Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Anal Chem ; 95(49): 17957-17961, 2023 12 12.
Article in English | MEDLINE | ID: mdl-38084380

ABSTRACT

Biotransformation leading to single residue modifications (e.g., deamidation, oxidation) can contribute to decreased efficacy/potency, poor pharmacokinetics, and/or toxicity/immunogenicity for protein therapeutics. Identifying and characterizing such liabilities in vivo are emerging needs for biologics drug discovery. In vitro stress assays involving PBS for deamidation or AAPH for oxidation are commonly used for predicting liabilities in manufacturing and storage and are sometimes considered a predictive tool for in vivo liabilities. However, reports discussing their in vivo translatability are limited. Herein, we introduce a mass spectrometry workflow that characterizes in vivo oxidation and deamidation in pharmacokinetically relevant compartments for diverse protein therapeutic modalities. The workflow has low bias of <10% in quantitating degradation in the relevant pharmacokinetic concentration range for monkey and rabbit serum/plasma (1-100 µg/mL) and allows for high sequence coverage (∼85%) for discovery/monitoring of amino acid modifications. For oxidation and deamidation, the assay was precise, with percent coefficient of variation of <8% at 1-100 µg/mL and ≤6% method-induced artifacts. A high degree of in vitro and in vivo correlation was observed for deamidation on the six diverse protein therapeutics (seven liability sites) tested. In vivo translatability for oxidation liabilities were not observed for the 11 molecules tested using in vitro AAPH stress. One of the molecules dosed in eyes resulted in a false positive and a false negative prediction for in vivo oxidation following AAPH stress. Finally, peroxide stress was also tested but resulted in limited success (1 out of 4 molecules) in predicting oxidation liabilities.


Subject(s)
Oxidation-Reduction , Animals , Rabbits , Biotransformation
2.
Anal Chem ; 95(11): 4834-4839, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36876898

ABSTRACT

The growing opportunities recognized for covalent drug inhibitors, like KRAS G12C inhibitors, are driving the need for mass spectrometry methods that can quickly and robustly measure therapeutic drug activity in vivo for drug discovery research and development. Effective front-end sample preparation is critical for proteins extracted from tumors but is generally labor intensive and impractical for large sample numbers typical in pharmacodynamic (PD) studies. Herein, we describe an automated and integrated sample preparation method for the measurement of activity levels of KRAS G12C drug inhibitor alkylation from complex tumor samples involving high throughput detergent removal and preconcentration followed by quantitation using mass spectrometry. We introduce a robust assay with an average intra-assay coefficient of variation (CV) of 4% and an interassay CV of 6% obtained from seven studies, enabling us to understand the relationship between KRAS G12C target occupancy and the therapeutic PD effect from mouse tumor samples. Further, the data demonstrated that the drug candidate GDC-6036, a KRAS G12C covalent inhibitor, shows dose-dependent target inhibition (KRAS G12C alkylation) and MAPK pathway inhibition, which correlate with high antitumor potency in the MIA PaCa-2 pancreatic xenograft model.


Subject(s)
Antineoplastic Agents , Proto-Oncogene Proteins p21(ras) , Humans , Animals , Mice , Proto-Oncogene Proteins p21(ras)/genetics , Cell Line, Tumor , Mutation , Antineoplastic Agents/pharmacology , Disease Models, Animal
3.
Anal Bioanal Chem ; 414(22): 6601-6610, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35821276

ABSTRACT

Proteases are critical proteins involved in cleaving substrates that may impact biological pathways, cellular processes, or disease progression. In the biopharmaceutical industry, modulating the levels of protease activity is an important strategy for mitigating many types of diseases. While a variety of analytical tools exist for characterizing substrate cleavages, in vitro functional screening for antibody inhibitors of protease activity using physiologically relevant intact protein substrates remains challenging. In addition, detecting such large protein substrates with high heterogeneity using high-throughput mass spectrometry screening has rarely been reported in the literature with concerns for assay robustness and sensitivity. In this study, we established a peptide-based in vitro functional screening assay for antibody inhibitors of mouse bone morphogenic protein 1 (mBMP1) metalloprotease using a heterogeneous recombinant 66-kDa mouse Procollagen I alpha 1 chain (mProcollagen) substrate. We compared several analytical tools including capillary gel electrophoresis Western blot (CE-Western blot), as well as both intact protein and peptide-based mass spectrometry (MS) to quantitate the mBMP1 proteolytic activity and its inhibition by antibodies using this heterogeneous mProcollagen substrate. We concluded that the peptide-based mass spectrometry screening assay was the most suitable approach in terms of throughput, sensitivity, and assay robustness. We then optimized our mBMP1 proteolysis reaction after characterizing the enzyme kinetics using the peptide-based MS assay. This assay resulted in Z' values ranging from 0.6 to 0.8 from the screening campaign. Among over 1200 antibodies screened, IC50 characterization was performed on the top candidate hits, which showed partial or complete inhibitory activities against mBMP1.


Subject(s)
Peptides , Procollagen , Animals , Mass Spectrometry , Mice , Peptide Hydrolases/metabolism , Peptides/metabolism , Procollagen/metabolism , Proteins/metabolism , Proteolysis , Substrate Specificity
4.
Proc Natl Acad Sci U S A ; 117(18): 9851-9856, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32327606

ABSTRACT

Toward the goal of increasing the throughput of high-resolution mass characterization of intact antibodies, we developed a RapidFire-mass spectrometry (MS) assay using electrospray ionization. We achieved unprecedented screening throughput as fast as 15 s/sample, which is an order of magnitude improvement over conventional liquid chromatography (LC)-MS approaches. The screening enabled intact mass determination as accurate as 7 ppm with baseline resolution at the glycoform level for intact antibodies. We utilized this assay to characterize and perform relative quantitation of antibody species from 248 samples of 62 different cell line clones at four time points in 2 h using RapidFire-time-of-flight MS screening. The screening enabled selection of clones with the highest purity of bispecific antibody production and the results significantly correlated with conventional LC-MS results. In addition, analyzing antibodies from a complex plasma sample using affinity-RapidFire-MS was also demonstrated and qualified. In summary, the platform affords high-throughput analyses of antibodies, including bispecific antibodies and potential mispaired side products, in cell culture media, or other complex matrices.


Subject(s)
Antibodies, Bispecific/blood , Antibodies/blood , High-Throughput Screening Assays/methods , Spectrometry, Mass, Electrospray Ionization/methods , Antibodies/isolation & purification , Antibodies, Bispecific/isolation & purification , Cell Line , Chromatography, Liquid/methods , Humans
5.
ACS Med Chem Lett ; 11(4): 541-549, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292562

ABSTRACT

A class of imidazoisoindole (III) heme-binding indoleamine-2,3-dioxygenase (IDO1) inhibitors were optimized via structure-based drug design into a series of tryptophan-2,3-dioxygenase (TDO)-selective inhibitors. Kynurenine pathway modulation was demonstrated in vivo, which enabled evaluation of TDO as a potential cancer immunotherapy target. As means of mitigating the risk of drug-drug interactions arising from cytochrome P450 inhibition, a novel property-based drug design parameter, herein referred to as the CYP Index, was implemented for the design of inhibitors with appreciable selectivity for TDO over CYP3A4. We anticipate the CYP Index will be a valuable design parameter for optimizing CYP inhibition of any small molecule inhibitor containing a Lewis basic motif capable of binding heme.

6.
Anal Chem ; 92(10): 6839-6843, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32309925

ABSTRACT

There are many pharmacokinetic challenges associated with administering protein therapeutics, including biotransformation via clipping, deamidation, isomerization, oxidation, etc. In the case of engineered multivalent tethered antibody formats, proteolysis or deconjugation at the fusion or conjugation site present further issues. Unlike degradations associated with antibody drug conjugates, such biotransformations of tethered antibody formats usually result in degraded products with large mass differences. These large differences can result in processing or mass spectrometry response bias among the resulting product species that can lead to inaccurate stability quantitation. Herein, we describe an assay strategy for characterizing and quantitating degradations accurately for multivalent antibodies by incorporating response bias corrections. For the multivalent tethered antibody molecules selected, an ∼30-80% difference in response, compared to the cleaved product, was observed. To correct for the response bias, selected tethered multivalent antibodies and an IgG antibody (representing the stable intact and the degraded product species, respectively) were spiked in serum at known ratios for analysis. Following affinity capture, we generated calibration curves (five-parameter logistic fit p < 0.05) by plotting the measured ratios of the MS ion responses against the known spiked-in ratios (CVs < 8% for calibration standards). The qualified calibration curve (accuracy within 8% and 2% for measuring degradations of 5% and 15% product, respectively) was then used, through interpolation, to determine stability profiles for the same multivalent tethered antibody formats from both in vitro serum and pharmacokinetic study samples.


Subject(s)
Antibodies/analysis , Immunoconjugates/analysis , Chromatography, Liquid , Mass Spectrometry
7.
MAbs ; 12(1): 1715705, 2020.
Article in English | MEDLINE | ID: mdl-31997712

ABSTRACT

For antibody-drug conjugates to be efficacious and safe, they must be stable in circulation to carry the payload to the site of the targeted cell. Several components of a drug-conjugated antibody are known to influence stability: 1) the site of drug attachment on the antibody, 2) the linker used to attach the payload to the antibody, and 3) the payload itself. In order to support the design and optimization of a high volume of drug conjugates and avoid unstable conjugates prior to testing in animal models, we wanted to proactively identify these potential liabilities. Therefore, we sought to establish an in vitro screening method that best correlated with in vivo stability. While traditionally plasma has been used to assess in vitro stability, our evaluation using a variety of THIOMABTM antibody-drug conjugates revealed several disconnects between the stability assessed in vitro and the in vivo outcomes when using plasma. When drug conjugates were incubated in vitro for 24 h in mouse whole blood rather than plasma and then analyzed by affinity capture LC-MS, we found an improved correlation to in vivo stability with whole blood (R2 = 0.87, coefficient of determination) compared to unfrozen or frozen mouse plasma (R2 = 0.34, 0.01, respectively). We further showed that this whole blood assay was also able to predict in vivo stability of other preclinical species such as rat and cynomolgus monkey, as well as in human. The screening method utilized short (24 h) incubation times, as well as a custom analysis software, allowing increased throughput and in-depth biotransformation characterization. While some instabilities that were more challenging to identify remain, the method greatly enhanced the process of screening, optimizing, and lead candidate selection, resulting in the substantial reduction of animal studies.


Subject(s)
Chromatography, Liquid/methods , Immunoconjugates/chemistry , Mass Spectrometry/methods , Animals , Humans , In Vitro Techniques , Protein Stability
8.
Anal Chem ; 92(2): 2186-2193, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31880920

ABSTRACT

With the rapid rise of therapeutic antibodies and antibody-drug conjugates, significant investments have been made in developing workflows that utilize mass spectrometry to detect these intact molecules, the large fragments generated by their selective digestion, and the peptides generated by traditional proteomics workflows. The resultant data is used to gain insight into a wide range of parameters, including primary sequence, disulfide bonding, glycosylation patterns, biotransformation, and more. However, many of the technologies utilized to couple these workflows to mass spectrometers have significant limitations that force nonoptimal modifications to upstream sample preparation steps, limit the throughput of high-volume workflows, and prevent the harmonization of diverse experiments onto a single hardware platform. Here, we describe a new analytical platform that enables direct and high-throughput coupling to electrospray ionization mass spectrometry. The SampleStream platform is compatible with both native and denaturing electrospray, operates with a throughput of up to 15 s/sample, provides extensive concentration of dilute samples, and affords similar sensitivity to comparable liquid chromatographic methods.


Subject(s)
Antibodies, Monoclonal/analysis , High-Throughput Screening Assays , Immunoconjugates/analysis , High-Throughput Screening Assays/instrumentation , Software , Spectrometry, Mass, Electrospray Ionization/instrumentation
9.
Drug Metab Dispos ; 47(10): 1156-1163, 2019 10.
Article in English | MEDLINE | ID: mdl-31085544

ABSTRACT

In cells, catalytic disulfide cleavage is an essential mechanism in protein folding and synthesis. However, detailed enzymatic catalytic mechanism relating cleavage of disulfide bonds in xenobiotics is not well understood. This study reports an enzymatic mechanism of cleavage of disulfide bonds in xenobiotic small molecules and antibody conjugate (ADC) linkers. The chemically stable disulfide bonds in substituted disulfide-containing pyrrolobenzodiazepine (PBD, pyrrolo[2,1-c][1,4]benzodiazepine) monomer prodrugs in presence of glutathione or cysteine were found to be unstable in incubations in whole blood of humans and rats. It was shown the enzymes involved were thioredoxin (TRX) and glutaredoxin (GRX). For a diverse set of drug-linker conjugates, we determined that TRX in the presence of TRX-reductase and NADPH generated the cleaved products that are consistent with catalytic disulfide cleavage and linker immolation. GRX was less rigorously studied; in the set of compounds studied, its role in the catalytic cleavage was also confirmed. Collectively, these in vitro experiments demonstrate that TRX as well as GRX can catalyze the cleavage of disulfide bonds in both small molecules and linkers of ADCs.


Subject(s)
Glutaredoxins/metabolism , Immunoconjugates/pharmacokinetics , Thioredoxins/metabolism , Animals , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Disulfides/chemistry , Disulfides/metabolism , Female , Humans , Immunoconjugates/chemistry , Male , Pyrroles/chemistry , Pyrroles/metabolism , Rats , Recombinant Proteins/metabolism , Thioredoxin-Disulfide Reductase/metabolism
10.
Anal Chem ; 91(1): 903-911, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30481450

ABSTRACT

High throughput protein-ligand interaction screening assays employing mass spectrometric detection are widely used in early stage drug discovery. Mass spectrometry-based screening approaches employ a target protein added to a pool of small-molecule compounds, and binding is assessed by measuring ligands denatured from the complexes. Direct analysis of protein-ligand interactions using native mass spectrometry has been demonstrated but is not widely used due to the detection limit on protein size, the requirement of volatile buffers, and the necessity for specialized instrumentation to preserve weak interactions under native conditions. Here we present a robust, quantitative, and automated online size-exclusion chromatography-native mass spectrometry (SEC-nMS) platform for measuring affinities of noncovalent protein-small-molecule interactions on an Orbitrap mass spectrometer. Indoleamine 2,3-dioxygenase 1, a catabolic enzyme, and inhibitory ligands were employed as a demonstration of the method. Efficient separation and elution enabled preservation of protein-ligand complexes and increased throughput. The high sensitivity and intra charge state resolution at high m/ z offered by the Exactive Plus EMR Orbitrap allowed for protein ligand affinity quantitation and resolved individual compounds close in mass. Vc50 values determined via collision-induced dissociation experiments enabled the evaluation of complex stability in the gas phase and were found to be independent of the extent of complex formation. For the first time, Vc50 determinations were achieved on an inline SEC-nMS platform. Systematic comparison of our method with optimized chip-based nanoelectrospray infusion served as a reference for ligand screening and affinity quantitation and further revealed the advantages of SEC-MS.


Subject(s)
Acetates/analysis , Enzyme Inhibitors/analysis , High-Throughput Screening Assays , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Small Molecule Libraries/analysis , Acetates/pharmacology , Chromatography, Gel , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Ligands , Mass Spectrometry , Small Molecule Libraries/pharmacology
11.
J Am Soc Mass Spectrom ; 29(7): 1532-1537, 2018 07.
Article in English | MEDLINE | ID: mdl-29845559

ABSTRACT

Antibody-drug conjugates (ADCs) present unique challenges for ligand-binding assays primarily due to the dynamic changes of the drug-to-antibody ratio (DAR) distribution in vivo and in vitro. Here, an automated on-tip affinity capture platform with subsequent mass spectrometry analysis was developed to accurately characterize the DAR distribution of ADCs from biological matrices. A variety of elution buffers were tested to offer optimal recovery, with trastuzumab serving as a surrogate to the ADCs. High assay repeatability (CV 3%) was achieved for trastuzumab antibody when captured below the maximal binding capacity of 7.5 µg. Efficient on-tip deglycosylation was also demonstrated in 1 h followed by affinity capture. Moreover, this tip-based platform affords higher throughput for DAR characterization when compared with a well-characterized bead-based method. Graphical Abstract ᅟ.


Subject(s)
Immunoconjugates/blood , Mass Spectrometry/methods , Animals , Antibodies, Monoclonal/blood , Chromatography, Liquid/methods , Haplorhini , Humans , Immunoconjugates/chemistry , Rats
12.
ACS Med Chem Lett ; 9(5): 417-421, 2018 May 10.
Article in English | MEDLINE | ID: mdl-29795752

ABSTRACT

Tryptophan 2,3-dioxygenase 2 (TDO2) catalyzes the conversion of tryptophan to the immunosuppressive metabolite kynurenine. TDO2 overexpression has been observed in a number of cancers; therefore, TDO inhibition may be a useful therapeutic intervention for cancers. We identified an aminoisoxazole series as potent TDO2 inhibitors from a high-throughput screen (HTS). An extensive medicinal chemistry effort revealed that both the amino group and the isoxazole moiety are important for TDO2 inhibitory activity. Computational modeling yielded a binding hypothesis and provided insight into the observed structure-activity relationships. The optimized compound 21 is a potent TDO2 inhibitor with modest selectivity over indolamine 2,3-dioxygenase 1 (IDO1) and with improved human whole blood stability.

13.
Bioconjug Chem ; 29(4): 1155-1167, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29481745

ABSTRACT

Previous investigations on antibody-drug conjugate (ADC) stability have focused on drug release by linker-deconjugation due to the relatively stable payloads such as maytansines. Recent development of ADCs has been focused on exploring technologies to produce homogeneous ADCs and new classes of payloads to expand the mechanisms of action of the delivered drugs. Certain new ADC payloads could undergo metabolism in circulation while attached to antibodies and thus affect ADC stability, pharmacokinetics, and efficacy and toxicity profiles. Herein, we investigate payload stability specifically and seek general guidelines to address payload metabolism and therefore increase the overall ADC stability. Investigation was performed on various payloads with different functionalities (e.g., PNU-159682 analog, tubulysin, cryptophycin, and taxoid) using different conjugation sites (HC-A118C, LC-K149C, and HC-A140C) on THIOMAB antibodies. We were able to reduce metabolism and inactivation of a broad range of payloads of THIOMAB antibody-drug conjugates by employing optimal conjugation sites (LC-K149C and HC-A140C). Additionally, further payload stability was achieved by optimizing the linkers. Coupling relatively stable sites with optimized linkers provided optimal stability and reduction of payloads metabolism in circulation in vivo.


Subject(s)
Antibodies/chemistry , Immunoconjugates/chemistry , Immunologic Factors/chemistry , Pharmaceutical Preparations/chemistry , Antigens/immunology , Binding Sites , Drug Stability , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Immunologic Factors/administration & dosage , Immunologic Factors/pharmacokinetics
14.
Bioconjug Chem ; 29(2): 473-485, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29425028

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , Cysteine/chemistry , Disulfides/chemistry , Immunoconjugates/chemistry , Maleimides/chemistry , Trastuzumab/chemistry , Animals , Antineoplastic Agents, Immunological/blood , Cysteine/blood , Cysteine/genetics , Disulfides/blood , Drug Stability , High-Throughput Screening Assays , Humans , Immunoconjugates/blood , Maleimides/blood , Models, Molecular , Mutagenesis, Site-Directed , Oligopeptides/blood , Oligopeptides/chemistry , Protein Aggregates , Protein Stability , Rats , Trastuzumab/blood , Trastuzumab/genetics
15.
J Atmos Sci ; 75(7): 2445-2472, 2018 Jul.
Article in English | MEDLINE | ID: mdl-33867577

ABSTRACT

We use 3-D cloud-resolving model (CRM) simulations of two mesoscale convective systems at mid-latitudes and a simple statistical ensemble method to diagnose the scale dependency of convective momentum transport (CMT) and CMT-related properties, and evaluate a parameterization scheme for convection-induced pressure gradient (CIPG) developed by Gregory et al. (GKI97). GKI97 relates CIPG to a constant coefficient multiplied by mass flux and vertical mean wind shear. CRM results show that mass fluxes and CMT exhibit strong scale dependency in temporal evolution and vertical structure. The prevalent understandings of CMT characteristics in terms of upgradient/downgradient transport are applicable to updrafts but not downdrafts across a wide range of grid spacings (4-512 km). For the small-to-median grid spacings (4~64 km), GKI97 reproduces some aspects of CIPG scale dependency except for underestimating the variations of CIPG as grid spacing decreases. However, for large grid spacings (128~512 km), GKI97 might even less adequately parameterize CIPG because it omits the contribution from either the nonlinear shear or buoyancy forcings. Further diagnosis of CRM results suggests that inclusion of nonlinear shear forcing in GKI97 is needed for the large grid spacings, and use of the three-updraft and one downdraft approach proposed in an earlier study may help a modified GKI97 capture more variations of CIPG as grid spacing decreases for the small-to-median grid spacings. Further, the optimal coefficients used in GKI97 seems insensitive to grid spacings, but they might be different for updrafts and downdrafts, for different MCS types, and for zonal and meridional components.

16.
J Med Chem ; 61(3): 989-1000, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29227683

ABSTRACT

Antibody-drug conjugates (ADCs) have become an important therapeutic modality for oncology, with three approved by the FDA and over 60 others in clinical trials. Despite the progress, improvements in ADC therapeutic index are desired. Peptide-based ADC linkers that are cleaved by lysosomal proteases have shown sufficient stability in serum and effective payload-release in targeted cells. If the linker can be preferentially hydrolyzed by tumor-specific proteases, safety margin may improve. However, the use of peptide-based linkers limits our ability to modulate protease specificity. Here we report the structure-guided discovery of novel, nonpeptidic ADC linkers. We show that a cyclobutane-1,1-dicarboxamide-containing linker is hydrolyzed predominantly by cathepsin B while the valine-citrulline dipeptide linker is not. ADCs bearing the nonpeptidic linker are as efficacious and stable in vivo as those with the dipeptide linker. Our results strongly support the application of the peptidomimetic linker and present new opportunities for improving the selectivity of ADCs.


Subject(s)
Cathepsin B/metabolism , Drug Discovery , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Peptidomimetics/chemistry , Peptidomimetics/metabolism , Humans , Intracellular Space/metabolism , Substrate Specificity
17.
Cancer Res ; 77(24): 7027-7037, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29046337

ABSTRACT

Antibody-drug conjugates (ADC) are designed to selectively bind to tumor antigens via the antibody and release their cytotoxic payload upon internalization. Controllable payload release through judicious design of the linker has been an early technological milestone. Here, we examine the effect of the protease-cleavable valine-citrulline [VC(S)] linker on ADC efficacy. The VC(S) linker was designed to be cleaved by cathepsin B, a lysosomal cysteine protease. Surprisingly, suppression of cathepsin B expression via CRISPR-Cas9 gene deletion or shRNA knockdown had no effect on the efficacy of ADCs with VC(S) linkers armed with a monomethyl auristatin E (MMAE) payload. Mass spectrometry studies of payload release suggested that other cysteine cathepsins can cleave the VC(S) linker. Also, ADCs with a nonprotease-cleavable enantiomer, the VC(R) isomer, mediated effective cell killing with a cysteine-VC(R)-MMAE catabolite generated by lysosomal catabolism. Based on these observations, we altered the payload to a pyrrolo[2,1-c][1,4]benzodiazepine dimer (PBD) conjugate that requires linker cleavage in order to bind its DNA target. Unlike the VC-MMAE ADCs, the VC(S)-PBD ADC is at least 20-fold more cytotoxic than the VC(R)-PBD ADC. Our findings reveal that the VC(S) linker has multiple paths to produce active catabolites and that antibody and intracellular targets are more critical to ADC efficacy. These results suggest that protease-cleavable linkers are unlikely to increase the therapeutic index of ADCs and that resistance based on linker processing is improbable. Cancer Res; 77(24); 7027-37. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/metabolism , Antineoplastic Agents/metabolism , Cathepsin B/physiology , Immunoconjugates/metabolism , Prodrugs/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Cathepsin B/metabolism , Cell Line, Tumor , Cells, Cultured , Citrulline/metabolism , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Immunoconjugates/therapeutic use , Oligopeptides , Prodrugs/therapeutic use , Proteolysis , Valine/metabolism
18.
J Med Chem ; 60(19): 8083-8102, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28929759

ABSTRACT

Significant data exists to suggest that dual leucine zipper kinase (DLK, MAP3K12) is a conserved regulator of neuronal degeneration following neuronal injury and in chronic neurodegenerative disease. Consequently, there is considerable interest in the identification of DLK inhibitors with a profile compatible with development for these indications. Herein, we use structure-based drug design combined with a focus on CNS drug-like properties to generate compounds with superior kinase selectivity and metabolic stability as compared to previously disclosed DLK inhibitors. These compounds, exemplified by inhibitor 14, retain excellent CNS penetration and are well tolerated following multiple days of dosing at concentrations that exceed those required for DLK inhibition in the brain.


Subject(s)
Alzheimer Disease/drug therapy , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Brain/drug effects , Brain/metabolism , Drug Design , Humans , Macaca fascicularis , Mice , Mice, Inbred C57BL , Models, Molecular , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
19.
SLAS Discov ; 22(10): 1211-1217, 2017 12.
Article in English | MEDLINE | ID: mdl-28581894

ABSTRACT

In recent years, the ability to unambiguously identify complex mixtures of analytes with high accuracy and resolving power in a label-free format continues to expand the application of mass spectrometry (MS) in the drug discovery process. This advantage combined with improved instrumentation makes MS suitable for targets with limited alternative assays for high-throughput screening (HTS). We describe a novel screening format using Self-Assembled Monolayers and matrix-assisted laser Desorption Ionization (SAMDI) technology. SAMDI enables affinity capture of a target protein for use in a small-molecule-binding assay format. Subsequent ionization enables the inferred identification of noncovalent compound interactions. SAMDI technology overcomes shot-to-shot variability by uniformly saturating the surface with captured protein, thereby minimizing matrix crystallization "hot spots." Furthermore, the combination with high-resolution matrix-assisted laser desorption/ionization time of flight significantly reduces interference of small-molecule detection from salt, detergent, and matrix. By using a pooled library format, the SAMDI assay can significantly improve the throughput of MS-based screening irrespective of enzyme activity. Finally, we demonstrate binding affinity rank ordering from a pool of compounds that correlates with potency data from a biochemical assay.


Subject(s)
Small Molecule Libraries/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Biological Assay , Cattle , Feasibility Studies , Solutions
20.
Bioorg Med Chem Lett ; 27(13): 2974-2981, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28512031

ABSTRACT

A high-throughput screening (HTS) of the Genentech/Roche library identified a novel, uncharged scaffold as a KDM5A inhibitor. Lacking insight into the binding mode, initial attempts to improve inhibitor potency failed to improve potency, and synthesis of analogs was further hampered by the presence of a C-C bond between the pyrrolidine and pyridine. Replacing this with a C-N bond significantly simplified synthesis, yielding pyrazole analog 35, of which we obtained a co-crystal structure with KDM5A. Using structure-based design approach, we identified 50 with improved biochemical, cell potency and reduced MW and lower lipophilicity (LogD) compared with the original hit. Furthermore, 50 showed lower clearance than 9 in mice. In combination with its remarkably low plasma protein binding (PPB) in mice (40%), oral dosing of 50 at 5mg/kg resulted in unbound Cmax ∼2-fold of its cell potency (PC9 H3K4Me3 0.96µM), meeting our criteria for an in vivo tool compound from a new scaffold.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Pyrazoles/pharmacology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Mice , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Rats , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...