Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Bioconjug Chem ; 35(6): 855-866, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38789102

RESUMO

Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.


Assuntos
Receptores de IgG , Animais , Camundongos , Receptores de IgG/imunologia , Humanos , Polietilenoglicóis/química , Citotoxicidade Celular Dependente de Anticorpos , Fagocitose/efeitos dos fármacos
2.
Mol Cancer Ther ; 20(2): 320-328, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33288628

RESUMO

Auristatins, a class of clinically validated anti-tubulin agents utilized as payloads in antibody-drug conjugates, are generally classified by their membrane permeability and the extent of cytotoxic bystander activity on neighboring cells after targeted delivery. The drugs typically fall within two categories: membrane permeable monomethyl auristatin E-type molecules with high bystander activities and susceptibility to efflux pumps, or charged and less permeable monomethyl auristatin F (MMAF) analogs with low bystander activities and resistance to efflux pumps. Herein, we report the development of novel auristatins that combine the attributes of each class by having both bystander activity and cytotoxicity on multidrug-resistant (MDR+) cell lines. Structure-based design focused on the hydrophobic functionalization of the N-terminal N-methylvaline of the MMAF scaffold to increase cell permeability. The resulting structure-activity relationships of the new auristatins demonstrate that optimization of hydrophobicity and structure can lead to highly active free drugs and antibody-drug conjugates with in vivo bystander activities.


Assuntos
Aminobenzoatos/uso terapêutico , Oligopeptídeos/uso terapêutico , Aminobenzoatos/farmacologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Oligopeptídeos/farmacologia , Ratos , Relação Estrutura-Atividade
3.
Bioconjug Chem ; 19(10): 1960-3, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18803412

RESUMO

Auristatins are highly potent antimitotic agents that have received considerable attention because of their activities when targeted to tumor cells in the form of antibody-drug conjugates (ADCs). Our lead agent, SGN-35, consists of the cAC10 antibody linked to the N-terminal amino acid of monomethylauristatin E (MMAE) via a valine-citrulline p-aminobenzylcarbamate (val-cit-PABC) linker that is cleaved by intracellular proteases such as cathepsin B. More recently, we developed an auristatin F (AF) derivative monomethylauristatin F (MMAF), which unlike MMAE contains the amino acid phenylalanine at the C-terminal position. Because of the negatively charged C-terminal residue, the potency of AF and MMAF is impaired. However, their ability to kill target cells is greatly enhanced through facilitated cellular uptake by internalizing mAbs. Here, we explore the effects of linker technology on AF-based ADC potency, activity, and tolerability by generating a diverse set of dipeptide linkers between the C-terminal residue and the mAb carrier. The resulting ADCs differed widely in activity, with some having significantly improved therapeutic indices compared to the original mAb-Val-Cit-PABC-MMAF conjugate. The therapeutic index was increased yet further by generating dipeptide-based ADCs utilizing new auristatins with methionine or tryptophan as the C-terminal drug residue. These results demonstrate that manipulation of the C-terminal peptide sequence used to attach auristatins to the mAb carrier can lead to highly potent and specific conjugates with greatly improved therapeutic windows.


Assuntos
Anticorpos/metabolismo , Antimitóticos/metabolismo , Imunoconjugados/metabolismo , Imunoconjugados/farmacologia , Oligopeptídeos/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imunoconjugados/toxicidade
4.
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
5.
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
6.
Mol Cancer Ther ; 16(1): 116-123, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28062707

RESUMO

The emergence of antibody-drug conjugates (ADC), such as brentuximab vedotin and ado-trastuzumab emtansine, has led to increased efforts to identify new payloads and develop improved drug-linker technologies. Most antibody payloads impart significant hydrophobicity to the ADC, resulting in accelerated plasma clearance and suboptimal in vivo activity, particularly for conjugates with high drug-to-antibody ratios (DAR). We recently reported on the incorporation of a discrete PEG24 polymer as a side chain in a ß-glucuronidase-cleavable monomethylauristatin E (MMAE) linker to provide homogeneous DAR 8 conjugates with decreased plasma clearance and increased antitumor activity in xenograft models relative to a non-PEGylated control. In this work, we optimized the drug-linker by minimizing the size of the PEG side chain and incorporating a self-stabilizing maleimide to prevent payload de-conjugation in vivo Multiple PEG-glucuronide-MMAE linkers were prepared with PEG size up to 24 ethylene oxide units, and homogeneous DAR 8 ADCs were evaluated. A clear relationship was observed between PEG length and conjugate pharmacology when tested in vivo Longer PEG chains resulted in slower clearance, with a threshold length of PEG8 beyond which clearance was not impacted. Conjugates bearing PEG of sufficient length to minimize plasma clearance provided a wider therapeutic window relative to faster clearing conjugates bearing shorter PEGs. A lead PEGylated glucuronide-MMAE linker was identified incorporating a self-stabilizing maleimide and a PEG12 side chain emerged from these efforts, enabling highly potent, homogeneous DAR 8 conjugates and is under consideration for future ADC programs. Mol Cancer Ther; 16(1); 116-23. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Imunoconjugados/farmacologia , Oligopeptídeos , Polietilenoglicóis , Animais , Anticorpos Monoclonais/química , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estabilidade de Medicamentos , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/química , Imunoconjugados/farmacocinética , Maleimidas/química , Maleimidas/farmacologia , Camundongos , Estrutura Molecular , Oligopeptídeos/química , Polietilenoglicóis/química , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Clin Cancer Res ; 11(2 Pt 1): 843-52, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15701875

RESUMO

Effective antibody-drug conjugates (ADC) combine high drug-linker stability in circulation and efficient intratumoral release of drug. Conjugation of monomethyl auristatin E (MMAE) to the anti-CD30 monoclonal antibody (mAb), cAC10, produced a selective and potent ADC against CD30(+) anaplastic large cell lymphoma and Hodgkin's disease models. This ADC, cAC10-valine-citrulline-MMAE, uses a protease-sensitive dipeptide linker designed to release MMAE by lysosomal cathepsin B in target cells but maintain a stable linkage and attenuate drug potency in circulation. To evaluate ADC stability in vivo, we developed methods for measuring drug/mAb ratios at progressive times in plasma from ADC-treated mice and nonhuman primates. Anti-idiotype mAb permitted the capture and quantitation of mAb cAC10, whereas antidrug mAb and MMAE-conjugated horseradish peroxidase reporter provided quantitative detection of conjugated drug following its in vitro release by cathepsin B. These data were validated by an alternative ELISA using anti-idiotype and anti-MMAE mAbs for capture and detection, respectively. Both methods differentiated ADC with variable levels of drug loading and were subsequently applied to stability studies in severe combined immunodeficient mice and cynomolgus monkeys. Evaluation of ADC from mouse circulation showed the linker half-life to be approximately 144 hours (6.0 days), significantly greater than that reported for disulfide- or hydrazone-linked ADCs in mice or human trials. In cynomolgus monkey, the apparent linker half-life was approximately 230 hours (9.6 days), suggesting that the drug-linker will be highly stable in humans. These data represent the longest reported drug-linker half-life to date and provide the basis for the pronounced specificity and antitumor activity of cAC10-valine-citrulline-MMAE.


Assuntos
Anticorpos Monoclonais/farmacologia , Doença de Hodgkin , Imunoconjugados/uso terapêutico , Antígeno Ki-1/imunologia , Linfoma Anaplásico de Células Grandes , Oligopeptídeos/uso terapêutico , Animais , Anticorpos Anti-Idiotípicos/uso terapêutico , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Dissulfetos , Estabilidade de Medicamentos , Feminino , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/imunologia , Doença de Hodgkin/metabolismo , Hidrazonas , Imunoconjugados/química , Imunoconjugados/metabolismo , Antígeno Ki-1/metabolismo , Linfoma Anaplásico de Células Grandes/tratamento farmacológico , Linfoma Anaplásico de Células Grandes/imunologia , Linfoma Anaplásico de Células Grandes/metabolismo , Macaca fascicularis , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Oligopeptídeos/química , Sensibilidade e Especificidade , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Cancer Res ; 10(23): 7842-51, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15585616

RESUMO

The anti-CD20 antibody rituximab is useful in the treatment of certain B-cell malignancies, most notably non-Hodgkin's lymphoma. Its efficacy has been increased when used in combination with chemotherapy, yet anti-CD20 monoclonal antibodies (mAbs) directly conjugated with drugs such as doxorubicin (Dox) have failed to deliver drug or to demonstrate antitumor activity. We have produced anti-CD20 antibody-drug conjugates that possess potent antitumor activity by using the anti-mitotic agent, monomethyl auristatin E (MMAE), linked via the lysosomally cleavable dipeptide, valine-citrulline (vc). Two anti-CD20 conjugates, rituximab-vcMMAE and 1F5-vcMMAE, were selectively cytotoxic against CD20(+) B-lymphoma cell lines, with IC(50) values ranging from 50 ng/mL to 1 microg/mL. Unlike rituximab, which showed diffuse surface localization, rituximab-vcMMAE capped and was internalized within 4 hours after binding to CD20(+) B cells. Internalization of rituximab-vcMMAE was followed by rapid G(2)-M phase arrest and onset of apoptosis. Anti-CD20 antibody-drug conjugates prepared with Dox were internalized and localized as with rituximab-vcMMAE, yet these were not effective for drug delivery (IC(50) > 50 microg/mL). Consistent with in vitro activity, rituximab-vcMMAE showed antitumor efficacy in xenograft models of CD20-positive lymphoma at doses where rituximab or rituximab-Dox conjugates were ineffective. These data indicate that anti-CD20-based antibody-drug conjugates are effective antitumor agents when prepared with a stable, enzyme-cleavable peptide linkage to highly potent cytotoxic agents such as MMAE.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD20/imunologia , Antineoplásicos/farmacologia , Linfoma de Células B , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Murinos , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Fase G2/efeitos dos fármacos , Humanos , Linfoma de Células B/tratamento farmacológico , Linfoma de Células B/imunologia , Camundongos , Oligopeptídeos/efeitos adversos , Oligopeptídeos/química , Rituximab , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Biotechnol ; 33(7): 733-5, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26076429

RESUMO

The in vitro potency of antibody-drug conjugates (ADCs) increases with the drug-to-antibody ratio (DAR); however, ADC plasma clearance also increases with DAR, reducing exposure and in vivo efficacy. Here we show that accelerated clearance arises from ADC hydrophobicity, which can be modulated through drug-linker design. We exemplify this using hydrophilic auristatin drug linkers and PEGylated ADCs that yield uniform, high-DAR ADCs with superior in vivo performance.


Assuntos
Química Farmacêutica , Imunoconjugados , Preparações Farmacêuticas , Animais , Linhagem Celular , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imunoconjugados/química , Imunoconjugados/farmacocinética , Camundongos , Camundongos SCID , Modelos Químicos , Modelos Moleculares , Preparações Farmacêuticas/química
10.
Nat Biotechnol ; 32(10): 1059-62, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25194818

RESUMO

Many antibody-drug conjugates (ADCs) are unstable in vivo because they are formed from maleimide-containing components conjugated to reactive thiols. These thiosuccinimide linkages undergo two competing reactions in plasma: elimination of the maleimide through a retro-Michael reaction, which results in loss of drug-linker from the ADC, and hydrolysis of the thiosuccinimide ring, which results in a derivative that is resistant to the elimination reaction. In an effort to create linker technologies with improved stability characteristics, we used diaminopropionic acid (DPR) to prepare a drug-linker incorporating a basic amino group adjacent to the maleimide, positioned to provide intramolecular catalysis of thiosuccinimide ring hydrolysis. This basic group induces the thiosuccinimide to undergo rapid hydrolysis at neutral pH and room temperature. Once hydrolyzed, the drug-linker is no longer subject to maleimide elimination reactions, preventing nonspecific deconjugation. In vivo studies demonstrate that the increased stability characteristics can lead to improved ADC antitumor activity and reduced neutropenia.


Assuntos
Anticorpos/química , Antineoplásicos/química , Excipientes/química , Imunoconjugados/química , Maleimidas/química , Animais , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Hidrólise , Imunoconjugados/farmacologia , Camundongos , Camundongos SCID , Plasma , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Bioorg Med Chem Lett ; 16(2): 358-62, 2006 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-16275070

RESUMO

Highly potent and novel derivatives of doxorubicin were linked to monoclonal antibodies (mAbs) for site-specific drug delivery. Drug linker 5 consisted of a dipeptide linker attached directly to the daunosamine nitrogen of the n-butyldiacetate doxorubicin derivative 2a. Upon hydrolysis of the peptide linker and acetate groups, the free daunosamine nitrogen is able to form the highly potent 2-pyrrolinodoxorubicin (3a). The second approach involved the use of an oxazolidine carbamate (13) to mask an activating aldehyde group until proteolytic hydrolysis releases 3a. Both drug linkers were shown to be substrates for the lysosomal enzyme cathepsin B. Each molecule was conjugated to the mAbs c1F6 (anti-CD70) and cAC10 (anti-CD30) to give potent drug conjugates against renal cell carcinoma and anaplastic large cell lymphoma cell lines, respectively. The activities were immunologically selective, since antigen negative cell lines were much less sensitive to treatment with the drug conjugates. The approaches described here for attaching highly potent doxorubicin derivatives to mAbs are novel and allow for control of drug stability while covalently bound to the delivery agent.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Dipeptídeos/química , Doxorrubicina/química , Imunotoxinas/química , Imunotoxinas/farmacologia , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Antígenos de Neoplasias/efeitos dos fármacos , Antígenos de Neoplasias/imunologia , Sítios de Ligação , Linhagem Celular Tumoral , Doxorrubicina/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Humanos , Hidrólise , Conformação Molecular , Relação Estrutura-Atividade
12.
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
13.
J Biol Chem ; 281(15): 10540-7, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16484228

RESUMO

The chimeric anti-CD30 monoclonal antibody cAC10, linked to the antimitotic agents monomethyl auristatin E (MMAE) or F (MMAF), produces potent and highly CD30-selective anti-tumor activity in vitro and in vivo. These drugs are appended via a valine-citrulline (vc) dipeptide linkage designed for high stability in serum and conditional cleavage and putative release of fully active drugs by lysosomal cathepsins. To characterize the biochemical processes leading to effective drug delivery, we examined the intracellular trafficking, internalization, and metabolism of the parent antibody and two antibody-drug conjugates, cAC10vc-MMAE and cAC10vc-MMAF, following CD30 surface antigen interaction with target cells. Both cAC10 and its conjugates bound to target cells and internalized in a similar manner. Subcellular fractionation and immunofluorescence studies demonstrated that the antibody and antibody-drug conjugates entering target cells migrated to the lysosomes. Trafficking of both species was blocked by inhibitors of clathrin-mediated endocytosis, suggesting that drug conjugation does not alter the fate of antibody-antigen complexes. Incubation of cAC10vc-MMAE or cAC10vc-MMAF with purified cathepsin B or with enriched lysosomal fractions prepared by subcellular fractionation resulted in the release of active, free drug. Cysteine protease inhibitors, but not aspartic or serine protease inhibitors, blocked antibody-drug conjugate metabolism and the ensuing cytotoxicity of target cells and yielded enhanced intracellular levels of the intact conjugates. These findings suggest that in addition to trafficking to the lysosomes, cathepsin B and perhaps other lysosomal cysteine proteases are requisite for drug release and provide a mechanistic basis for developing antibody-drug conjugates cleavable by intracellular proteases for the targeted delivery of anti-cancer therapeutics.


Assuntos
Antígeno Ki-1/química , Lisossomos/metabolismo , Oligopeptídeos/química , Anticorpos/química , Antígenos CD20/química , Antineoplásicos/farmacologia , Western Blotting , Catepsina B/química , Linhagem Celular , Cisteína Endopeptidases/química , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Endocitose , Endopeptidases/química , Citometria de Fluxo , Humanos , Concentração Inibidora 50 , Microscopia de Fluorescência , Modelos Químicos , Peptídeo Hidrolases/química , Peptídeos/química , Ligação Proteica , Frações Subcelulares/metabolismo , Temperatura , Fatores de Tempo , beta-Galactosidase/metabolismo
14.
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