofucose and 5-alkynylfucose derivatives that depleted cells of GDP-fucose, the substrate used by fucosyltransferases to incorporate fucose into protein and cellular glycans. The inhibitors were used in vitro to generate fucose-deficient antibodies with enhanced antibody-dependent cellular cytotoxicity activities. When given orally to mice, 2-fluorofucose inhibited fucosylation of endogenously produced antibodies, tumor xenograft membranes, and neutrophil adhesion glycans. We show that oral 2-fluorofucose treatment afforded complete protection from tumor engraftment in a syngeneic tumor vaccine model, inhibited neutrophil extravasation, and delayed the outgrowth of tumor xenografts in immune-deficient mice. The results point to several potential therapeutic applications for molecules that selectively block the endogenous generation of fucosylated glycan structures.


Assuntos
Anticorpos Monoclonais/metabolismo , Vacinas Anticâncer/farmacologia , Fucose/farmacologia , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/metabolismo , Polissacarídeos/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cromatografia Líquida , Cricetinae , Cricetulus , Desenho de Fármacos , Feminino , Fucose/química , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Neutrófilos/metabolismo
2.
Bioconjug Chem ; 24(10): 1650-5, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24050213

RESUMO

The role that carbohydrates play in antibody function and pharmacokinetics has made them important targets for modification. The terminal fucose of the N-linked glycan structure, which has been shown to be involved in modulation of antibody-directed cellular cytotoxicity, is a particularly interesting location for potential modification through incorporation of alternative sugar structures. A library of fucose analogues was evaluated for their ability to incorporate into antibody carbohydrates in place of the native fucose. A number of efficiently incorporated molecules were identified, demonstrating the ability of fucosyltransferase VIII to utilize a variety of non-natural sugars as substrates. Among these structures was a thiolated analogue, 6-thiofucose, which was incorporated into the antibody carbohydrate with good efficiency. This unnatural thio-sugar could then be used for conjugation using maleimide chemistry to produce antibody-drug conjugates with pronounced cytotoxic activities and improved homogeneity compared to drug attachment through hinge disulfides.


Assuntos
Anticorpos Monoclonais/química , Carboidratos/química , Fucose/análogos & derivados , Imunoconjugados/química , Anticorpos Monoclonais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Carboidratos/imunologia , Linhagem Celular , Dissulfetos/química , Fucose/imunologia , Humanos , Imunoconjugados/imunologia , Engenharia Metabólica
3.
Bioconjug Chem ; 20(6): 1242-50, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19469529

RESUMO

Antibody-drug conjugates (ADCs) were prepared with potent camptothecin analogues attached to monoclonal antibodies (mAbs) via dipeptide or glucuronide-based linkers. Aniline-containing camptothecin analogues were employed to provide a site of linker attachment via carbamate bonds that would be stable in circulation. The camptothecin analogues, 7-butyl-10-amino-camptothecin and 7-butyl-9-amino-10,11-methylenedioxy-camptothecin, are generally 10-1000 times more potent than camptothecin. Dipeptide and glucuronide drug linkers were employed containing self-immolative spacers that release drug following lysosomal degradation upon ADC internalization into antigen-positive cell lines. The camptothecin drug linkers were conjugated to three antibodies: chimeric BR96, chimeric AC10, and humanized 1F6, which bind to the Lewis-Y antigen on carcinomas, CD30 on hematologic malignancies, and CD70 present on hematologic malignancies and renal cell carcinoma, respectively. ADCs bearing the potent camptothecin analogue, 7-butyl-9-amino-10,11-methylenedioxy-camptothecin, were highly potent and immunologically specific on a panel of cancer cell lines in vitro, and efficacious at well-tolerated doses in a renal cell carcinoma xenograft model.


Assuntos
Anticorpos Monoclonais/química , Antineoplásicos/química , Antineoplásicos/farmacologia , Camptotecina/química , Camptotecina/farmacologia , Imunoconjugados/química , Imunoconjugados/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Linhagem Celular Tumoral , Desenho de Fármacos , Estabilidade de Medicamentos , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos BALB C
4.
Bioorg Med Chem Lett ; 19(10): 2650-3, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19386499

RESUMO

Cytotoxic agents streptonigrin and 17-amino-geldanamycin were linked to monoclonal antibodies (mAbs), forming antibody-drug conjugates (ADCs) for antigen-mediated targeting to cancer cells. The drugs were conjugated with a linker construct that is labile to lysosomal proteases and incorporates a valine-alanine-p-aminobenzyl (PAB)-amino linkage for direct attachment to the electron-deficient amine functional groups present in both drugs. The resulting ADCs release drug following internalization into antigen-positive cancer cells. The drug linkers were conjugated to mAbs cAC10 (anti-CD30) and h1F6 (anti-CD70) via alkylation of reduced interchain disulfides to give ADCs loaded with 4 drugs/mAb. The streptonigrin ADCs were potent and immunologically specific on a panel of cancer cell lines in vitro and in a Hodgkin lymphoma xenograft model. We conclude that streptonigrin ADCs are candidates for further research, and that the novel linker system used to make them is well-suited for the conjugation of cytotoxic agents containing electron-deficient amine functional groups.


Assuntos
Benzoquinonas/síntese química , Benzilaminas/química , Dipeptídeos/química , Imunoconjugados/química , Lactamas Macrocíclicas/síntese química , Estreptonigrina/síntese química , Animais , Anticorpos Monoclonais/química , Benzoquinonas/química , Benzilaminas/síntese química , Ligante CD27/metabolismo , Linhagem Celular Tumoral , Dipeptídeos/síntese química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoconjugados/farmacologia , Antígeno Ki-1/metabolismo , Lactamas Macrocíclicas/química , Camundongos , Estreptonigrina/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cancer Res ; 66(4): 2328-37, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16489038

RESUMO

Metastatic renal cell carcinoma (RCC) is an aggressive disease refractory to most existing therapeutic modalities. Identifying new markers for disease progression and drug targets for RCC will benefit this unmet medical need. We report a subset of clear cell and papillary cell RCC aberrantly expressing the lymphocyte activation marker CD70, a member of the tumor necrosis factor superfamily. Importantly, CD70 expression was found to be maintained at the metastatic sites of RCC. Anti-CD70 antibody-drug conjugates (ADC) consisting of auristatin phenylalanine phenylenediamine (AFP) or monomethyl auristatin phenylalanine (MMAF), two novel derivatives of the anti-tubulin agent auristatin, mediated potent antigen-dependent cytotoxicity in CD70-expressing RCC cells. Cytotoxic activity of these anti-CD70 ADCs was associated with their internalization and subcellular trafficking through the endosomal-lysosomal pathway, disruption of cellular microtubule network, and G2-M phase cell cycle arrest. The efficiency of drug delivery using anti-CD70 as vehicle was illustrated by the much enhanced cytotoxicity of antibody-conjugated MMAF compared with free MMAF. Hence, ADCs targeted to CD70 can selectively recognize RCC, internalize, and reach the appropriate subcellular compartment(s) for drug release and tumor cell killing. In vitro cytotoxicity of these ADCs was confirmed in xenograft models using RCC cell lines. Our findings provide evidence that CD70 is an attractive target for antibody-based therapeutics against metastatic RCC and suggest that anti-CD70 ADCs can provide a new treatment approach for advanced RCC patients who currently have no chemotherapeutic options.


Assuntos
Antígenos CD/biossíntese , Antineoplásicos/administração & dosagem , Carcinoma de Células Renais/imunologia , Imunoconjugados/imunologia , Imunoconjugados/farmacologia , Neoplasias Renais/imunologia , Proteínas de Membrana/biossíntese , Fatores de Necrose Tumoral/biossíntese , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Apoptose/efeitos dos fármacos , Ligante CD27 , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/patologia , Ciclo Celular/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Ativação Linfocitária , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Nus , Microtúbulos/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Necrose Tumoral/genética , Fatores de Necrose Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Biotechnol ; 21(7): 778-84, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12778055

RESUMO

We describe the in vitro and in vivo properties of monoclonal antibody (mAb)-drug conjugates consisting of the potent synthetic dolastatin 10 analogs auristatin E (AE) and monomethylauristatin E (MMAE), linked to the chimeric mAbs cBR96 (specific to Lewis Y on carcinomas) and cAC10 (specific to CD30 on hematological malignancies). The linkers used for conjugate formation included an acid-labile hydrazone and protease-sensitive dipeptides, leading to uniformly substituted conjugates that efficiently released active drug in the lysosomes of antigen-positive (Ag+) tumor cells. The peptide-linked mAb-valine-citrulline-MMAE and mAb-phenylalanine-lysine-MMAE conjugates were much more stable in buffers and plasma than the conjugates of mAb and the hydrazone of 5-benzoylvaleric acid-AE ester (AEVB). As a result, the mAb-Val-Cit-MMAE conjugates exhibited greater in vitro specificity and lower in vivo toxicity than corresponding hydrazone conjugates. In vivo studies demonstrated that the peptide-linked conjugates induced regressions and cures of established tumor xenografts with therapeutic indices as high as 60-fold. These conjugates illustrate the importance of linker technology, drug potency and conjugation methodology in developing safe and efficacious mAb-drug conjugates for cancer therapy.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Oligopeptídeos/química , Oligopeptídeos/uso terapêutico , Animais , Anticorpos Monoclonais/efeitos adversos , Antineoplásicos/efeitos adversos , Antineoplásicos/síntese química , Células Cultivadas/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Estabilidade de Medicamentos , Humanos , Imunotoxinas/efeitos adversos , Imunotoxinas/química , Imunotoxinas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Transplante de Neoplasias , Oligopeptídeos/efeitos adversos , Oligopeptídeos/síntese química , Ligação Proteica , Resultado do Tratamento
7.
Mol Cancer Ther ; 15(5): 938-45, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26944920

RESUMO

A quaternary ammonium-based drug-linker has been developed to expand the scope of antibody-drug conjugate (ADC) payloads to include tertiary amines, a functional group commonly present in biologically active compounds. The linker strategy was exemplified with a ß-glucuronidase-cleavable auristatin E construct. The drug-linker was found to efficiently release free auristatin E (AE) in the presence of ß-glucuronidase and provide ADCs that were highly stable in plasma. Anti-CD30 conjugates comprised of the glucuronide-AE linker were potent and immunologically specific in vitro and in vivo, displaying pharmacologic properties comparable with a carbamate-linked glucuronide-monomethylauristatin E control. The quaternary ammonium linker was then applied to a tubulysin antimitotic drug that contained an N-terminal tertiary amine that was important for activity. A glucuronide-tubulysin quaternary ammonium linker was synthesized and evaluated as an ADC payload, in which the resulting conjugates were found to be potent and immunologically specific in vitro, and displayed a high level of activity in a Hodgkin lymphoma xenograft. Furthermore, the results were superior to those obtained with a related tubulysin derivative containing a secondary amine N-terminus for conjugation using previously known linker technology. The quaternary ammonium linker represents a significant advance in linker technology, enabling stable conjugation of payloads with tertiary amine residues. Mol Cancer Ther; 15(5); 938-45. ©2016 AACR.


Assuntos
Compostos de Amônio/química , Anticorpos Monoclonais/química , Anticorpos Monoclonais/farmacologia , Imunoconjugados/química , Imunoconjugados/farmacologia , Animais , Anticorpos Monoclonais/farmacocinética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Humanos , Imunoconjugados/farmacocinética , Cinética , Camundongos , Estrutura Molecular , Ligação Proteica , Ratos , Tubulina (Proteína) , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Bioconjug Chem ; 17(1): 114-24, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16417259

RESUMO

We have previously shown that antibody-drug conjugates (ADCs) consisting of cAC10 (anti-CD30) linked to the antimitotic agent monomethylauristatin E (MMAE) lead to potent in vitro and in vivo activities against antigen positive tumor models. MMAF is a new antimitotic auristatin derivative with a charged C-terminal phenylalanine residue that attenuates its cytotoxic activity compared to its uncharged counterpart, MMAE, most likely due to impaired intracellular access. In vitro cytotoxicity studies indicated that mAb-maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-MMAF (mAb-L1-MMAF) conjugates were >2200-fold more potent than free MMAF on a large panel of CD30 positive hematologic cell lines. As with cAC10-L1-MMAE, the corresponding MMAF ADC induced cures and regressions of established xenograft tumors at well tolerated doses. To further optimize the ADC, several new linkers were generated in which various components within the L1 linker were either altered or deleted. One of the most promising linkers contained a noncleavable maleimidocaproyl (L4) spacer between the drug and the mAb. cAC10-L4-MMAF was approximately as potent in vitro as cAC10-L1-MMAF against a large panel of cell lines and was equally potent in vivo. Importantly, cAC10-L4-MMAF was tolerated at >3 times the MTD of cAC10-L1-MMAF. LCMS studies indicated that drug released from cAC10-L4-MMAF was the cysteine-L4-MMAF adduct, which likely arises from mAb degradation within the lysosomes of target cells. This new linker technology appears to be ideally suited for drugs that are both relatively cell-impermeable and tolerant of substitution with amino acids. Thus, alterations of the linker have pronounced impacts on toxicity and lead to new ADCs with greatly improved therapeutic indices.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Imunoconjugados/uso terapêutico , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Oligopeptídeos/uso terapêutico , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Antineoplásicos/administração & dosagem , Antineoplásicos/síntese química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Concentração Inibidora 50 , Linfoma Difuso de Grandes Células B/patologia , Dose Máxima Tolerável , Camundongos , Camundongos SCID , Oligopeptídeos/administração & dosagem , Oligopeptídeos/síntese química , Resultado do Tratamento , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Bioconjug Chem ; 17(2): 410-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16536473

RESUMO

CC49 is a clinically validated antibody with specificity for TAG-72, a carbohydrate epitope that is overexpressed and exposed on the cell surface in a large fraction of solid malignancies. We constructed a single-chain fragment (scFv) based on CC49 and fused it to beta-lactamase (BLA). Following optimization of the scFv domain by combinatorial consensus mutagenesis (CCM) for increased expression and stability, we characterized the protein variant for binding, in vivo pharmacokinetics (PK), and antitumor efficacy. The fusion protein TAB2.5 possessed a similar binding specificity relative to the parent antibody CC49. TAB2.5 also showed prolonged retention (T(1/2) = 36.9 h) in tumor-bearing mice with tumor/plasma ratios of up to 1000. Preliminary evaluation of TAB2.5, in combination with a novel prodrug, GC-Mel, resulted in significant efficacy in a colorectal xenograft tumor model and supports the utility of the protein as an agent for tumor-selective prodrug activation.


Assuntos
Anticorpos Antineoplásicos , Cefalosporinas/química , Cefalosporinas/metabolismo , Cefalosporinas/uso terapêutico , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/metabolismo , Compostos de Mostarda Nitrogenada/uso terapêutico , Pró-Fármacos , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , beta-Lactamases , Animais , Antibióticos Antineoplásicos/uso terapêutico , Anticorpos Antineoplásicos/química , Anticorpos Antineoplásicos/genética , Anticorpos Antineoplásicos/metabolismo , Anticorpos Antineoplásicos/uso terapêutico , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Camptotecina/análogos & derivados , Camptotecina/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Doxorrubicina/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Fragmentos de Imunoglobulinas/genética , Fragmentos de Imunoglobulinas/metabolismo , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/metabolismo , Irinotecano , Melfalan/química , Melfalan/metabolismo , Melfalan/uso terapêutico , Camundongos , Camundongos Nus , Estrutura Molecular , Transplante de Neoplasias , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Pró-Fármacos/uso terapêutico , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , beta-Lactamases/química , beta-Lactamases/genética , beta-Lactamases/metabolismo , beta-Lactamases/uso terapêutico
10.
J Org Chem ; 67(6): 1866-72, 2002 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11895404

RESUMO

A new anticancer prodrug activation strategy based on the 1,6-elimination reaction of p-aminobenzyl ethers is described. Model studies were undertaken with the N-protected peptide benzyloxycarbonyl-valine-citrulline (Z-val-cit), which was attached to the amino groups of p-aminobenzyl ether derivatives of 1-naphthol and N-acetylnorephedrine. The amide bond that formed was designed for hydrolysis by cathepsin B, a protease associated with rapidly growing and metastatic carcinomas. Upon treatment with the enzyme, the Z-val-cit-p-amidobenzyl ether of 1-naphthol (2) underwent peptide bond hydrolysis with the rapid release of 1-naphthol. The aliphatic Z-val-cit-p-amidobenzyl ether of N-acetylnorephedrine (5) also underwent amide bond hydrolysis, but without the ensuing elimination of N-acetylnorephedrine. On the basis of these results, the phenolic anticancer drugs etoposide (6) and combretastatin A-4 (7) were attached to the Z-val-cit-p-amidobenzyl alcohol through ether linkages, forming the peptide-drug derivatives 8 and 9, respectively. Both compounds were stable in aqueous buffers and serum and underwent ether fragmentation upon treatment with cathepsin B, resulting in the release of the parent drugs in chemically unmodified forms. The released drugs were 13-50 times more potent than were the prodrug precursors on a panel of cancer cell lines. In contrast, the corresponding carbonate derivative of combretastatin A-4 (13) was unstable in aqueous environments and was as cytotoxic as combretastatin A-4. This result extends the use of the self-immolative p-aminobenzyl group for the fragmentation of aromatic ethers and provides a new strategy for anticancer prodrug development.


Assuntos
Álcoois Benzílicos/química , Dipeptídeos/química , Endopeptidases/metabolismo , Éteres Fenílicos/química , Pró-Fármacos/química , Antineoplásicos , Catálise , Catepsina B/metabolismo , Cromatografia Líquida de Alta Pressão , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Hidrólise , Neoplasias Pulmonares , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Estrutura Molecular , Naftóis/química , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Blood ; 102(4): 1458-65, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12714494

RESUMO

The chimeric monoclonal antibody cAC10, directed against CD30, induces growth arrest of CD30+ cell lines in vitro and has pronounced antitumor activity in severe combined immunodeficiency (SCID) mouse xenograft models of Hodgkin disease. We have significantly enhanced these activities by conjugating to cAC10 the cytotoxic agent monomethyl auristatin E (MMAE) to create the antibody-drug conjugate cAC10-vcMMAE. MMAE, a derivative of the cytotoxic tubulin modifier auristatin E, was covalently coupled to cAC10 through a valine-citrulline peptide linker. The drug was stably attached to the antibody, showing only a 2% release of MMAE following 10-day incubation in human plasma, but it was readily cleaved by lysosomal proteases after receptor-mediated internalization. Release of MMAE into the cytosol induced G2/M-phase growth arrest and cell death through the induction of apoptosis. In vitro, cAC10-vcMMAE was highly potent and selective against CD30+ tumor lines (IC50 less than 10 ng/mL) but was more than 300-fold less active on antigen-negative cells. In SCID mouse xenograft models of anaplastic large cell lymphoma or Hodgkin disease, cAC10-vcMMAE was efficacious at doses as low as 1 mg/kg. Mice treated at 30 mg/kg cAC10-vcMMAE showed no signs of toxicity. These data indicate that cAC10-vcMMAE may be a highly effective and selective therapy for the treatment of CD30+ neoplasias.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Imunotoxinas/farmacologia , Antígeno Ki-1/imunologia , Oligopeptídeos/farmacologia , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/química , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Brentuximab Vedotin , Ciclo Celular/efeitos dos fármacos , Estabilidade de Medicamentos , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/imunologia , Doença de Hodgkin/metabolismo , Humanos , Imunoconjugados/química , Imunotoxinas/química , Concentração Inibidora 50 , Antígeno Ki-1/metabolismo , Camundongos , Camundongos SCID , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA