Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Bioconjug Chem ; 32(4): 746-754, 2021 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-33689309

RESUMEN

Although peptide motifs represent the majority of cleavable linkers used in clinical-stage antibody-drug conjugates (ADCs), the sequences are often sensitive to cleavage by extracellular enzymes, such as elastase, which leads to systemic release of the cytotoxic payload. This action reduces the therapeutic index by causing off-target toxicities that can be dose-limiting. For example, a common side-effect of ADCs made using peptide-cleavable linkers is myelosuppression, including neutropenia. Only a few reports describe methods for optimizing peptide linkers to maintain efficient and potent tumor payload delivery while enhancing circulating stability. Herein, we address these critical limitations through the development of a tandem-cleavage linker strategy, where two sequential enzymatic cleavage events mediate payload release. We prepared dipeptides that are protected from degradation in the circulation by a sterically encumbering glucuronide moiety. Upon ADC internalization and lysosomal degradation, the monosaccharide is removed and the exposed dipeptide is degraded, which liberates the attached payload inside the target cell. We used CD79b-targeted monomethyl auristatin E (MMAE) conjugates as our model system and compared the stability, efficacy, and tolerability of ADCs made with tandem-cleavage linkers to ADCs made using standard technology with the vedotin linker. The results, where rat studies showed dramatically improved tolerability in the hematopoietic compartment, highlight the role that linker stability plays in efficacy and tolerability and also offer a means of improving an ADC's therapeutic index for improved patient outcomes.


Asunto(s)
Antineoplásicos/toxicidad , Antígenos CD79/toxicidad , Inmunoconjugados/toxicidad , Animales , Antineoplásicos/química , Antígenos CD79/química , Endocitosis , Femenino , Hidrólisis , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nat Chem Biol ; 15(10): 949-958, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451760

RESUMEN

Antibody-drug conjugates (ADCs) selectively deliver chemotherapeutic agents to target cells and are important cancer therapeutics. However, the mechanisms by which ADCs are internalized and activated remain unclear. Using CRISPR-Cas9 screens, we uncover many known and novel endolysosomal regulators as modulators of ADC toxicity. We identify and characterize C18ORF8/RMC1 as a regulator of ADC toxicity through its role in endosomal maturation. Through comparative analysis of screens with ADCs bearing different linkers, we show that a subset of late endolysosomal regulators selectively influence toxicity of noncleavable linker ADCs. Surprisingly, we find cleavable valine-citrulline linkers can be processed rapidly after internalization without lysosomal delivery. Lastly, we show that sialic acid depletion enhances ADC lysosomal delivery and killing in diverse cancer cell types, including with FDA (US Food and Drug Administration)-approved trastuzumab emtansine (T-DM1) in Her2-positive breast cancer cells. Together, these results reveal new regulators of endolysosomal trafficking, provide important insights for ADC design and identify candidate combination therapy targets.


Asunto(s)
Sistemas CRISPR-Cas , Estudio de Asociación del Genoma Completo , Inmunoconjugados/toxicidad , Maitansina/análogos & derivados , Ácido N-Acetilneuramínico/farmacología , Trastuzumab/farmacología , Ado-Trastuzumab Emtansina , Antineoplásicos Inmunológicos/farmacología , Proteínas Portadoras , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Lisosomas , Maitansina/farmacología
3.
Bioconjug Chem ; 30(11): 2982-2988, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31671265

RESUMEN

Antibody-drug conjugates (ADCs) are an established modality for the tissue-specific delivery of chemotherapeutics. However, due to the hydrophobic nature of many cytotoxic payloads, challenges remain in developing chemically stable ADCs with high drug loading. In previous studies, payload structure, unique stimuli-responsive chemistries, and PEGylated cross-linkers have been used to decrease ADC hydrophobicity. In this work, we investigate the effect of a new parameter, cross-linker sequence. A support-free synthesis of PEGylated, sequence-defined cross-linkers was developed and applied to the synthesis of three constitutionally isomeric ADCs containing PEG side chains and a monomethyl auristatin E payload. Placement of PEG side chains distally from the payload was found to yield an ADC with altered hydrophilicity, antigen binding, and in vitro potency. This work establishes a versatile method for synthesizing multifunctional cross-linkers and identifies cross-linker sequence as a new handle for modulating the performance of ADCs.


Asunto(s)
Anticuerpos Monoclonales/química , Proliferación Celular , Reactivos de Enlaces Cruzados/química , Interacciones Hidrofóbicas e Hidrofílicas , Inmunoconjugados/química , Inmunoconjugados/farmacología , Oligopéptidos/química , Femenino , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/inmunología , Células Tumorales Cultivadas
4.
J Biol Chem ; 290(25): 15730-15745, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-25931126

RESUMEN

To further our aim of synthesizing aldehyde-tagged proteins for research and biotechnology applications, we developed methods for recombinant production of aerobic formylglycine-generating enzyme (FGE) in good yield. We then optimized the FGE biocatalytic reaction conditions for conversion of cysteine to formylglycine in aldehyde tags on intact monoclonal antibodies. During the development of these conditions, we discovered that pretreating FGE with copper(II) is required for high turnover rates and yields. After further investigation, we confirmed that both aerobic prokaryotic (Streptomyces coelicolor) and eukaryotic (Homo sapiens) FGEs contain a copper cofactor. The complete kinetic parameters for both forms of FGE are described, along with a proposed mechanism for FGE catalysis that accounts for the copper-dependent activity.


Asunto(s)
Proteínas Bacterianas/química , Coenzimas/química , Cobre/química , Streptomyces coelicolor/enzimología , Sulfatasas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Coenzimas/metabolismo , Cobre/metabolismo , Cisteína/química , Cisteína/metabolismo , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro , Streptomyces coelicolor/genética , Sulfatasas/genética , Sulfatasas/metabolismo
5.
BMC Biotechnol ; 16: 23, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26911368

RESUMEN

BACKGROUND: The ability to site-specifically conjugate a protein to a payload of interest (e.g., a fluorophore, small molecule pharmacophore, oligonucleotide, or other protein) has found widespread application in basic research and drug development. For example, antibody-drug conjugates represent a class of biotherapeutics that couple the targeting specificity of an antibody with the chemotherapeutic potency of a small molecule drug. While first generation antibody-drug conjugates (ADCs) used random conjugation approaches, next-generation ADCs are employing site-specific conjugation. A facile way to generate site-specific protein conjugates is via the aldehyde tag technology, where a five amino acid consensus sequence (CXPXR) is genetically encoded into the protein of interest at the desired location. During protein expression, the Cys residue within this consensus sequence can be recognized by ectopically-expressed formylglycine generating enzyme (FGE), which converts the Cys to a formylglycine (fGly) residue. The latter bears an aldehyde functional group that serves as a chemical handle for subsequent conjugation. RESULTS: The yield of Cys conversion to fGly during protein production can be variable and is highly dependent on culture conditions. We set out to achieve consistently high yields by modulating culture conditions to maximize FGE activity within the cell. We recently showed that FGE is a copper-dependent oxidase that binds copper in a stoichiometric fashion and uses it to activate oxygen, driving enzymatic turnover. Building upon that work, here we show that by supplementing cell culture media with copper we can routinely reach high yields of highly converted protein. We demonstrate that cells incorporate copper from the media into FGE, which results in increased specific activity of the enzyme. The amount of copper required is compatible with large scale cell culture, as demonstrated in fed-batch cell cultures with antibody titers of 5 g · L(-1), specific cellular production rates of 75 pg · cell(-1) · d(-1), and fGly conversion yields of 95-98 %. CONCLUSIONS: We describe a process with a high yield of site-specific formylglycine (fGly) generation during monoclonal antibody production in CHO cells. The conversion of Cys to fGly depends upon the activity of FGE, which can be ensured by supplementing the culture media with 50 uM copper(II) sulfate.


Asunto(s)
Aldehídos/química , Anticuerpos/química , Cobre/metabolismo , Medios de Cultivo/química , Glicina/metabolismo , Aldehídos/análisis , Aldehídos/metabolismo , Animales , Anticuerpos/análisis , Anticuerpos/metabolismo , Células CHO , Cricetinae , Cricetulus , Glicina/química
6.
J Cell Sci ; 126(Pt 10): 2151-6, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23525003

RESUMEN

Trafficking of the chitin synthase Chs2p from the endoplasmic reticulum (ER) to the bud-neck in late mitosis is tightly regulated by the cell cycle via phosphorylation of serine residues in the N-terminus of the protein. Here, we describe the effects of Chs2p phosphorylation on the interaction with coat protein complex II (COPII). Identification of a cdc5(ts) mutant, which fails to transport Chs2p-3xGFP to the bud-neck and instead accumulates the protein in intracellular puncta, led us to discover that Chs2p-3xGFP accumulates at ER exit sites in metaphase-arrested wild-type cells. Using an in vitro ER vesicle formation assay we showed that phosphorylation of Chs2p by the cyclin-dependent kinase CDK1 prevents packaging into COPII vesicles, whereas dephosphorylation of Chs2p by the phosphatase Cdc14p stimulates selection into the vesicles. We found that the cytoplasmic N-terminal domain of Chs2p, which contains the CDK1 phosphorylation sites, interacts with the COPII component Sec24p in a yeast two-hybrid assay and that phosphomimetic substitutions of serines at the CDK1 consensus sites reduces the interaction. Our data suggest that dephosphorylation functions as a molecular switch for regulated ER exit of Chs2p.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Vesículas Cubiertas por Proteínas de Revestimiento/fisiología , Quitina Sintasa/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Biomimética , Ciclo Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quitina Sintasa/genética , Ingeniería Genética , Proteínas de la Membrana/genética , Mutación/genética , Fosforilación/genética , Unión Proteica , Transporte de Proteínas/genética , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Técnicas del Sistema de Dos Híbridos , Proteínas de Transporte Vesicular
7.
Bioconjug Chem ; 25(7): 1331-41, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-24924618

RESUMEN

It is becoming increasingly clear that site-specific conjugation offers significant advantages over conventional conjugation chemistries used to make antibody-drug conjugates (ADCs). Site-specific payload placement allows for control over both the drug-to-antibody ratio (DAR) and the conjugation site, both of which play an important role in governing the pharmacokinetics (PK), disposition, and efficacy of the ADC. In addition to the DAR and site of conjugation, linker composition also plays an important role in the properties of an ADC. We have previously reported a novel site-specific conjugation platform comprising linker payloads designed to selectively react with site-specifically engineered aldehyde tags on an antibody backbone. This chemistry results in a stable C-C bond between the antibody and the cytotoxin payload, providing a uniquely stable connection with respect to the other linker chemistries used to generate ADCs. The flexibility and versatility of the aldehyde tag conjugation platform has enabled us to undertake a systematic evaluation of the impact of conjugation site and linker composition on ADC properties. Here, we describe the production and characterization of a panel of ADCs bearing the aldehyde tag at different locations on an IgG1 backbone conjugated using Hydrazino-iso-Pictet-Spengler (HIPS) chemistry. We demonstrate that in a panel of ADCs with aldehyde tags at different locations, the site of conjugation has a dramatic impact on in vivo efficacy and pharmacokinetic behavior in rodents; this advantage translates to an improved safety profile in rats as compared to a conventional lysine conjugate.


Asunto(s)
Aldehídos/química , Anticuerpos Monoclonales/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Receptor ErbB-2/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunoconjugados/farmacología , Ratones , Ratones SCID , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Bioconjug Chem ; 24(6): 846-51, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23731037

RESUMEN

Aldehyde- and ketone-functionalized biomolecules have found widespread use in biochemical and biotechnological fields. They are typically conjugated with hydrazide or aminooxy nucleophiles under acidic conditions to yield hydrazone or oxime products that are relatively stable, but susceptible to hydrolysis over time. We introduce a new reaction, the hydrazino-Pictet-Spengler (HIPS) ligation, which has two distinct advantages over hydrazone and oxime ligations. First, the HIPS ligation proceeds quickly near neutral pH, allowing for one-step labeling of aldehyde-functionalized proteins under mild conditions. Second, the HIPS ligation product is very stable (>5 days) in human plasma relative to an oxime-linked conjugate (∼1 day), as demonstrated by monitoring protein-fluorophore conjugates by ELISA. Thus, the HIPS ligation exhibits a combination of product stability and speed near neutral pH that is unparalleled by current carbonyl bioconjugation chemistries.


Asunto(s)
Materiales Biocompatibles/química , Hidrazinas/química , Proteínas/química , Aldehídos/química , Humanos , Concentración de Iones de Hidrógeno , Hidrólisis , Cetonas/química , Modelos Moleculares , Estructura Molecular , Oximas/química
9.
Mol Cancer Ther ; 19(9): 1866-1874, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32651200

RESUMEN

Trastuzumab and the related ADC, ado-trastuzumab emtansine (T-DM1), both target HER2-overexpressing cells. Together, these drugs have treatment indications in both early-stage and metastatic settings for HER2+ breast cancer. T-DM1 retains the antibody functionalities of trastuzumab and adds the potency of a cytotoxic maytansine payload. Interestingly, in the clinic, T-DM1 cannot always replace the use of trastuzumab plus chemotherapy administered together as single agents. We hypothesize that this failure may be due, in part, to the limited systemic exposure achieved by T-DM1 relative to trastuzumab because of toxicity-related dosing constraints on the ADC. We have developed a trastuzumab-based ADC site specifically conjugated to maytansine through a noncleavable linker. This construct, termed CAT-01-106, has a drug-to-antibody ratio (DAR) of 1.8, approximately half the average DAR of T-DM1, which comprises a mixture of antibodies variously conjugated with DARs ranging from 0 to 8. The high DAR species present in T-DM1 contribute to its toxicity and limit its clinical dose. CAT-01-106 showed superior in vivo efficacy compared with T-DM1 at equal payload dosing and was equally or better tolerated compared with T-DM1 at equal payload dosing up to 120 mg/kg in Sprague-Dawley rats and 60 mg/kg in cynomolgus monkeys. CAT-01-106 also showed improved pharmacokinetics in rats relative to T-DM1, with 40% higher ADC exposure levels. Together, the data suggest that CAT-01-106 may be sufficiently tolerable to enable clinical dosing at trastuzumab-equivalent exposure levels, combining the functions of both the antibody and the payload in one drug and potentially improving patient outcomes.


Asunto(s)
Ado-Trastuzumab Emtansina/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Inmunoconjugados/administración & dosificación , Maitansina/química , Trastuzumab/química , Ado-Trastuzumab Emtansina/efectos adversos , Ado-Trastuzumab Emtansina/farmacocinética , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunoconjugados/efectos adversos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Macaca fascicularis , Dosis Máxima Tolerada , Ratas , Ratas Sprague-Dawley , Receptor ErbB-2/metabolismo , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Methods Mol Biol ; 2033: 131-147, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31332752

RESUMEN

As a critical feature of the next generation of antibody-drug conjugates (ADCs), site-specific bioconjugation approaches can help to optimize stability, pharmacokinetics, efficacy, and safety as well as improve manufacturing consistency. The SMARTag® technology platform offers a practical and efficient chemoenzymatic solution for site-specific protein modifications. A bioorthogonal aldehyde handle is introduced through the oxidation of a cysteine residue, embedded in a specific peptide sequence (CxPxR), to the aldehyde-bearing formylglycine (fGly). This enzymatic modification is carried out by the formylglycine-generating enzyme (FGE). The broad recognition of this short sequence by FGE within the context of heterologous proteins allows for the introduction of fGly residues at chosen sites in proteins expressed in prokaryotic and eukaryotic systems. The protocol presented here describes the methods for expressing fGly-containing antibodies in eukaryotic cells and subsequent site-specific conjugation with a payload-linker using aldehyde-specific Hydrazino-Iso-Pictet-Spengler (HIPS) chemistry.


Asunto(s)
Inmunoconjugados/genética , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Ingeniería de Proteínas/métodos , Proteínas/química , Aldehídos/química , Glicina/análogos & derivados , Humanos , Inmunoconjugados/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/genética , Péptidos/química , Péptidos/genética , Procesamiento Proteico-Postraduccional/genética , Proteínas/genética
11.
Oncoimmunology ; 8(4): e1565859, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30906660

RESUMEN

Oncology treatment has been revolutionized by the introduction of immune checkpoint inhibitor drugs, which enable 20-40% of patients to generate anti-tumor immune responses. Combination treatment approaches with chemotherapeutic drugs may enable responses in the remaining patient cohorts. In this regard, a handful of drugs are promising due to their ability to induce immunogenic cell death in target cells. However, these agents are systemically delivered and indiscriminately cytotoxic to proliferating cells. By contrast, antibody-drug conjugates can selectively deliver a cytotoxic payload to a tumor, sparing most healthy cells. The ability of antibody-drug conjugates to induce immunogenic cell death in target cells has not yet been determined, although preclinical in vivo studies suggest this possibility. Here, we describe for the first time production of the in vitro hallmarks of immunogenic cell death - ecto-calreticulin and secreted ATP and HMGB1 protein - by cells in response to treatment with antibody-drug conjugates bearing a maytansine payload.

12.
Antibodies (Basel) ; 8(4)2019 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-31694242

RESUMEN

A promising molecular target for aggressive cancers is the urokinase receptor (uPAR). A fully human, recombinant antibody that binds uPAR to form a stable complex that blocks uPA-uPAR interactions (2G10) and is internalized primarily through endocytosis showed efficacy in a mouse xenograft model of highly aggressive, triple negative breast cancer (TNBC). Antibody-drug conjugates (ADCs) of 2G10 were designed and produced bearing tubulin inhibitor payloads ligated through seven different linkers. Aldehyde tag technology was employed for linking, and either one or two tags were inserted into the antibody heavy chain, to produce site-specifically conjugated ADCs with drug-to-antibody ratios of either two or four. Both cleavable and non-cleavable linkers were combined with two different antimitotic toxins-MMAE (monomethylauristatin E) and maytansine. Nine different 2G10 ADCs were produced and tested for their ability to target uPAR in cell-based assays and a mouse model. The anti-uPAR ADC that resulted in tumor regression comprised an MMAE payload with a cathepsin B cleavable linker, 2G10-RED-244-MMAE. This work demonstrates in vitro activity of the 2G10-RED-244-MMAE in TNBC cell lines and validates uPAR as a therapeutic target for TNBC.

13.
Methods Mol Biol ; 1728: 3-16, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29404988

RESUMEN

Enzymatic modification of proteins can generate uniquely reactive chemical functionality, enabling site-specific reactions on the protein surface. Formylglycine-generating enzyme (FGE) is one enzyme that can be exploited in this fashion. FGE binds its consensus sequence (CXPXR, known as the "aldehyde-tag") and converts the cysteine to a formylglycine (fGly). fGly-containing proteins contain a bioorthogonal aldehyde on their surface that can be modified selectively in the presence of the 20 canonical amino acids. Here, we describe protocols for the generation of a site-specifically modified protein, an antibody-drug conjugate (ADC), using aldehyde-tagging protocols and aldehyde-reactive conjugation chemistry.


Asunto(s)
Enzimas/química , Enzimas/metabolismo , Glicina/análogos & derivados , Inmunoconjugados/química , Aldehídos/química , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Células CHO , Catálisis , Línea Celular , Cromatografía Liquida , Cricetulus , Cisteína/química , Enzimas/genética , Glicina/biosíntesis , Glicina/química , Humanos , Inmunoconjugados/metabolismo , Mutagénesis Sitio-Dirigida , Especificidad por Sustrato , Espectrometría de Masas en Tándem
14.
Antibodies (Basel) ; 7(4)2018 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-31544890

RESUMEN

We hypothesized that systematic liquid chromatography-tandem mass spectrometry investigations of an antibody-drug conjugate (ADC), its small and large molecular components, and surrogate small-molecule conjugates might comprise a simple and efficient approach for the extended characterization of ADCs. Furthermore, we envisioned that results from this work might allow us to assign specific composition changes in the ADC based on monoisotopic mass shifts of conjugatable modifications as detected in the surrogate small-molecule conjugates. We tested our hypothesis with a case study using an aldehyde-tag-based ADC conjugated to a noncleavable linker bearing a maytansine payload. Nearly quantitative bioconversion from cysteine to formylglycine was observed in the monoclonal antibody, and bioorthogonal conjugation was detected only on the formylglycine residues in the ADC. Using our method, both conjugatable and nonconjugatable modifications were discovered in the linker/payload; however, only conjugatable modifications were observed on the ADC. Based on these results, we anticipate that our approach to systematic mass spectrometric investigations can be successfully applied to other ADCs and therapeutic bioconjugates for investigational new drug (IND)-enabling extended characterization.

15.
MAbs ; 10(8): 1182-1189, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30252630

RESUMEN

The advantages of site-specific over stochastic bioconjugation technologies include homogeneity of product, minimal perturbation of protein structure/function, and - increasingly - the ability to perform structure activity relationship studies at the conjugate level. When selecting the optimal location for site-specific payload placement, many researchers turn to in silico modeling of protein structure to identify regions predicted to offer solvent-exposed conjugatable sites while conserving protein function. Here, using the aldehyde tag as our site-specific technology platform and human IgG1 antibody as our target protein, we demonstrate the power of taking an unbiased scanning approach instead. Scanning insertion of the human formylglycine generating enzyme (FGE) recognition sequence, LCTPSR, at each of the 436 positions in the light and heavy chain antibody constant regions followed by co-expression with FGE yielded a library of antibodies bearing an aldehyde functional group ready for conjugation. Each of the variants was expressed, purified, and conjugated to a cytotoxic payload using the Hydrazinyl Iso-Pictet-Spengler ligation to generate an antibody-drug conjugate (ADC), which was analyzed in terms of conjugatability (assessed by drug-to-antibody ratio, DAR) and percent aggregate. We searched for insertion sites that could generate manufacturable ADCs, defined as those variants yielding reasonable antibody titers, DARs of ≥ 1.3, and ≥ 95% monomeric species. Through this process, we discovered 58 tag insertion sites that met these metrics, including 14 sites in the light chain, a location that had proved refractory to the placement of manufacturable tag sites using in silico modeling/rational approaches.


Asunto(s)
Aldehídos/inmunología , Inmunoconjugados/inmunología , Regiones Constantes de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Aldehídos/química , Secuencia de Aminoácidos , Sitios de Unión , Simulación por Computador , Composición de Medicamentos/métodos , Glicina/análogos & derivados , Glicina/química , Glicina/genética , Glicina/inmunología , Humanos , Inmunoconjugados/química , Inmunoconjugados/genética , Regiones Constantes de Inmunoglobulina/química , Regiones Constantes de Inmunoglobulina/genética , Inmunoglobulina G/química , Inmunoglobulina G/genética , Biblioteca de Péptidos , Unión Proteica
16.
Mol Cancer Ther ; 17(1): 161-168, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29142069

RESUMEN

Hematologically derived tumors make up ∼10% of all newly diagnosed cancer cases in the United States. Of these, the non-Hodgkin lymphoma (NHL) designation describes a diverse group of cancers that collectively rank among the top 10 most commonly diagnosed cancers worldwide. Although long-term survival trends are improving, there remains a significant unmet clinical need for treatments to help patients with relapsed or refractory disease, one cause of which is drug efflux through upregulation of xenobiotic pumps, such as MDR1. CD22 is a clinically validated target for the treatment of NHL, but no anti-CD22 agents have yet been approved for this indication. Recent approval of an anti-CD22 antibody-drug conjugate (ADC) for the treatment of relapsed/refractory ALL supports the rationale for targeting this protein. An opportunity exists for a next-generation anti-CD22 antibody-drug conjugate (ADC) to address unmet medical needs in the relapsed/refractory NHL population. We describe a site-specifically conjugated antibody-drug conjugate, made using aldehyde tag technology, targeted against CD22 and bearing a noncleavable maytansine payload that is resistant to MDR1-mediated efflux. The construct was efficacious against CD22+ NHL xenografts and could be repeatedly dosed in cynomolgus monkeys at 60 mg/kg with no observed significantly adverse effects. Exposure to total ADC at these doses (as assessed by AUC0-inf) indicated that the exposure needed to achieve efficacy was below tolerable limits. Together, the data suggest that this drug has the potential to be used effectively in patients with CD22+ tumors that have developed MDR1-related resistance to prior therapies. Mol Cancer Ther; 17(1); 161-8. ©2017 AACR.


Asunto(s)
Inmunoconjugados/farmacología , Maitansina/administración & dosificación , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Animales , Resistencia a Antineoplásicos , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
18.
Artículo en Inglés | MEDLINE | ID: mdl-27311685

RESUMEN

Antibody-drug conjugates represent a growing class of biologic drugs that use the targeted specificity of an antibody to direct the localization of a small molecule drug, often a cytotoxic payload. After conjugation, antibody-drug conjugate preparations typically retain a residual amount of free (unconjugated) linker-payload. Monitoring this free small molecule drug component is important due to the potential for free payload to mediate unintended (off-target) toxicity. We developed a simple RP-HPLC/MRM-MS-based assay that can be rapidly employed to quantify free linker-payload. The method uses low sample volumes and offers an LLOQ of 10nM with 370pg on column. This analytical approach was used to monitor free linker-payload removal during optimization of the tangential flow filtration manufacturing step.


Asunto(s)
Cromatografía Liquida/métodos , Inmunoconjugados/química , Espectrometría de Masas/métodos , Preparaciones Farmacéuticas/química
19.
ACS Med Chem Lett ; 7(11): 994-998, 2016 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-27882197

RESUMEN

Expanded ligation techniques are sorely needed to generate unique linkages for the growing field of functionally enhanced proteins. To address this need, we present a unique chemical ligation that involves the double addition of a pyrazolone moiety with an aldehyde-labeled protein. This ligation occurs via a tandem Knoevenagel condensation-Michael addition. A pyrazolone reacts with an aldehyde to generate an enone, which undergoes subsequent attack by a second pyrazolone to generate a bis-pyrazolone species. This rapid and facile ligation technique is performed under mild conditions in the absence of catalyst to generate new architectures that were previously inaccessible via conventional ligation reactions. Using this unique ligation, we generated three site-specifically labeled antibody-drug conjugates (ADCs) with an average of four drugs to one antibody. The in vitro and in vivo efficacies along with pharmacokinetic data of the site-specific ADCs are reported.

20.
Chem Biol ; 22(2): 293-8, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25619935

RESUMEN

There is a need for facile chemistries that allow for chemo- and regioselectivity in bioconjugation reactions. To address this need, we are pioneering site-specific bioconjugation methods that use formylglycine as a bioorthogonal handle on a protein surface. Here we introduce aldehyde-specific bioconjugation chemistry, the trapped-Knoevenagel ligation. The speed and stability of the trapped-Knoevenagel ligation further advances the repertoire of aldehyde-based bioconjugations and expands the toolbox for site-specific protein modifications. The trapped-Knoevenagel ligation reaction can be run at near neutral pH in the absence of catalysts to produce conjugates that are stable under physiological conditions. Using this new ligation, we generated an antibody-drug conjugate that demonstrates excellent efficacy in vitro and in vivo.


Asunto(s)
Carbono/química , Proteínas/química , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/toxicidad , Brentuximab Vedotina , Catálisis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Inmunoconjugados/química , Pirazoles/química , Trastuzumab/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA