Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
MAbs ; 15(1): 2229101, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37639687

RESUMO

The antibody-drug conjugate (ADC) field has undergone a renaissance, with substantial recent developmental investment and subsequent drug approvals over the past 6 y. In November 2022, ElahereTM became the latest ADC to be approved by the US Food and Drug Administration (FDA). To date, over 260 ADCs have been tested in the clinic against various oncology indications. Here, we review the clinical landscape of ADCs that are currently FDA approved (11), agents currently in clinical trials but not yet approved (164), and candidates discontinued following clinical testing (92). These clinically tested ADCs are further analyzed by their targeting tumor antigen(s), linker, payload choices, and highest clinical stage achieved, highlighting limitations associated with the discontinued drug candidates. Lastly, we discuss biologic engineering modifications preclinically demonstrated to improve the therapeutic index that if incorporated may increase the proportion of molecules that successfully transition to regulatory approval.


Assuntos
Antineoplásicos , Imunoconjugados , Estados Unidos , Imunoconjugados/uso terapêutico , Anticorpos Monoclonais , United States Food and Drug Administration
2.
MAbs ; 14(1): 2115213, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36206404

RESUMO

T cell-engaging bispecific antibodies (TCEs) are clinically effective treatments for hematological cancers. While the utility of TCEs in solid malignancies is being explored, toxicities arising from antigen expression on normal tissues have slowed or halted several clinical trials. Here, we describe the development of TCEs that preferentially drive T cell-mediated death against target cells co-expressing two tumor-associated antigens. We show that Ly6E and B7-H4 are simultaneously expressed on approximately 50% of breast cancers, whereas normal tissue expression is limited and mostly orthogonal. Traditional bispecific TCEs targeting a singular antigen, either Ly6E or B7-H4, are active when paired with high-affinity CD3-engagers, but normal tissue expression presents a toxicity risk. Treatment with a murine cross-reactive B7-H4-TCE results in rapid and severe weight loss in mice along with damage to B7-H4-expressing tissues. To overcome on-target toxicity, we designed trispecific antibodies co-targeting Ly6E, B7-H4, and CD3 and characterized the impact of dual-antigen binding and the relative placement of each binding domain on tumor killing in vitro and in vivo. In vitro killing of tumor cells co-expressing both antigens correlates to the placement of the higher affinity B7-H4 binding domain, with only modest enhancements seen upon addition of Ly6E binding. In xenograft models, avid binding of appropriately designed trispecific TCEs enables tumor growth inhibition while evading the poor tolerability seen with active bispecific TCEs. Collectively these data highlight the potential for dual-antigen targeting to improve safety and efficacy, and expand the scope of tumors that may effectively be treated by TCEs.Abbreviations: Chimeric antigen receptor T cells (CAR-Ts), dual-antigen targeted T cell engagers (DAT-TCE), Fragment antigen-binding (Fab), Hematoxylin and eosin (H&E), Institutional Animal Care and Use Committee (IACUC), Immunoglobulin G (IgG), immunohistochemistry (IHC), NOD SCID gamma (NSG), peripheral blood mononuclear cells (PBMCs), surface plasmon resonance (SPR), T cell-engagers (TCEs).


Assuntos
Anticorpos Biespecíficos , Receptores de Antígenos Quiméricos , Animais , Anticorpos Biespecíficos/farmacologia , Linhagem Celular Tumoral , Amarelo de Eosina-(YS) , Hematoxilina , Humanos , Imunoglobulina G , Leucócitos Mononucleares , Camundongos , Camundongos SCID , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Med Chem ; 64(5): 2534-2575, 2021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33596065

RESUMO

The biological and medicinal impacts of proteolysis-targeting chimeras (PROTACs) and related chimeric molecules that effect intracellular degradation of target proteins via ubiquitin ligase-mediated ubiquitination continue to grow. However, these chimeric entities are relatively large compounds that often possess molecular characteristics, which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. We therefore explored the conjugation of such molecules to monoclonal antibodies using technologies originally developed for cytotoxic payloads so as to provide alternate delivery options for these novel agents. In this report, we describe the first phase of our systematic development of antibody-drug conjugates (ADCs) derived from bromodomain-containing protein 4 (BRD4)-targeting chimeric degrader entities. We demonstrate the antigen-dependent delivery of the degrader payloads to PC3-S1 prostate cancer cells along with related impacts on MYC transcription and intracellular BRD4 levels. These experiments culminate with the identification of one degrader conjugate, which exhibits antigen-dependent antiproliferation effects in LNCaP prostate cancer cells.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Dipeptídeos/farmacologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Imunoconjugados/farmacologia , Proteólise/efeitos dos fármacos , Fatores de Transcrição/antagonistas & inibidores , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/síntese química , Dipeptídeos/farmacocinética , Compostos Heterocíclicos com 3 Anéis/síntese química , Compostos Heterocíclicos com 3 Anéis/farmacocinética , Humanos , Imunoconjugados/química , Imunoconjugados/imunologia , Oxirredutases/imunologia , Células PC-3 , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
5.
Bioconjug Chem ; 29(2): 473-485, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29425028

RESUMO

THIOMAB antibody technology utilizes cysteine residues engineered onto an antibody to allow for site-specific conjugation. The technology has enabled the exploration of different attachment sites on the antibody in combination with small molecules, peptides, or proteins to yield antibody conjugates with unique properties. As reported previously ( Shen , B. Q. , et al. ( 2012 ) Nat. Biotechnol. 30 , 184 - 189 ; Pillow , T. H. , et al. ( 2017 ) Chem. Sci. 8 , 366 - 370 ), the specific location of the site of conjugation on an antibody can impact the stability of the linkage to the engineered cysteine for both thio-succinimide and disulfide bonds. High stability of the linkage is usually desired to maximize the delivery of the cargo to the intended target. In the current study, cysteines were individually substituted into every position of the anti-HER2 antibody (trastuzumab), and the stabilities of drug conjugations at those sites were evaluated. We screened a total of 648 THIOMAB antibody-drug conjugates, each generated from a trastuzamab prepared by sequentially mutating non-cysteine amino acids in the light and heavy chains to cysteine. Each THIOMAB antibody variant was conjugated to either maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E (MC-vc-PAB-MMAE) or pyridyl disulfide monomethyl auristatin E (PDS-MMAE) using a high-throughput, on-bead conjugation and purification method. Greater than 50% of the THIOMAB antibody variants were successfully conjugated to both MMAE derivatives with a drug to antibody ratio (DAR) of >0.5 and <50% aggregation. The relative in vitro plasma stabilities for approximately 750 conjugates were assessed using enzyme-linked immunosorbent assays, and stable sites were confirmed with affinity-capture LC/MS-based detection methods. Highly stable conjugation sites for the two types of MMAE derivatives were identified on both the heavy and light chains. Although the stabilities of maleimide conjugates were shown to be greater than those of the disulfide conjugates, many sites were identified that were stable for both. Furthermore, in vitro stabilities of selected stable sites translated across different cytotoxic payloads and different target antibodies as well as to in vivo stability.


Assuntos
Antineoplásicos Imunológicos/química , Cisteína/química , Dissulfetos/química , Imunoconjugados/química , Maleimidas/química , Trastuzumab/química , Animais , Antineoplásicos Imunológicos/sangue , Cisteína/sangue , Cisteína/genética , Dissulfetos/sangue , Estabilidade de Medicamentos , Ensaios de Triagem em Larga Escala , Humanos , Imunoconjugados/sangue , Maleimidas/sangue , Modelos Moleculares , Mutagênese Sítio-Dirigida , Oligopeptídeos/sangue , Oligopeptídeos/química , Agregados Proteicos , Estabilidade Proteica , Ratos , Trastuzumab/sangue , Trastuzumab/genética
6.
Mol Cancer Ther ; 17(3): 638-649, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29282299

RESUMO

Luminal A (hormone receptor-positive) breast cancer constitutes 70% of total breast cancer patients. In an attempt to develop a targeted therapeutic for this cancer indication, we have identified and characterized Glial cell line-Derived Neurotrophic Factor (GDNF) Family Receptor Alpha 1 (GFRA1) antibody-drug conjugates (ADC) using a cleavable valine-citrulline-MMAE (vcMMAE) linker-payload. RNAseq and IHC analysis confirmed the abundant expression of GFRA1 in luminal A breast cancer tissues, whereas minimal or no expression was observed in most normal tissues. Anti-GFRA-vcMMAE ADC internalized to the lysosomes and exhibited target-dependent killing of GFRA1-expressing cells both in vitro and in vivo The ADCs using humanized anti-GFRA1 antibodies displayed robust therapeutic activity in clinically relevant cell line-derived (MCF7 and KPL-1) tumor xenograft models. The lead anti-GFRA1 ADC cross-reacts with rodent and cynomolgus monkey GFRA1 antigen and showed optimal pharmacokinetic properties in both species. These properties subsequently enabled a target-dependent toxicity study in rats. Anti-GFRA1 ADC is well tolerated in rats, as seen with other vcMMAE linker-payload based ADCs. Overall, these data suggest that anti-GFRA1-vcMMAE ADC may provide a targeted therapeutic opportunity for luminal A breast cancer patients. Mol Cancer Ther; 17(3); 638-49. ©2017 AACR.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/antagonistas & inibidores , Imunoconjugados/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Anticorpos/química , Anticorpos/imunologia , Anticorpos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/imunologia , Células HEK293 , Humanos , Imunoconjugados/imunologia , Imunoconjugados/farmacocinética , Células MCF-7 , Macaca fascicularis , Camundongos Nus , Camundongos SCID , Ratos Sprague-Dawley , Receptores de Esteroides/metabolismo , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética
7.
Chem Sci ; 8(1): 366-370, 2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-28451181

RESUMO

Disulfide bonds provide a bioactivatable connection with applications in imaging and therapy. The circulation stability and intracellular release of disulfides are problematically coupled in that increasing stability causes a corresponding decrease in cleavage and payload release. However, an antibody offers the potential for a reversible stabilization. We examined this by attaching a small molecule directly to engineered cysteines in an antibody. At certain sites this unhindered disulfide was stable in circulation yet cellular internalization and antibody catabolism generated a disulfide catabolite that was rapidly reduced. We demonstrated that this stable connection and facile release is applicable to a variety of payloads. The ability to reversibly stabilize a labile functional group with an antibody may offer a way to improve targeted probes and therapeutics.

8.
MAbs ; 9(3): 430-437, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28125314

RESUMO

Bispecific antibodies are a growing class of therapeutic molecules. Many of the current bispecific formats require DNA engineering to convert the parental monoclonal antibodies into the final bispecific molecules. We describe here a method to generate bispecific molecules from hybridoma IgGs in 3-4 d using chemical conjugation of antigen-binding fragments (Fabs) (bisFabs). Proteolytic digestion conditions for each IgG isotype were analyzed to optimize the yield and quality of the final conjugates. The resulting bisFabs showed no significant amounts of homodimers or aggregates. The predictive value of murine bisFabs was tested by comparing the T-cell redirected cytotoxic activity of a panel of antibodies in either the bisFab or full-length IgG formats. A variety of antigens with different structures and expression levels was used to extend the comparison to a wide range of binding geometries and antigen densities. The activity observed for different murine bisFabs correlated with those observed for the full-length IgG format across multiple different antigen targets, supporting the use of bisFabs as a screening tool. Our method may also be used for the screening of bispecific antibodies with other mechanisms of action, allowing for a more rapid selection of lead therapeutic candidates.


Assuntos
Anticorpos Biespecíficos/biossíntese , Fragmentos Fab das Imunoglobulinas/biossíntese , Imunoglobulina G/isolamento & purificação , Engenharia de Proteínas/métodos , Animais , Anticorpos Biespecíficos/imunologia , Anticorpos Biespecíficos/isolamento & purificação , Humanos , Hibridomas , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Imunoglobulina G/imunologia , Camundongos
9.
Pharm Res ; 32(6): 1884-93, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25446772

RESUMO

PURPOSE: THIOMAB™ drug conjugates (TDCs) with engineered cysteine residues allow site-specific drug conjugation and defined Drug-to-Antibody Ratios (DAR). In order to help elucidate the impact of drug-loading, conjugation site, and subsequent deconjugation on pharmacokinetics and efficacy, we have developed an integrated mathematical model to mechanistically characterize pharmacokinetic behavior and preclinical efficacy of MMAE conjugated TDCs with different DARs. General applicability of the model structure was evaluated with two different TDCs. METHOD: Pharmacokinetics studies were conducted for unconjugated antibody and purified TDCs with DAR-1, 2 and 4 for trastuzumab TDC and Anti-STEAP1 TDC in mice. Total antibody concentrations and individual DAR fractions were measured. Efficacy studies were performed in tumor-bearing mice. RESULTS: An integrated model consisting of distinct DAR species (DAR0-4), each described by a two-compartment model was able to capture the experimental data well. Time series measurements of each Individual DAR species allowed for the incorporation of site-specific drug loss through deconjugation and the results suggest a higher deconjugation rate from heavy chain site HC-A114C than the light chain site LC-V205C. Total antibody concentrations showed multi-exponential decline, with a higher clearance associated with higher DAR species. The experimentally observed effects of TDC on tumor growth kinetics were successfully described by linking pharmacokinetic profiles to DAR-dependent killing of tumor cells. CONCLUSION: Results from the integrated model evaluated with two different TDCs highlight the impact of DAR and site of conjugation on pharmacokinetics and efficacy. The model can be used to guide future drug optimization and in-vivo studies.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antineoplásicos/farmacocinética , Modelos Biológicos , Compostos de Sulfidrila/farmacocinética , Trastuzumab/metabolismo , Administração Intravenosa , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Antígenos de Neoplasias/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Disponibilidade Biológica , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Cisteína , Feminino , Masculino , Taxa de Depuração Metabólica , Camundongos Nus , Camundongos SCID , Transplante de Neoplasias , Compostos de Sulfidrila/administração & dosagem , Compostos de Sulfidrila/química , Trastuzumab/administração & dosagem , Trastuzumab/química
10.
J Med Chem ; 57(19): 7890-9, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25191794

RESUMO

Antibody-drug conjugates (ADCs) have a significant impact toward the treatment of cancer, as evidenced by the clinical activity of the recently approved ADCs, brentuximab vedotin for Hodgkin lymphoma and ado-trastuzumab emtansine (trastuzumab-MCC-DM1) for metastatic HER2+ breast cancer. DM1 is an analog of the natural product maytansine, a microtubule inhibitor that by itself has limited clinical activity and high systemic toxicity. However, by conjugation of DM1 to trastuzumab, the safety was improved and clinical activity was demonstrated. Here, we report that through chemical modification of the linker-drug and antibody engineering, the therapeutic activity of trastuzumab maytansinoid ADCs can be further improved. These improvements include eliminating DM1 release in the plasma and increasing the drug load by engineering four cysteine residues into the antibody. The chemical synthesis of highly stable linker-drugs and the modification of cysteine residues of engineered site-specific antibodies resulted in a homogeneous ADC with increased therapeutic activity compared to the clinically approved ADC, trastuzumab-MCC-DM1.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/síntese química , Imunoconjugados/farmacologia , Maitansina/análogos & derivados , Engenharia de Proteínas , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Trastuzumab
11.
J Biotechnol ; 180: 10-6, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24704608

RESUMO

Transient transfection of mammalian cells provides a rapid method of producing protein for research purposes. Combining the transient transfection protein expression system with new automation technologies developed for the biotechnology industry would enable a high throughput protein production platform that could be utilized to generate a variety of different proteins in a short amount of time. These proteins could be used for an assortment of studies including proof of concept, antibody development, and biological structure and function. Here we describe such a platform: a semi-automated process for PEI-mediated transient protein production in tubespins at a throughput of 96 transfections at a time using a Biomek FX(P) liquid handling system. In one batch, 96 different proteins can be produced in milligram amounts by PEI transfection of HEK293 cells cultured in 50 mL tubespins. Methods were developed for the liquid handling system to automate the different processes associated with transient transfections such as initial cell seeding, DNA:PEI complex activation and DNA:PEI complex addition to the cells. Increasing DNA:PEI complex incubation time resulted in lower protein expression. To minimize protein production variability, the methods were further optimized to achieve consistent cell seeding, control the DNA:PEI incubation time and prevent cross-contamination among different tubespins. This semi-automated transfection process was applied to express 520 variants of a human IgG1 (hu IgG1) antibody.


Assuntos
Ensaios de Triagem em Larga Escala/instrumentação , Ensaios de Triagem em Larga Escala/métodos , Transfecção/instrumentação , Transfecção/métodos , Automação Laboratorial , DNA/química , Células HEK293 , Humanos , Iminas/química , Imunoglobulina G/análise , Imunoglobulina G/química , Polietilenos/química , Robótica
12.
MAbs ; 6(1): 34-45, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24423619

RESUMO

Antibody therapeutics have revolutionized the treatment of cancer over the past two decades. Antibodies that specifically bind tumor surface antigens can be effective therapeutics; however, many unmodified antibodies lack therapeutic activity. These antibodies can instead be applied successfully as guided missiles to deliver potent cytotoxic drugs in the form of antibody drug conjugates (ADCs). The success of ADCs is dependent on four factors--target antigen, antibody, linker, and payload. The field has made great progress in these areas, marked by the recent approval by the US Food and Drug Administration of two ADCs, brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). However, the therapeutic window for many ADCs that are currently in pre-clinical or clinical development remains narrow and further improvements may be required to enhance the therapeutic potential of these ADCs. Production of ADCs is an area where improvement is needed because current methods yield heterogeneous mixtures that may include 0-8 drug species per antibody molecule. Site-specific conjugation has been recently shown to eliminate heterogeneity, improve conjugate stability, and increase the therapeutic window. Here, we review and describe various site-specific conjugation strategies that are currently used for the production of ADCs, including use of engineered cysteine residues, unnatural amino acids, and enzymatic conjugation through glycotransferases and transglutaminases. In addition, we also summarize differences among these methods and highlight critical considerations when building next-generation ADC therapeutics.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Especificidade de Anticorpos , Sistemas de Liberação de Medicamentos , Imunoconjugados/uso terapêutico , Maitansina/análogos & derivados , Neoplasias/tratamento farmacológico , Ado-Trastuzumab Emtansina , Brentuximab Vedotin , Humanos , Maitansina/uso terapêutico , Trastuzumab , Estados Unidos , United States Food and Drug Administration
13.
MAbs ; 6(1): 95-107, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24121517

RESUMO

Multi-transmembrane proteins are especially difficult targets for antibody generation largely due to the challenge of producing a protein that maintains its native conformation in the absence of a stabilizing membrane. Here, we describe an immunization strategy that successfully resulted in the identification of monoclonal antibodies that bind specifically to extracellular epitopes of a 12 transmembrane protein, multi-drug resistant protein 4 (MRP4). These monoclonal antibodies were developed following hydrodynamic tail vein immunization with a cytomegalovirus (CMV) promoter-based plasmid expressing MRP4 cDNA and were characterized by flow cytometry. As expected, the use of the immune modulators fetal liver tyrosine kinase 3 ligand (Flt3L) and granulocyte-macrophage colony-stimulating factor positively enhanced the immune response against MRP4. Imaging studies using CMV-based plasmids expressing luciferase showed that the in vivo half-life of the target antigen was less than 48 h using CMV-based plasmids, thus necessitating frequent boosting with DNA to achieve an adequate immune response. We also describe a comparison of plasmids, which contained MRP4 cDNA with either the CMV or CAG promoters, used for immunizations. The observed luciferase activity in this comparison demonstrated that the CAG promoter-containing plasmid pCAGGS induced prolonged constitutive expression of MRP4 and an increased anti-MRP4 specific immune response even when the plasmid was injected less frequently. The method described here is one that can be broadly applicable as a general immunization strategy to develop antibodies against multi-transmembrane proteins, as well as target antigens that are difficult to express or purify in native and functionally active conformation.


Assuntos
Anticorpos/imunologia , Imunização , Proteínas Associadas à Resistência a Múltiplos Medicamentos/imunologia , Plasmídeos , Vacinas de DNA , Animais , Linhagem Celular , DNA Complementar/imunologia , DNA Complementar/farmacologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Plasmídeos/imunologia , Plasmídeos/farmacologia , Estrutura Secundária de Proteína , Vacinas de DNA/imunologia , Vacinas de DNA/farmacologia
14.
Methods Mol Biol ; 1045: 189-203, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23913148

RESUMO

Antibody conjugates are used in many therapeutic and research applications and are generated by chemically linking a cysteine or lysine residue to potent chemotherapeutic drugs or other functional groups through a flexible linker. Recently, we have engineered THIOMABs (antibodies with engineered reactive cysteine residues) for site-specific conjugation and showed that these antibody conjugates display homogeneous labeling with optimal in vitro and in vivo characteristics. Here, we describe protocols for engineering, selection, and site-specific conjugation of THIOMABs with thiol-reactive linkers.


Assuntos
Cisteína/química , Imunoconjugados/química , Imunoconjugados/genética , Engenharia de Proteínas/métodos , Sítios de Ligação , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida , Especificidade por Substrato
15.
Nat Biotechnol ; 30(2): 184-9, 2012 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-22267010

RESUMO

The reactive thiol in cysteine is used for coupling maleimide linkers in the generation of antibody conjugates. To assess the impact of the conjugation site, we engineered cysteines into a therapeutic HER2/neu antibody at three sites differing in solvent accessibility and local charge. The highly solvent-accessible site rapidly lost conjugated thiol-reactive linkers in plasma owing to maleimide exchange with reactive thiols in albumin, free cysteine or glutathione. In contrast, a partially accessible site with a positively charged environment promoted hydrolysis of the succinimide ring in the linker, thereby preventing this exchange reaction. The site with partial solvent-accessibility and neutral charge displayed both properties. In a mouse mammary tumor model, the stability and therapeutic activity of the antibody conjugate were affected positively by succinimide ring hydrolysis and negatively by maleimide exchange with thiol-reactive constituents in plasma. Thus, the chemical and structural dynamics of the conjugation site can influence antibody conjugate performance by modulating the stability of the antibody-linker interface.


Assuntos
Anticorpos/sangue , Anticorpos/imunologia , Sítios de Ligação de Anticorpos/imunologia , Imunoconjugados/química , Imunoconjugados/imunologia , Imunoglobulina G/química , Engenharia de Proteínas , Aminobenzoatos/química , Aminobenzoatos/imunologia , Animais , Anticorpos/química , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cisteína/química , Humanos , Imunoconjugados/administração & dosagem , Imunoglobulina G/imunologia , Macaca fascicularis , Maleimidas/química , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/imunologia , Maitansina/química , Maitansina/imunologia , Camundongos , Camundongos Nus , Modelos Moleculares , Oligopeptídeos/química , Oligopeptídeos/imunologia , Conformação Proteica , Ratos , Relação Estrutura-Atividade , Compostos de Sulfidrila/química , Trastuzumab
16.
Clin Cancer Res ; 16(19): 4769-78, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20805300

RESUMO

PURPOSE: Antibody drug conjugates (ADCs) combine the ideal properties of both antibodies and cytotoxic drugs by targeting potent drugs to the antigen-expressing tumor cells, thereby enhancing their antitumor activity. Successful ADC development for a given target antigen depends on optimization of antibody selection, linker stability, cytotoxic drug potency, and mode of linker-drug conjugation to the antibody. Here, we systematically examined the in vitro potency as well as in vivo preclinical efficacy and safety profiles of a heterogeneous preparation of conventional trastuzumab-mcc-DM1 (TMAb-mcc-DM1) ADC with that of a homogeneous engineered thio-trastuzumab-mpeo-DM1 (thioTMAb-mpeo-DM1) conjugate. EXPERIMENTAL DESIGN AND RESULTS: To generate thioTMAb-mpeo-DM1, one drug maytansinoid 1 (DM1) molecule was conjugated to an engineered cysteine residue at Ala114 (Kabat numbering) on each trastuzumab-heavy chain, resulting in two DM1 molecules per antibody. ThioTMAb-mpeo-DM1 retained similar in vitro anti-cell proliferation activity and human epidermal growth factor receptor 2 (HER2) binding properties to that of the conventional ADC. Furthermore, it showed improved efficacy over the conventional ADC at DM1-equivalent doses (µg/m(2)) and retained efficacy at equivalent antibody doses (mg/kg). An improved safety profile of >2-fold was observed in a short-term target-independent rat safety study. In cynomolgus monkey safety studies, thioTMAb-mpeo-DM1 was tolerated at higher antibody doses (up to 48 mg/kg or 6,000 µg DM1/m(2)) compared with the conventional ADC that had dose-limiting toxicity at 30 mg/kg (6,000 µg DM1/m(2)). CONCLUSIONS: The engineered thioTMAb-mpeo-DM1 with broadened therapeutic index represents a promising antibody drug conjugate for future clinical development of HER2-positive targeted breast cancer therapies.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Maitansina/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Compostos de Sulfidrila/farmacologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais Humanizados , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Feminino , Humanos , Macaca fascicularis , Maitansina/química , Camundongos , Ratos , Ratos Sprague-Dawley , Receptor ErbB-2/química , Compostos de Sulfidrila/química , Trastuzumab
17.
Nat Biotechnol ; 26(8): 925-32, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18641636

RESUMO

Antibody-drug conjugates enhance the antitumor effects of antibodies and reduce adverse systemic effects of potent cytotoxic drugs. However, conventional drug conjugation strategies yield heterogenous conjugates with relatively narrow therapeutic index (maximum tolerated dose/curative dose). Using leads from our previously described phage display-based method to predict suitable conjugation sites, we engineered cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not perturb immunoglobulin folding and assembly, or alter antigen binding. When conjugated to monomethyl auristatin E, an antibody against the ovarian cancer antigen MUC16 is as efficacious as a conventional conjugate in mouse xenograft models. Moreover, it is tolerated at higher doses in rats and cynomolgus monkeys than the same conjugate prepared by conventional approaches. The favorable in vivo properties of the near-homogenous composition of this conjugate suggest that our strategy offers a general approach to retaining the antitumor efficacy of antibody-drug conjugates, while minimizing their systemic toxicity.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Citotoxinas/farmacologia , Imunotoxinas/farmacocinética , Neoplasias Ovarianas/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Antineoplásicos/genética , Especificidade de Anticorpos , Sítios de Ligação , Antígeno Ca-125/imunologia , Linhagem Celular Tumoral , Química Farmacêutica/métodos , Cisteína/genética , Feminino , Humanos , Macaca fascicularis , Proteínas de Membrana/imunologia , Camundongos , Mutagênese Sítio-Dirigida , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Compostos de Sulfidrila/farmacologia
18.
J Immunol Methods ; 332(1-2): 41-52, 2008 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-18230399

RESUMO

Cysteines with reactive thiol groups are attractive tools for site-specific labeling of proteins. Engineering a reactive cysteine residue into proteins with multiple disulfide bonds is often a challenging task as it may interfere with structural and functional properties of the protein. Here we developed a phage display-based biochemical assay, PHESELECTOR (Phage ELISA for Selection of Reactive Thiols) to rapidly screen reactive thiol groups on antibody fragments without interfering with their antigen binding, using trastuzumab-Fab (hu4D5Fab) as a model system. The solvent accessibility values for all the amino acid residues in the hu4D5Fab were calculated using available crystal structure information. Serine, alanine and valine residues with highest solvent accessibility values were selected and tested to compare structure-based design with the PHESELECTOR biochemical method. Cysteine substitutions at partially solvent-accessible alanine or valine residues exhibited better thiol reactivity values than substitutions at serine residues. The poor correlation between fractional solvent accessibility and thiol reactivity of the engineered hu4D5Fab variants indicated the value of PHESELECTOR biochemical assay to identify reactive thiol groups on the antibody-Fab surface. Mass spectrometric analysis of biotinylated ThioFab (Fab with engineered cysteine) variants confirmed that conjugation occurred only at the engineered cysteine thiols of either light or heavy chains. ThioFabs with engineered cysteine residues in the constant domains (CL and CH(1)) should allow universal application for site-specific conjugation of antibody-Fabs.


Assuntos
Anticorpos Monoclonais/química , Cisteína/química , Fragmentos Fab das Imunoglobulinas/química , Anticorpos Monoclonais/imunologia , Reações Antígeno-Anticorpo , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Biotina/química , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Cisteína/genética , Cisteína/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo/métodos , Regulação da Expressão Gênica/genética , Engenharia Genética , Variação Genética , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Maleimidas/química , Espectrometria de Massas/métodos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Plasmídeos/genética , Sensibilidade e Especificidade , Coloração e Rotulagem/métodos , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA