60 results on '"Koerber JT"'
Search Results
2. Surface immobilization of hexa-histidine-tagged adeno-associated viral vectors for localized gene delivery
- Author
-
Jang, J-H, Koerber, JT, Gujraty, K, Bethi, SR, Kane, RS, and Schaffer, DV
- Subjects
Biotechnology ,Genetics ,Gene Therapy ,5.2 Cellular and gene therapies ,Development of treatments and therapeutic interventions ,Cells ,Cultured ,Dependovirus ,Gene Transfer Techniques ,Genetic Therapy ,Genetic Vectors ,Histidine ,Humans ,Oligopeptides ,Transduction ,Genetic ,AAV ,localized gene delivery ,substrate-mediated gene delivery ,hexa-histidine ,Biological Sciences ,Medical and Health Sciences - Abstract
Adeno-associated viral (AAV) vectors, which are undergoing broad exploration in clinical trials, have significant promise for therapeutic gene delivery because of their safety and delivery efficiency. Gene delivery technologies capable of mediating localized gene expression may further enhance the potential of AAV in a variety of therapeutic applications by reducing spread outside a target region, which may thereby reduce off-target side effects. We have genetically engineered an AAV variant capable of binding to surfaces with high affinity through a hexa-histidine metal-binding interaction. This immobilized AAV vector system mediates high-efficiency delivery to cells that contact the surface and thus may have promise for localized gene delivery, which may aid numerous applications of AAV delivery to gene therapy.
- Published
- 2010
3. Abstract P6-17-20: Withdrawn
- Author
-
Junttila, TT, primary, Ellerman, D, additional, Lombana, N, additional, Hristopoulos, M, additional, Clark, R, additional, Li, J, additional, Vij, R, additional, Koerber, JT, additional, Johnston, J, additional, Shelton, A, additional, Mai, E, additional, Gadkar, K, additional, Lo, AA, additional, Totpal, K, additional, Prell, R, additional, Lee, G, additional, Spiess, C, additional, and Slaga, D, additional
- Published
- 2019
- Full Text
- View/download PDF
4. A novel, label-free, pre-equilibrium assay to determine the association and dissociation rate constants of therapeutic antibodies on living cells.
- Author
-
Janezic EM, Doan A, Mai E, Bravo DD, Wang J, Kim HS, Spiess C, Bewley K, ElSohly A, Liang WC, Koerber JT, Richalet P, Vanhove M, and Comps-Agrar L
- Subjects
- Humans, Animals, Kinetics, Immunoglobulin G, Cricetulus, CHO Cells, Rabbits, Antibodies, Monoclonal chemistry
- Abstract
Background and Purpose: Monoclonal antibodies (Ab) represent the fastest growing drug class. Knowledge of the biophysical parameters (k
on , koff and KD ) that dictate Ab:receptor interaction is critical during the drug discovery process. However, with the increasing complexity of Ab formats and their targets, it became apparent that existing technologies present limitations and are not always suitable to determine these parameters. Therefore, novel affinity determination methods represent an unmet assay need., Experimental Approach: We developed a pre-equilibrium kinetic exclusion assay using recent mathematical advances to determine the kon , koff and KD of monoclonal Ab:receptor interactions on living cells. The assay is amenable to all human IgG1 and rabbit Abs., Key Results: Using our novel assay, we demonstrated for several monoclonal Ab:receptor pairs that the calculated kinetic rate constants were comparable with orthogonal methods that were lower throughput or more resource consuming. We ran simulations to predict the critical conditions to improve the performance of the assays. We further showed that this method could successfully be applied to both suspension and adherent cells. Finally, we demonstrated that kon and koff , but not KD , correlate with in vitro potency for a panel of monoclonal Abs., Conclusions and Implications: Our novel assay has the potential to systematically probe binding kinetics of monoclonal Abs to cells and can be incorporated in a screening cascade to identify new therapeutic candidates. Wide-spread adoption of pre-equilibrium assays using physiologically relevant systems will lead to a more holistic understanding of how Ab binding kinetics influence their potency., (© 2023 British Pharmacological Society.)- Published
- 2024
- Full Text
- View/download PDF
5. A real-time antibody-dependent cellular phagocytosis assay by live cell imaging.
- Author
-
Shi Y, Sun Y, Seki A, Rutz S, Koerber JT, and Wang J
- Subjects
- Humans, Antibody-Dependent Cell Cytotoxicity, Animals, Mice, Cell Line, Tumor, Phagocytosis, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Macrophages immunology, Macrophages metabolism
- Abstract
Antibody-dependent cellular phagocytosis (ADCP) is a cellular process by which antibody-opsonized targets (pathogens or cells) activate the Fc receptors on the surface of phagocytes to induce phagocytosis, resulting in internalization and degradation of pathogens or target cells through phagosome acidification. Besides NK cells-mediated antibody-dependent cellular cytotoxicity (ADCC), tumor-infiltrated monocytes and macrophages can directly kill tumor cells in the presence of tumor antigen-specific antibodies through ADCP, representing another attractive strategy for cancer immunotherapy. Even though several methods have been developed to measure ADCP, an automated and high-throughput quantitative assay should offer highly desirable advantages for drug discovery. In this study we established a new ADCP assay to identify therapeutical monoclonal antibodies (mAbs) that facilitate macrophages phagocytosis of live target cells. We used Incucyte, an imaging system for live cell analysis. By labeling the live target cells with a pH sensitive dye (pHrodo), we successfully monitored the ADCP in real time. We demonstrated that our image-based assay is robust and quantitative, suitable for screening and characterization of therapeutical mAbs that directly kill target cells through ADCP. Furthermore, we found different subtypes of macrophages have distinct ADCP activities using both mouse and human primary macrophages differentiated in vitro. By studying various mAbs with mutations in their Fc regions using our assay, we showed that the variants with increased binding to Fc gamma receptors (FcγRs) have enhanced ADCP activities., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
6. Preclinical and translational pharmacology of afucosylated anti-CCR8 antibody for depletion of tumour-infiltrating regulatory T cells.
- Author
-
Gampa G, Spinosa P, Getz J, Zhong Y, Halpern W, Esen E, Davies J, Chou C, Kwong M, Wang Y, Arenzana TL, Shivva V, Huseni M, Hsieh R, Schartner J, Koerber JT, Rutz S, and Hosseini I
- Subjects
- Animals, Humans, Mice, Female, Male, Translational Research, Biomedical, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal administration & dosage, Neoplasms drug therapy, Neoplasms immunology, Dose-Response Relationship, Drug, Antibody-Dependent Cell Cytotoxicity drug effects, Macaca fascicularis, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory immunology, Receptors, CCR8 antagonists & inhibitors, Receptors, CCR8 immunology
- Abstract
Background and Purpose: RO7502175 is an afucosylated antibody designed to eliminate C-C motif chemokine receptor 8 (CCR8)
+ Treg cells in the tumour microenvironment through enhanced antibody-dependent cellular cytotoxicity (ADCC)., Experimental Approach: We report findings from preclinical studies characterizing pharmacology, pharmacokinetics (PK)/pharmacodynamics (PD) and safety profile of RO7502175 and discuss the translational PK/PD approach used to inform first-in-human (FiH) dosing strategy and clinical development in solid tumour indications., Key Results: RO7502175 demonstrated selective ADCC against human CCR8+ Treg cells from dissociated tumours in vitro. In cynomolgus monkeys, RO7502175 exhibited a biphasic concentration-time profile consistent with immunoglobulin G1 (IgG1) antibodies, reduced CCR8+ Treg cells in the blood, induced minimal and transient cytokine secretion, and was well tolerated with a no-observed-adverse-effect level (NOAEL) of 100 mg·kg-1 . Moreover, RO7502175 caused minimal cytokine release from peripheral blood mononuclear cells (PBMCs) in vitro. A quantitative model was developed to capture surrogate anti-murine CCR8 antibody PK/PD and tumour dynamics in mice and RO7502175 PK/PD in cynomolgus monkeys. Subsequently, the model was used to project RO7502175 human PK and receptor occupancy (RO) in patients. Because traditional approaches resulted in a low FiH dose for this molecule, even with its superior preclinical safety profile, an integrated approach based on the totality of preclinical data and modelling insights was used for starting dose selection., Conclusion and Implications: This work demonstrates a translational research strategy for collecting and utilizing relevant nonclinical data, developing a mechanistic PK/PD model and using a comprehensive approach to inform clinical study design for RO7502175., (© 2024 Genentech, Inc. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
7. Application of N-Terminal Site-Specific Biotin and Digoxigenin Conjugates to Clinical Anti-drug Antibody Assay Development.
- Author
-
Wang X, Chang W, Khosraviani M, Phung W, Peng L, Cohen S, Andrews BT, Sun Y, Davies CW, Koerber JT, Yang J, and Song A
- Subjects
- Digoxigenin chemistry, Antibodies, Amines, Biotin chemistry, Lysine
- Abstract
Biotin- and digoxigenin (DIG)-conjugated therapeutic drugs are critical reagents used for the development of anti-drug antibody (ADA) assays for the assessment of immunogenicity. The current practice of generating biotin and DIG conjugates is to label a therapeutic antibody with biotin or DIG via primary amine groups on lysine or N-terminal residues. This approach modifies lysine residues nonselectively, which can impact the ability of an ADA assay to detect those ADAs that recognize epitopes located at or near the modified lysine residue(s). The impact of the lysine modification is considered greater for therapeutic antibodies that have a limited number of lysine residues, such as the variable heavy domain of heavy chain (VHH) antibodies. In this paper, for the first time, we report the application of site-specifically conjugated biotin- and DIG-VHH reagents to clinical ADA assay development using a model molecule, VHHA. The site-specific conjugation of biotin or DIG to VHHA was achieved by using an optimized reductive alkylation approach, which enabled the majority of VHHA molecules labeled with biotin or DIG at the desirable N-terminus, thereby minimizing modification of the protein after labeling and reducing the possibility of missing detection of ADAs. Head-to-head comparison of biophysical characterization data revealed that the site-specific biotin and DIG conjugates demonstrated overall superior quality to biotin- and DIG-VHHA prepared using the conventional amine coupling method, and the performance of the ADA assay developed using site-specific biotin and DIG conjugates met all acceptance criteria. The approach described here can be applied to the production of other therapeutic-protein- or antibody-based critical reagents that are used to support ligand binding assays.
- Published
- 2024
- Full Text
- View/download PDF
8. Structural basis of antibody inhibition and chemokine activation of the human CC chemokine receptor 8.
- Author
-
Sun D, Sun Y, Janezic E, Zhou T, Johnson M, Azumaya C, Noreng S, Chiu C, Seki A, Arenzana TL, Nicoludis JM, Shi Y, Wang B, Ho H, Joshi P, Tam C, Payandeh J, Comps-Agrar L, Wang J, Rutz S, Koerber JT, and Masureel M
- Subjects
- Humans, Receptors, CCR8 genetics, Ligands, Chemokine CCL1 metabolism, Antibodies, Chemokines, CC metabolism, Chemokines, CC pharmacology, Receptors, Chemokine genetics
- Abstract
The C-C motif chemokine receptor 8 (CCR8) is a class A G-protein coupled receptor that has emerged as a promising therapeutic target in cancer. Targeting CCR8 with an antibody has appeared to be an attractive therapeutic approach, but the molecular basis for chemokine-mediated activation and antibody-mediated inhibition of CCR8 are not fully elucidated. Here, we obtain an antagonist antibody against human CCR8 and determine structures of CCR8 in complex with either the antibody or the endogenous agonist ligand CCL1. Our studies reveal characteristic antibody features allowing recognition of the CCR8 extracellular loops and CCL1-CCR8 interaction modes that are distinct from other chemokine receptor - ligand pairs. Informed by these structural insights, we demonstrate that CCL1 follows a two-step, two-site binding sequence to CCR8 and that antibody-mediated inhibition of CCL1 signaling can occur by preventing the second binding event. Together, our results provide a detailed structural and mechanistic framework of CCR8 activation and inhibition that expands our molecular understanding of chemokine - receptor interactions and offers insight into the development of therapeutic antibodies targeting chemokine GPCRs., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
9. High-Throughput Analyses of Therapeutic Antibodies Using High-Field Asymmetric Waveform Ion Mobility Spectrometry Combined with SampleStream and Intact Protein Mass Spectrometry.
- Author
-
Shi RL, Dillon MA, Compton PD, Sawyer WS, Thorup JR, Kwong M, Chan P, Chiu CPC, Li R, Yadav R, Lee GY, Gober JG, Li Z, ElSohly AM, Ovacik AM, Koerber JT, Spiess C, Josephs JL, and Tran JC
- Subjects
- Mass Spectrometry methods, Chromatography, Liquid, Ions chemistry, Ion Mobility Spectrometry methods, Immunoglobulin G
- Abstract
Intact protein mass spectrometry (MS) coupled with liquid chromatography was applied to characterize the pharmacokinetics and stability profiles of therapeutic proteins. However, limitations from chromatography, including throughput and carryover, result in challenges with handling large sample numbers. Here, we combined intact protein MS with multiple front-end separations, including affinity capture, SampleStream, and high-field asymmetric waveform ion mobility spectrometry (FAIMS), to perform high-throughput and specific mass measurements of a multivalent antibody with one antigen-binding fragment (Fab) fused to an immunoglobulin G1 (IgG1) antibody. Generic affinity capture ensures the retention of both intact species 1Fab-IgG1 and the tentative degradation product IgG1. Subsequently, the analytes were directly loaded into SampleStream, where each injection occurs within ∼30 s. By separating ions prior to MS detection, FAIMS further offered improvement in signal-overnoise by ∼30% for denatured protein MS via employing compensation voltages that were optimized for different antibody species. When enhanced FAIMS transmission of 1Fab-IgG1 was employed, a qualified assay was established for spiked-in serum samples between 0.1 and 25 μg/mL, resulting in ∼10% accuracy bias and precision coefficient of variation. Selective FAIMS transmission of IgG1 as the degradation surrogate product enabled more sensitive detection of clipped species for intact 1Fab-IgG1 at 5 μg/mL in serum, generating an assay to measure 1Fab-IgG1 truncation between 2.5 and 50% with accuracy and precision below 20% bias and coefficient of variation. Our results revealed that the SampleStream-FAIMS-MS platform affords high throughput, selectivity, and sensitivity for characterizing therapeutic antibodies from complex biomatrices qualitatively and quantitatively.
- Published
- 2023
- Full Text
- View/download PDF
10. Combined PD-L1/TGFβ blockade allows expansion and differentiation of stem cell-like CD8 T cells in immune excluded tumors.
- Author
-
Castiglioni A, Yang Y, Williams K, Gogineni A, Lane RS, Wang AW, Shyer JA, Zhang Z, Mittman S, Gutierrez A, Astarita JL, Thai M, Hung J, Yang YA, Pourmohamad T, Himmels P, De Simone M, Elstrott J, Capietto AH, Cubas R, Modrusan Z, Sandoval W, Ziai J, Gould SE, Fu W, Wang Y, Koerber JT, Sanjabi S, Mellman I, Turley SJ, and Müller S
- Subjects
- Female, Animals, Mice, Cell Differentiation, Stem Cells, Interferon-gamma immunology, T-Cell Exhaustion, Mice, Inbred BALB C, Cell Line, Tumor, RNA-Seq, CD8-Positive T-Lymphocytes immunology, B7-H1 Antigen antagonists & inhibitors, Transforming Growth Factor beta antagonists & inhibitors, Immune Checkpoint Inhibitors pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms immunology
- Abstract
TGFβ signaling is associated with non-response to immune checkpoint blockade in patients with advanced cancers, particularly in the immune-excluded phenotype. While previous work demonstrates that converting tumors from excluded to inflamed phenotypes requires attenuation of PD-L1 and TGFβ signaling, the underlying cellular mechanisms remain unclear. Here, we show that TGFβ and PD-L1 restrain intratumoral stem cell-like CD8 T cell (T
SCL ) expansion and replacement of progenitor-exhausted and dysfunctional CD8 T cells with non-exhausted T effector cells in the EMT6 tumor model in female mice. Upon combined TGFβ/PD-L1 blockade IFNγhi CD8 T effector cells show enhanced motility and accumulate in the tumor. Ensuing IFNγ signaling transforms myeloid, stromal, and tumor niches to yield an immune-supportive ecosystem. Blocking IFNγ abolishes the anti-PD-L1/anti-TGFβ therapy efficacy. Our data suggest that TGFβ works with PD-L1 to prevent TSCL expansion and replacement of exhausted CD8 T cells, thereby maintaining the T cell compartment in a dysfunctional state., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
11. Mini-PBPK-Based Population Model and Covariate Analysis to Assess the Complex Pharmacokinetics and Pharmacodynamics of RO7449135, an Anti-KLK5/KLK7 Bispecific Antibody in Cynomolgus Monkeys.
- Author
-
Cai H, Tao X, Shim J, Bauer RN, Bremer M, Bu W, LaMar J, Basile R, Dere E, Nguyen T, Laing S, Chan P, Yi T, Koerber JT, Sperinde G, and Stefanich E
- Subjects
- Animals, Macaca fascicularis, Kallikreins, Skin metabolism, Antibodies, Bispecific pharmacokinetics
- Abstract
RO7449135, an anti-kallikrein (KLK)5/KLK7 bispecific antibody, is in development as a potential therapy against Netherton's syndrome (NS). In cynomolgus monkey studies, RO7449135 bound to KLK5 and KLK7, causing considerable accumulation of total KLKs, but with non-dose-proportional increase. To understand the complex PKPD, a population model with covariate analysis was developed accounting for target binding in skin and migration of bound targets from skin to blood. The covariate analysis suggested the animal batch as the categorical covariate impacting the different KLK5 synthesis rates between the repeat-dose study and single-dose study, and the dose as continuous covariate impacting the internalization rate of the binary and ternary complexes containing KLK7. To comprehend the mechanism underlying, we hypothesized that inhibition of KLK5 by RO7449135 prevented its cleavage of the pro-enzyme of KLK7 (pro-KLK7) and altered the proportion between pro-KLK7 and KLK7. Besides the pro-KLK7, RO7449135 can interact with other proteins like LEKTI through KLK7 connection in a dose-dependent manner. The different high-order complexes formed by RO7449135 interacting with pro-KLK7 or LEKTI-like proteins can be subject to faster internalization rate. Accounting for the dose and animal batch as covariates, the model-predicted free target suppression is well aligned with the visual target engagement check. The population PKPD model with covariate analysis provides the scientific input for the complex PKPD analysis, successfully predicts the target suppression in cynomolgus monkeys, and thereby can be used for the human dose projection of RO7449135., (© 2023. The Author(s), under exclusive licence to American Association of Pharmaceutical Scientists.)
- Published
- 2023
- Full Text
- View/download PDF
12. Development of an 18 F-labeled anti-human CD8 VHH for same-day immunoPET imaging.
- Author
-
Sriraman SK, Davies CW, Gill H, Kiefer JR, Yin J, Ogasawara A, Urrutia A, Javinal V, Lin Z, Seshasayee D, Abraham R, Haas P, Koth C, Marik J, Koerber JT, and Williams SP
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Positron-Emission Tomography methods, Cell Line, Tumor, Neoplasms diagnostic imaging, Single-Domain Antibodies
- Abstract
Purpose: Cancer immunotherapies (CITs) have revolutionized the treatment of certain cancers, but many patients fail to respond or relapse from current therapies, prompting the need for new CIT agents. CD8
+ T cells play a central role in the activity of many CITs, and thus, the rapid imaging of CD8+ cells could provide a critical biomarker for new CIT agents. However, existing89 Zr-labeled CD8 PET imaging reagents exhibit a long circulatory half-life and high radiation burden that limit potential applications such as same-day and longitudinal imaging., Methods: To this end, we discovered and developed a 13-kDa single-domain antibody (VHH5v2) against human CD8 to enable high-quality, same-day imaging with a reduced radiation burden. To enable sensitive and rapid imaging, we employed a site-specific conjugation strategy to introduce an18 F radiolabel to the VHH., Results: The anti-CD8 VHH, VHH5v2, demonstrated binding to a membrane distal epitope of human CD8 with a binding affinity (KD ) of 500 pM. Subsequent imaging experiments in several xenografts that express varying levels of CD8 demonstrated rapid tumor uptake and fast clearance from the blood. High-quality images were obtained within 1 h post-injection and could quantitatively differentiate the tumor models based on CD8 expression level., Conclusion: Our work reveals the potential of this anti-human CD8 VHH [18 F]F-VHH5v2 to enable rapid and specific imaging of CD8+ cells in the clinic., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2023
- Full Text
- View/download PDF
13. Systematic pharmacological analysis of agonistic and antagonistic fibroblast growth factor receptor 1 MAbs reveals a similar unique mode of action.
- Author
-
Chan J, Chan J, Shao L, Stawicki SS, Pham VC, Akita RW, Hafner M, Crocker L, Yu K, Koerber JT, Schaefer G, and Comps-Agrar L
- Subjects
- Animals, Humans, Mice, Neoplasms, Receptor, Fibroblast Growth Factor, Type 1 agonists, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Signal Transduction, Antibodies, Monoclonal pharmacology
- Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a receptor tyrosine kinase that plays a major role in developmental processes and metabolism. The dysregulation of FGFR1 through genetic aberrations leads to skeletal and metabolic diseases as well as cancer. For this reason, FGFR1 is a promising therapeutic target, yet a very challenging one due to potential on-target toxicity. More puzzling is that both agonistic and antagonistic FGFR1 antibodies are reported to exhibit similar toxicity profiles in vivo, namely weight loss. In this study, we aimed to assess and compare the mechanism of action of these molecules to better understand this apparent contradiction. By systematically comparing the binding of these antibodies and the activation or the inhibition of the major FGFR1 signaling events, we demonstrated that the molecules displayed similar properties and can behave either as an agonist or antagonist depending on the presence or the absence of the endogenous ligand. We further demonstrated that these findings translated in xenografts mice models. In addition, using time-resolved FRET and mass spectrometry analysis, we showed a functionally distinct FGFR1 active conformation in the presence of an antibody that preferentially activates the FGFR substrate 2 (FRS2)-dependent signaling pathway, demonstrating that modulating the geometry of a FGFR1 dimer can effectively change the signaling outputs and ultimately the activity of the molecule in preclinical studies. Altogether, our results highlighted how bivalent antibodies can exhibit both agonistic and antagonistic activities and have implications for targeting other receptor tyrosine kinases with antibodies., Competing Interests: Conflict of interest All authors were employees of Genentech, Inc. at the time of this study., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. Dual antibody inhibition of KLK5 and KLK7 for Netherton syndrome and atopic dermatitis.
- Author
-
Chavarria-Smith J, Chiu CPC, Jackman JK, Yin J, Zhang J, Hackney JA, Lin WY, Tyagi T, Sun Y, Tao J, Dunlap D, Morton WD, Ghodge SV, Maun HR, Li H, Hernandez-Barry H, Loyet KM, Chen E, Liu J, Tam C, Yaspan BL, Cai H, Balazs M, Arron JR, Li J, Wittwer AJ, Pappu R, Austin CD, Lee WP, Lazarus RA, Sudhamsu J, Koerber JT, and Yi T
- Subjects
- Mice, Humans, Animals, Serine Peptidase Inhibitor Kazal-Type 5 metabolism, Epidermis pathology, Antibodies metabolism, Kallikreins metabolism, Netherton Syndrome genetics, Netherton Syndrome metabolism, Netherton Syndrome pathology, Dermatitis, Atopic pathology, Skin Diseases metabolism
- Abstract
The epidermis is a barrier that prevents water loss while keeping harmful substances from penetrating the host. The impermeable cornified layer of the stratum corneum is maintained by balancing continuous turnover driven by epidermal basal cell proliferation, suprabasal cell differentiation, and corneal shedding. The epidermal desquamation process is tightly regulated by balance of the activities of serine proteases of the Kallikrein-related peptidases (KLK) family and their cognate inhibitor lymphoepithelial Kazal type-related inhibitor (LEKTI), which is encoded by the serine peptidase inhibitor Kazal type 5 gene. Imbalance of proteolytic activity caused by a deficiency of LEKTI leads to excessive desquamation due to increased activities of KLK5, KLK7, and KLK14 and results in Netherton syndrome (NS), a debilitating condition with an unmet clinical need. Increased activity of KLKs may also be pathological in other dermatoses such as atopic dermatitis (AD). Here, we describe the discovery of inhibitory antibodies against murine KLK5 and KLK7 that could compensate for the deficiency of LEKTI in NS. These antibodies are protective in mouse models of NS and AD and, when combined, promote improved skin barrier integrity and reduced inflammation. To translate these findings, we engineered a humanized bispecific antibody capable of potent inhibition of human KLK5 and KLK7. A crystal structure of KLK5 bound to the inhibitory Fab revealed that the antibody binds distal to its active site and uses a relatively unappreciated allosteric inhibition mechanism. Treatment with the bispecific anti-KLK5/7 antibody represents a promising therapy for clinical development in NS and other inflammatory dermatoses.
- Published
- 2022
- Full Text
- View/download PDF
15. Structure of the core human NADPH oxidase NOX2.
- Author
-
Noreng S, Ota N, Sun Y, Ho H, Johnson M, Arthur CP, Schneider K, Lehoux I, Davies CW, Mortara K, Wong K, Seshasayee D, Masureel M, Payandeh J, Yi T, and Koerber JT
- Subjects
- Humans, Immunoglobulin Fragments, NADPH Oxidase 2 genetics, NADPH Oxidase 2 metabolism, Reactive Oxygen Species metabolism, NADPH Oxidases metabolism, Superoxides metabolism
- Abstract
NOX2 is the prototypical member of the NADPH oxidase NOX superfamily and produces superoxide (O
2 •- ), a key reactive oxygen species (ROS) that is essential in innate and adaptive immunity. Mutations that lead to deficiency in NOX2 activity correlate with increased susceptibility to bacterial and fungal infections, resulting in chronic granulomatous disease. The core of NOX2 is formed by a heterodimeric transmembrane complex composed of NOX2 (formerly gp91) and p22, but a detailed description of its structural architecture is lacking. Here, we present the structure of the human NOX2 core complex bound to a selective anti-NOX2 antibody fragment. The core complex reveals an intricate extracellular topology of NOX2, a four-transmembrane fold of the p22 subunit, and an extensive transmembrane interface which provides insights into NOX2 assembly and activation. Functional assays uncover an inhibitory activity of the 7G5 antibody mediated by internalization-dependent and internalization-independent mechanisms. Overall, our results provide insights into the NOX2 core complex architecture, disease-causing mutations, and potential avenues for selective NOX2 pharmacological modulation., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
16. Genome-wide association study identifies kallikrein 5 in type 2 inflammation-low asthma.
- Author
-
Jackman JK, Stockwell A, Choy DF, Xie MM, Lu P, Jia G, Li H, Abbas AR, Bronson PG, Lin WY, Chiu CPC, Maun HR, Roose-Girma M, Tam L, Zhang J, Modrusan Z, Graham RR, Behrens TW, White SR, Naureckas T, Ober C, Ferreira M, Sedlacek R, Wu J, Lee WP, Lazarus RA, Koerber JT, Arron JR, Yaspan BL, and Yi T
- Subjects
- Antibodies, Neutralizing genetics, Chemokines genetics, Cytokines metabolism, Humans, Inflammation genetics, Interleukin-13 genetics, Interleukin-4 genetics, Kallikreins genetics, Kallikreins metabolism, Asthma genetics, Genome-Wide Association Study
- Abstract
Background: Clinical studies of type 2 (T2) cytokine-related neutralizing antibodies in asthma have identified a substantial subset of patients with low levels of T2 inflammation who do not benefit from T2 cytokine neutralizing antibody treatment. Non-T2 mechanisms are poorly understood in asthma but represent a redefined unmet medical need., Objective: We sought to gain a better understanding of genetic contributions to T2-low asthma., Methods: We utilized an unbiased genome-wide association study of patients with moderate to severe asthma stratified by T2 serum biomarker periostin. We also performed additional expression and biological analysis for the top genetic hits., Results: We identified a novel protective single nucleotide polymorphism at chr19q13.41, which is selectively associated with T2-low asthma and establishes Kallikrein-related peptidase 5 (KLK5) as the causal gene mediating this association. Heterozygous carriers of the single nucleotide polymorphisms have reduced KLK5 expression. KLK5 is secreted by human bronchial epithelial cells and elevated in asthma bronchial alveolar lavage. T2 cytokines IL-4 and IL-13 downregulate KLK5 in human bronchial epithelial cells. KLK5, dependent on its catalytic function, induces epithelial chemokine/cytokine expression. Finally, overexpression of KLK5 in airway or lack of an endogenous KLK5 inhibitor, SPINK5, leads to spontaneous airway neutrophilic inflammation., Conclusion: Our data identify KLK5 to be the causal gene at a novel locus at chr19q13.41 associated with T2-low asthma., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. Development of an SPR-based binding assay for characterization of anti-CD20 antibodies to CD20 expressed on extracellular vesicles.
- Author
-
Wang X, Phan MM, Sun Y, Koerber JT, Ho H, Chen Y, and Yang J
- Subjects
- Antigens, CD20, Membrane Proteins, Rituximab, Extracellular Vesicles metabolism, Surface Plasmon Resonance
- Abstract
Characterization of anti-CD20 antibody binding to CD20 is critical to development of anti-CD20 therapeutics. While SPR is widely used to characterize binding of therapeutics to their targets, its application to the characterization of anti-CD20 therapeutics has been limited by the challenges of obtaining recombinant or native full-length CD20 suitable for ligand binding assays. Extracellular vesicles (EVs) are nanoparticles naturally released from cells that provide a favorable microenvironment for membrane proteins such as CD20 to maintain proper conformation and activity. Here, we report a novel SPR-based assay that enables elucidation of binding kinetics and affinity measurements for anti-CD20 antibody binding to EV-expressed CD20. Our SPR assay is label-free, easy to perform, and demonstrates specific interaction of rituximab and obinutuzumab to CD20 expressed on EVs. The SPR assay revealed that rituximab and obinutuzumab have different binding kinetics and mechanisms to CD20 although both bind to CD20 with high affinity. Our results are consistent with existing literature and verified the validity of this method. The detailed binding kinetics information may also contribute to a better understanding of the interaction between these two antibodies and CD20. Moreover, our method provides a platform with which to characterize other therapeutic antibodies binding to EV-expressed membrane proteins., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. Conformation-locking antibodies for the discovery and characterization of KRAS inhibitors.
- Author
-
Davies CW, Oh AJ, Mroue R, Steffek M, Bruning JM, Xiao Y, Feng S, Jayakar S, Chan E, Arumugam V, Uribe SC, Drummond J, Frommlet A, Lu C, Franke Y, Merchant M, Koeppen H, Quinn JG, Malhotra S, Do S, Gazzard L, Purkey HE, Rudolph J, Mulvihill MM, Koerber JT, Wang W, and Evangelista M
- Subjects
- Humans, Ligands, Mutation, Antibodies chemistry, Neoplasms, Proto-Oncogene Proteins p21(ras) antagonists & inhibitors
- Abstract
Small molecules that stabilize inactive protein conformations are an underutilized strategy for drugging dynamic or otherwise intractable proteins. To facilitate the discovery and characterization of such inhibitors, we created a screening platform to identify conformation-locking antibodies for molecular probes (CLAMPs) that distinguish and induce rare protein conformational states. Applying the approach to KRAS, we discovered CLAMPs that recognize the open conformation of KRAS
G12C stabilized by covalent inhibitors. One CLAMP enables the visualization of KRASG12C covalent modification in vivo and can be used to investigate response heterogeneity to KRASG12C inhibitors in patient tumors. A second CLAMP enhances the affinity of weak ligands binding to the KRASG12C switch II region (SWII) by stabilizing a specific conformation of KRASG12C , thereby enabling the discovery of such ligands that could serve as leads for the development of drugs in a high-throughput screen. We show that combining the complementary properties of antibodies and small molecules facilitates the study and drugging of dynamic proteins., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
19. Loss of the intracellular enzyme QPCTL limits chemokine function and reshapes myeloid infiltration to augment tumor immunity.
- Author
-
Barreira da Silva R, Leitao RM, Pechuan-Jorge X, Werneke S, Oeh J, Javinal V, Wang Y, Phung W, Everett C, Nonomiya J, Arnott D, Lu C, Hsiao YC, Koerber JT, Hötzel I, Ziai J, Modrusan Z, Pillow TH, Roose-Girma M, Schartner JM, Merchant M, Rutz S, Eidenschenk C, Mellman I, and Albert ML
- Subjects
- Animals, Immunotherapy, Leukemic Infiltration, Mice, Mice, Knockout, Monocytes, Aminoacyltransferases genetics, Aminoacyltransferases metabolism, CD8-Positive T-Lymphocytes pathology, Chemokines metabolism, Neoplasms immunology
- Abstract
Tumor-associated macrophages are composed of distinct populations arising from monocytes or tissue macrophages, with a poorly understood link to disease pathogenesis. Here, we demonstrate that mouse monocyte migration was supported by glutaminyl-peptide cyclotransferase-like (QPCTL), an intracellular enzyme that mediates N-terminal modification of several substrates, including the monocyte chemoattractants CCL2 and CCL7, protecting them from proteolytic inactivation. Knockout of Qpctl disrupted monocyte homeostasis, attenuated tumor growth and reshaped myeloid cell infiltration, with loss of monocyte-derived populations with immunosuppressive and pro-angiogenic profiles. Antibody targeting of the receptor CSF1R, which more broadly eliminates tumor-associated macrophages, reversed tumor growth inhibition in Qpctl
-/- mice and prevented lymphocyte infiltration. Modulation of QPCTL synergized with anti-PD-L1 to expand CD8+ T cells and limit tumor growth. QPCTL inhibition constitutes an effective approach for myeloid cell-targeted cancer immunotherapy., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
20. Imaging Reveals Importance of Shape and Flexibility for Glomerular Filtration of Biologics.
- Author
-
Rafidi H, Estevez A, Ferl GZ, Mandikian D, Stainton S, Sermeño L, Williams SP, Kamath AV, Koerber JT, and Boswell CA
- Subjects
- Animals, Antibodies, Bispecific administration & dosage, Antibodies, Monoclonal administration & dosage, Female, Glomerular Filtration Barrier drug effects, Humans, Immunoglobulin G classification, Mice, SCID, Mice, Antibodies, Bispecific immunology, Antibodies, Monoclonal immunology, Glomerular Filtration Barrier metabolism, Immunoglobulin G immunology, Single Photon Emission Computed Tomography Computed Tomography methods, Viral Envelope Proteins immunology
- Abstract
Advances in antibody engineering have enabled the construction of novel molecular formats in diverse shapes and sizes, providing new opportunities for cancer immunotherapeutic drug discovery while also revealing limitations in knowledge of structure-activity relationships. The current understanding of renal filtration originates largely from data reported for dextrans, IgG, albumin, and selected globular proteins. For a one-armed IgG-based T-cell imaging agent, we observed higher renal signal than typically observed for bivalent IgGs, prompting us to explore the factors governing renal filtration of biologics. We constructed a small representative library of IgG-like formats with varied shapes and hinge flexibilities falling broadly into two categories: branched molecules including bivalent IgG and (scFv)
2 Fc, and nonbranched molecules including one-armed IgG, one-armed IgG with stacked Fab, and one-armed IgG with a rigid IgA2 hinge. Transmission electron microscopy revealed Y-shaped structures for the branched molecules and pseudo-linear structures for the nonbranched molecules. Single-photon emission CT imaging, autoradiography, and tissue harvest studies demonstrated higher renal uptake and catabolism for nonbranched molecules relative to branched molecules. Among the nonbranched molecules, the one-armed IgG with rigid IgA2 hinge molecule demonstrated higher kidney uptake and decreased systemic exposure relative to molecules with a more flexible hinge. Our results show that differences in shape and hinge flexibility drive the increased glomerular filtration of one-armed relative to bivalent antibodies and highlight the practical advantages of using imaging to assess renal filtration properties. These findings are particularly relevant for T-cell-dependent bispecific molecules, many of which have nonstandard antibody structures., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
21. Antibody toolkit reveals N-terminally ubiquitinated substrates of UBE2W.
- Author
-
Davies CW, Vidal SE, Phu L, Sudhamsu J, Hinkle TB, Chan Rosenberg S, Schumacher FR, Zeng YJ, Schwerdtfeger C, Peterson AS, Lill JR, Rose CM, Shaw AS, Wertz IE, Kirkpatrick DS, and Koerber JT
- Subjects
- Amino Acid Sequence, Animals, Antibodies immunology, Cells, Cultured, Crystallography, X-Ray methods, Humans, Mass Spectrometry methods, Protein Binding, Proteomics methods, Rabbits, Ubiquitin-Conjugating Enzymes chemistry, Ubiquitin-Conjugating Enzymes immunology, Ubiquitination, Antibodies chemistry, Peptides chemistry, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitinated Proteins metabolism
- Abstract
The ubiquitin conjugating enzyme UBE2W catalyzes non-canonical ubiquitination on the N-termini of proteins, although its substrate repertoire remains unclear. To identify endogenous N-terminally-ubiquitinated substrates, we discover four monoclonal antibodies that selectively recognize tryptic peptides with an N-terminal diglycine remnant, corresponding to sites of N-terminal ubiquitination. Importantly, these antibodies do not recognize isopeptide-linked diglycine (ubiquitin) modifications on lysine. We solve the structure of one such antibody bound to a Gly-Gly-Met peptide to reveal the molecular basis for its selective recognition. We use these antibodies in conjunction with mass spectrometry proteomics to map N-terminal ubiquitination sites on endogenous substrates of UBE2W. These substrates include UCHL1 and UCHL5, where N-terminal ubiquitination distinctly alters deubiquitinase (DUB) activity. This work describes an antibody toolkit for enrichment and global profiling of endogenous N-terminal ubiquitination sites, while revealing functionally relevant substrates of UBE2W., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
22. Homeostatic functions of monocytes and interstitial lung macrophages are regulated via collagen domain-binding receptor LAIR1.
- Author
-
Keerthivasan S, Şenbabaoğlu Y, Martinez-Martin N, Husain B, Verschueren E, Wong A, Yang YA, Sun Y, Pham V, Hinkle T, Oei Y, Madireddi S, Corpuz R, Tam L, Carlisle S, Roose-Girma M, Modrusan Z, Ye Z, Koerber JT, and Turley SJ
- Subjects
- Animals, Apoptosis physiology, Bone Marrow metabolism, Bone Marrow pathology, COS Cells, Cell Differentiation physiology, Cell Line, Cell Line, Tumor, Cell Lineage physiology, Cell Proliferation physiology, Chlorocebus aethiops, Female, Humans, Lung pathology, Macrophages, Alveolar pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes pathology, Myeloid Cells metabolism, Myeloid Cells pathology, Neoplasm Metastasis pathology, Proteomics methods, Signal Transduction physiology, Homeostasis physiology, Lung metabolism, Macrophages, Alveolar metabolism, Monocytes metabolism, Receptors, Immunologic metabolism
- Abstract
Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1
-/- mice had reduced frequencies of Ly6C- monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease., Competing Interests: Declarations of interests All authors are stockholders of Genentech/Roche except B.H., A.W., E.V., and S.C., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
23. Discovery of a caspase cleavage motif antibody reveals insights into noncanonical inflammasome function.
- Author
-
Davies CW, Stowe I, Phung QT, Ho H, Bakalarski CE, Gupta A, Zhang Y, Lill JR, Payandeh J, Kayagaki N, and Koerber JT
- Subjects
- Amino Acid Sequence, Caspases chemistry, Models, Molecular, Peptides chemistry, Peptides metabolism, Protein Binding, Protein Interaction Domains and Motifs, Proteolysis, Signal Transduction, Structure-Activity Relationship, Amino Acid Motifs, Antibodies chemistry, Antibodies metabolism, Binding Sites, Caspases metabolism, Inflammasomes metabolism
- Abstract
Inflammasomes sense a number of pathogen and host damage signals to initiate a signaling cascade that triggers inflammatory cell death, termed pyroptosis. The inflammatory caspases (1/4/5/11) are the key effectors of this process through cleavage and activation of the pore-forming protein gasdermin D. Caspase-1 also activates proinflammatory interleukins, IL-1β and IL-18, via proteolysis. However, compared to the well-studied apoptotic caspases, the identity of substrates and therefore biological functions of the inflammatory caspases remain limited. Here, we construct, validate, and apply an antibody toolset for direct detection of neo-C termini generated by inflammatory caspase proteolysis. By combining rabbit immune phage display with a set of degenerate and defined target peptides, we discovered two monoclonal antibodies that bind peptides with a similar degenerate recognition motif as the inflammatory caspases without recognizing the canonical apoptotic caspase recognition motif. Crystal structure analyses revealed the molecular basis of this strong yet paradoxical degenerate mode of peptide recognition. One antibody selectively immunoprecipitated cleaved forms of known and unknown inflammatory caspase substrates, allowing the identification of over 300 putative substrates of the caspase-4 noncanonical inflammasome, including caspase-7. This dataset will provide a path toward developing blood-based biomarkers of inflammasome activation. Overall, our study establishes tools to discover and detect inflammatory caspase substrates and functions, provides a workflow for designing antibody reagents to study cell signaling, and extends the growing evidence of biological cross talk between the apoptotic and inflammatory caspases., Competing Interests: Competing interest statement: All authors are employees of Genentech, Inc.
- Published
- 2021
- Full Text
- View/download PDF
24. Preclinical optimization of Ly6E-targeted ADCs for increased durability and efficacy of anti-tumor response.
- Author
-
Dela Cruz Chuh J, Go M, Chen Y, Guo J, Rafidi H, Mandikian D, Sun Y, Lin Z, Schneider K, Zhang P, Vij R, Sharpnack D, Chan P, de la Cruz C, Sadowsky J, Seshasayee D, Koerber JT, Pillow TH, Phillips GD, Rowntree RK, Boswell CA, Kozak KR, Polson AG, Polakis P, Yu SF, Dragovich PS, and Agard NJ
- Subjects
- Animals, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal pharmacology, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Antineoplastic Agents, Immunological pharmacokinetics, Antineoplastic Agents, Immunological pharmacology, Cell Line, Tumor, Cell Survival drug effects, Cell Survival immunology, Female, GPI-Linked Proteins immunology, HEK293 Cells, Humans, Immunoconjugates pharmacokinetics, Immunoconjugates pharmacology, Mice, SCID, Rats, Sprague-Dawley, Tumor Burden drug effects, Tumor Burden immunology, Mice, Rats, Antibodies, Monoclonal immunology, Antigens, Surface immunology, Antineoplastic Agents immunology, Immunoconjugates immunology, Oligopeptides immunology, Xenograft Model Antitumor Assays methods
- Abstract
Early success with brentuximab vedotin in treating classical Hodgkin lymphoma spurred an influx of at least 20 monomethyl auristatin E (MMAE) antibody-drug conjugates (ADCs) into clinical trials. While three MMAE-ADCs have been approved, most of these conjugates are no longer being investigated in clinical trials. Some auristatin conjugates show limited or no efficacy at tolerated doses, but even for drugs driving initial remissions, tumor regrowth and metastasis often rapidly occur. Here we describe the development of second-generation therapeutic ADCs targeting Lymphocyte antigen 6E (Ly6E) where the tubulin polymerization inhibitor MMAE (Compound 1 ) is replaced with DNA-damaging agents intended to drive increased durability of response. Comparison of a seco -cyclopropyl benzoindol-4-one (CBI)-dimer (compound 2 ) to MMAE showed increased potency, activity across more cell lines, and resistance to efflux by P -glycoprotein, a drug transporter commonly upregulated in tumors. Both anti-Ly6E-CBI and -MMAE conjugates drove single-dose efficacy in xenograft and patient-derived xenograft models, but seco- CBI-dimer conjugates showed reduced tumor outgrowth following multiple weeks of treatment, suggesting that they are less susceptible to developing resistance. In parallel, we explored approaches to optimize the targeting antibody. In contrast to immunization with recombinant Ly6E or Ly6E DNA, immunization with virus-like particles generated a high-affinity anti-Ly6E antibody. Conjugates to this antibody improve efficacy versus a previous clinical candidate both in vitro and in vivo with multiple cytotoxics. Conjugation of compound 2 to the second-generation antibody results in a substantially improved ADC with promising preclinical efficacy.
- Published
- 2021
- Full Text
- View/download PDF
25. Bivalent antibody pliers inhibit β-tryptase by an allosteric mechanism dependent on the IgG hinge.
- Author
-
Maun HR, Vij R, Walters BT, Morando A, Jackman JK, Wu P, Estevez A, Chen X, Franke Y, Lipari MT, Dennis MS, Kirchhofer D, Ciferri C, Loyet KM, Yi T, Eigenbrot C, Lazarus RA, and Koerber JT
- Subjects
- Allosteric Regulation drug effects, Amino Acid Sequence, Heparin pharmacology, Humans, Immunoglobulin Fab Fragments metabolism, Immunoglobulin G chemistry, Models, Molecular, Mutant Proteins chemistry, Protein Binding drug effects, Protein Multimerization, Tryptases chemistry, Antibodies, Monoclonal metabolism, Immunoglobulin G metabolism, Tryptases metabolism
- Abstract
Human β-tryptase, a tetrameric trypsin-like serine protease, is an important mediator of allergic inflammatory responses in asthma. Antibodies generally inhibit proteases by blocking substrate access by binding to active sites or exosites or by allosteric modulation. The bivalency of IgG antibodies can increase potency via avidity, but has never been described as essential for activity. Here we report an inhibitory anti-tryptase IgG antibody with a bivalency-driven mechanism of action. Using biochemical and structural data, we determine that four Fabs simultaneously occupy four exosites on the β-tryptase tetramer, inducing allosteric changes at the small interface. In the presence of heparin, the monovalent Fab shows essentially no inhibition, whereas the bivalent IgG fully inhibits β-tryptase activity in a hinge-dependent manner. Our results suggest a model where the bivalent IgG acts akin to molecular pliers, pulling the tetramer apart into inactive β-tryptase monomers, and may provide an alternative strategy for antibody engineering.
- Published
- 2020
- Full Text
- View/download PDF
26. Development of a specific immunoassay to selectively measure active tryptase in airway samples.
- Author
-
Sperinde G, Bremer M, Maun HR, Baruch A, Lazarus RA, Koerber JT, Vij R, Yi T, Fischer SK, and Staton T
- Subjects
- Humans, Immunoassay methods, Immunologic Tests methods, Mast Cells pathology, Tryptases metabolism
- Abstract
Aim: Tryptase is a tetrameric trypsin-like serine protease contained within the secretory granules of mast cells and is an important mediator of allergic inflammatory responses in respiratory diseases. Detection of active tryptase in the airway may provide important information about asthma and other respiratory diseases. Materials & Methods: An activity based probe has been incorported within an immunoassay to allow for measurement of active tryptase in human tissues. Results: A specific Simoa immunoassay to measure active tryptase in nasosorption samples was developed and qualified using an activity-based probe label and a specific antitryptase capture antibody. Conclusion: The assay was capable of measuring active tryptase in human samples, which will enable evaluation of the role of tryptase proteolytic activity in human disease.
- Published
- 2020
- Full Text
- View/download PDF
27. Effect of Modulating FcRn Binding on Direct and Pretargeted Tumor Uptake of Full-length Antibodies.
- Author
-
Nazarova L, Rafidi H, Mandikian D, Ferl GZ, Koerber JT, Davies CW, Ulufatu S, Ho J, Lau J, Yu SF, Ernst J, Sadowsky JD, and Boswell CA
- Subjects
- Animals, Breast Neoplasms diagnostic imaging, Breast Neoplasms metabolism, Breast Neoplasms pathology, Click Chemistry, Female, Image Processing, Computer-Assisted, Mice, Mice, SCID, Receptor, ErbB-2 metabolism, Single Photon Emission Computed Tomography Computed Tomography, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal pharmacology, Breast Neoplasms drug therapy, Histocompatibility Antigens Class I metabolism, Radiopharmaceuticals metabolism, Receptors, Fc metabolism
- Abstract
Full-length antibodies lack ideal pharmacokinetic properties for rapid targeted imaging, prompting the pursuit of smaller peptides and fragments. Nevertheless, studying the disposition properties of antibody-based imaging agents can provide critical insight into the pharmacology of their therapeutic counterparts, particularly for those coupled with potent payloads. Here, we evaluate modulation of binding to the neonatal Fc receptor (FcRn) as a protein engineering-based pharmacologic strategy to minimize the overall blood pool background with directly labeled antibodies and undesirable systemic click reaction of radiolabeled tetrazine with circulating pretargeted trans -cyclooctene (TCO)-modified antibodies. Noninvasive SPECT imaging of mice bearing HER2-expressing xenografts was performed both directly (
111 In-labeled antibody) and indirectly (pretargeted TCO-modified antibody followed by111 In-labeled tetrazine). Pharmacokinetic modulation of antibodies was achieved by two distinct methods: Fc engineering to reduce binding affinity to FcRn, and delayed administration of an antibody that competes with binding to FcRn. Tumor imaging with directly labeled antibodies was feasible in the absence of FcRn binding, rapidly attaining high tumor-to-blood ratios, but accompanied by moderate liver and spleen uptake. Pretargeted imaging of tumors with non-FcRn-binding antibody was also feasible, but systemic click reaction still occurred, albeit at lower levels than with parental antibody. Our findings demonstrate that FcRn binding impairment of full-length IgG antibodies moderately lowers tumor accumulation of radioactivity, and shifts background activity from blood pool to liver and spleen. Furthermore, reduction of FcRn binding did not eliminate systemic click reaction, but yielded greater improvements in tumor-to-blood ratio when imaging with directly labeled antibodies than with pretargeting., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
28. Structure of CD20 in complex with the therapeutic monoclonal antibody rituximab.
- Author
-
Rougé L, Chiang N, Steffek M, Kugel C, Croll TI, Tam C, Estevez A, Arthur CP, Koth CM, Ciferri C, Kraft E, Payandeh J, Nakamura G, Koerber JT, and Rohou A
- Subjects
- Antigens, CD20 immunology, Complement System Proteins immunology, Cryoelectron Microscopy, Humans, Immunoglobulin Fab Fragments chemistry, Protein Conformation, Protein Multimerization, Rituximab immunology, Antigens, CD20 chemistry, Rituximab chemistry
- Abstract
Cluster of differentiation 20 (CD20) is a B cell membrane protein that is targeted by monoclonal antibodies for the treatment of malignancies and autoimmune disorders but whose structure and function are unknown. Rituximab (RTX) has been in clinical use for two decades, but how it activates complement to kill B cells remains poorly understood. We obtained a structure of CD20 in complex with RTX, revealing CD20 as a compact double-barrel dimer bound by two RTX antigen-binding fragments (Fabs), each of which engages a composite epitope and an extensive homotypic Fab:Fab interface. Our data suggest that RTX cross-links CD20 into circular assemblies and lead to a structural model for complement recruitment. Our results further highlight the potential relevance of homotypic Fab:Fab interactions in targeting oligomeric cell-surface markers., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
29. An Allosteric Anti-tryptase Antibody for the Treatment of Mast Cell-Mediated Severe Asthma.
- Author
-
Maun HR, Jackman JK, Choy DF, Loyet KM, Staton TL, Jia G, Dressen A, Hackney JA, Bremer M, Walters BT, Vij R, Chen X, Trivedi NN, Morando A, Lipari MT, Franke Y, Wu X, Zhang J, Liu J, Wu P, Chang D, Orozco LD, Christensen E, Wong M, Corpuz R, Hang JQ, Lutman J, Sukumaran S, Wu Y, Ubhayakar S, Liang X, Schwartz LB, Babina M, Woodruff PG, Fahy JV, Ahuja R, Caughey GH, Kusi A, Dennis MS, Eigenbrot C, Kirchhofer D, Austin CD, Wu LC, Koerber JT, Lee WP, Yaspan BL, Alatsis KR, Arron JR, Lazarus RA, and Yi T
- Published
- 2020
- Full Text
- View/download PDF
30. An Allosteric Anti-tryptase Antibody for the Treatment of Mast Cell-Mediated Severe Asthma.
- Author
-
Maun HR, Jackman JK, Choy DF, Loyet KM, Staton TL, Jia G, Dressen A, Hackney JA, Bremer M, Walters BT, Vij R, Chen X, Trivedi NN, Morando A, Lipari MT, Franke Y, Wu X, Zhang J, Liu J, Wu P, Chang D, Orozco LD, Christensen E, Wong M, Corpuz R, Hang JQ, Lutman J, Sukumaran S, Wu Y, Ubhayakar S, Liang X, Schwartz LB, Babina M, Woodruff PG, Fahy JV, Ahuja R, Caughey GH, Kusi A, Dennis MS, Eigenbrot C, Kirchhofer D, Austin CD, Wu LC, Koerber JT, Lee WP, Yaspan BL, Alatsis KR, Arron JR, Lazarus RA, and Yi T
- Subjects
- Adolescent, Allosteric Regulation immunology, Animals, Cell Line, Female, Humans, Macaca fascicularis, Male, Mice, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, SCID, Rabbits, Antibodies, Monoclonal, Humanized therapeutic use, Asthma therapy, Mast Cells enzymology, Mast Cells immunology, Tryptases antagonists & inhibitors, Tryptases immunology
- Abstract
Severe asthma patients with low type 2 inflammation derive less clinical benefit from therapies targeting type 2 cytokines and represent an unmet need. We show that mast cell tryptase is elevated in severe asthma patients independent of type 2 biomarker status. Active β-tryptase allele count correlates with blood tryptase levels, and asthma patients carrying more active alleles benefit less from anti-IgE treatment. We generated a noncompetitive inhibitory antibody against human β-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a β-tryptase/antibody complex coupled with biochemical studies reveal the molecular basis for allosteric destabilization of small and large interfaces required for tetramerization. This anti-tryptase antibody potently blocks tryptase enzymatic activity in a humanized mouse model, reducing IgE-mediated systemic anaphylaxis, and inhibits airway tryptase in Ascaris-sensitized cynomolgus monkeys with favorable pharmacokinetics. These data provide a foundation for developing anti-tryptase as a clinical therapy for severe asthma., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
31. Massive antibody discovery used to probe structure-function relationships of the essential outer membrane protein LptD.
- Author
-
Storek KM, Chan J, Vij R, Chiang N, Lin Z, Bevers J 3rd, Koth CM, Vernes JM, Meng YG, Yin J, Wallweber H, Dalmas O, Shriver S, Tam C, Schneider K, Seshasayee D, Nakamura G, Smith PA, Payandeh J, Koerber JT, Comps-Agrar L, and Rutherford ST
- Subjects
- Animals, Anti-Bacterial Agents pharmacology, Binding Sites, Epitope Mapping, Epitopes metabolism, Escherichia coli growth & development, Escherichia coli metabolism, Mice, Inbred BALB C, Protein Structure, Secondary, Rats, Sprague-Dawley, Structure-Activity Relationship, Antibodies, Monoclonal metabolism, Bacterial Outer Membrane Proteins chemistry, Bacterial Outer Membrane Proteins metabolism, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism
- Abstract
Outer membrane proteins (OMPs) in Gram-negative bacteria dictate permeability of metabolites, antibiotics, and toxins. Elucidating the structure-function relationships governing OMPs within native membrane environments remains challenging. We constructed a diverse library of >3000 monoclonal antibodies to assess the roles of extracellular loops (ECLs) in LptD, an essential OMP that inserts lipopolysaccharide into the outer membrane of Escherichia coli . Epitope binning and mapping experiments with LptD-loop-deletion mutants demonstrated that 7 of the 13 ECLs are targeted by antibodies. Only ECLs inaccessible to antibodies were required for the structure or function of LptD. Our results suggest that antibody-accessible loops evolved to protect key extracellular regions of LptD, but are themselves dispensable. Supporting this hypothesis, no α-LptD antibody interfered with essential functions of LptD. Our experimental workflow enables structure-function studies of OMPs in native cellular environments, provides unexpected insight into LptD, and presents a method to assess the therapeutic potential of antibody targeting., Competing Interests: KS, JC, RV, NC, ZL, JB, CK, JV, YM, JY, HW, OD, SS, CT, KS, DS, GN, PS, JP, JK, LC, SR Employee of Genentech, Inc, a member of the Roche Group, and shareholder in Roche., (© 2019, Storek et al.)
- Published
- 2019
- Full Text
- View/download PDF
32. The Escherichia coli β-Barrel Assembly Machinery Is Sensitized to Perturbations under High Membrane Fluidity.
- Author
-
Storek KM, Vij R, Sun D, Smith PA, Koerber JT, and Rutherford ST
- Subjects
- Amino Acid Substitution, Bacterial Outer Membrane Proteins genetics, DNA Mutational Analysis, Escherichia coli genetics, Escherichia coli Proteins genetics, Bacterial Outer Membrane Proteins metabolism, Escherichia coli enzymology, Escherichia coli Proteins metabolism, Lipoproteins metabolism, Membrane Fluidity, Protein Folding, Protein Multimerization
- Abstract
Integral β-barrel membrane proteins are folded and inserted into the Gram-negative bacterial outer membrane by the β-barrel assembly machine (BAM). This essential complex, composed of a β-barrel protein, BamA, and four lipoproteins, BamB, BamC, BamD, and BamE, resides in the outer membrane, a unique asymmetrical lipid bilayer that is difficult to recapitulate in vitro Thus, the probing of BAM function in living cells is critical to fully understand the mechanism of β-barrel folding. We recently identified an anti-BamA monoclonal antibody, MAB1, that is a specific and potent inhibitor of BamA function. Here, we show that the inhibitory effect of MAB1 is enhanced when BAM function is perturbed by either lowering the level of BamA or removing the nonessential BAM lipoproteins, BamB, BamC, or BamE. The disruption of BAM reduces BamA activity, increases outer membrane (OM) fluidity, and activates the σ
E stress response, suggesting the OM environment and BAM function are interconnected. Consistent with this idea, an increase in the membrane fluidity through changes in the growth environment or alterations to the lipopolysaccharide in the outer membrane is sufficient to provide resistance to MAB1 and enable the BAM to tolerate these perturbations. Amino acid substitutions in BamA at positions in the outer membrane spanning region or the periplasmic space remote from the extracellular MAB1 binding site also provide resistance to the inhibitory antibody. Our data highlight that the outer membrane environment is a critical determinant in the efficient and productive folding of β-barrel membrane proteins by BamA. IMPORTANCE BamA is an essential component of the β-barrel assembly machine (BAM) in the outer membranes of Gram-negative bacteria. We have used a recently described inhibitory anti-BamA antibody, MAB1, to identify the molecular requirements for BAM function. Resistance to this antibody can be achieved through changes to the outer membrane or by amino acid substitutions in BamA that allosterically affect the response to MAB1. Sensitivity to MAB1 is achieved by perturbing BAM function. By using MAB1 activity and functional assays as proxies for BAM function, we link outer membrane fluidity to BamA activity, demonstrating that an increase in membrane fluidity sensitizes the cells to BAM perturbations. Thus, the search for potential inhibitors of BamA function must consider the membrane environment in which β-barrel folding occurs., (Copyright © 2018 Storek et al.)- Published
- 2018
- Full Text
- View/download PDF
33. Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3.
- Author
-
Slaga D, Ellerman D, Lombana TN, Vij R, Li J, Hristopoulos M, Clark R, Johnston J, Shelton A, Mai E, Gadkar K, Lo AA, Koerber JT, Totpal K, Prell R, Lee G, Spiess C, and Junttila TT
- Subjects
- Antibodies, Bispecific immunology, Cell Line, Tumor, Humans, Immunoglobulin Fab Fragments metabolism, Immunoglobulin G metabolism, Lymphocyte Activation immunology, Protein Binding, Antibody Affinity immunology, CD3 Complex immunology, Cytotoxicity, Immunologic, Receptor, ErbB-2 immunology
- Abstract
A primary barrier to the success of T cell-recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, we developed an anti-HER2/CD3 T cell-dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab-immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell-directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
34. Analysis of the effect of promoter type and skin pretreatment on antigen expression and antibody response after gene gun-based immunization.
- Author
-
Vij R, Lin Z, Schneider K, Seshasayee D, and Koerber JT
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antigens, Viral immunology, Claudin-4 immunology, Gene Expression, HEK293 Cells, Humans, Mice, Antibodies, Viral immunology, Antigens, Viral genetics, Biolistics methods, Immunization methods, Promoter Regions, Genetic genetics, Skin
- Abstract
Monoclonal antibodies (mAbs) have enabled numerous basic research discoveries and therapeutic approaches for many protein classes. However, there still exist a number of target classes, such as multi-pass membrane proteins, for which antibody discovery is difficult, due in part to lack of high quality, recombinant protein. Here we describe the impact of several parameters on antigen expression and the development of mAbs against human claudin 4 (CLDN4), a potential multi-indication cancer target. Using gene gun-based DNA delivery and bioluminescence imaging, we optimize promoter type by comparing expression profiles of four robust in vivo promoters. In addition, we observe that most vectors rapidly lose expression, ultimately reaching almost background levels by three days post-delivery. Recognizing this limitation, we next explored skin pretreatment strategies as an orthogonal method to further boost the efficiency of mAb generation. We show that SDS pretreatment can boost antigen expression, but fails to significantly increase mAb discovery efficiency. In contrast, we find that sandpaper pretreatment yields 5-fold more FACS+ anti-CLDN4 hybridomas, without impacting antigen expression. Our findings coupled with other strategies to improve DNA immunizations should improve the success of mAb discovery against other challenging targets and enable the generation of critical research tools and therapeutic candidates., Competing Interests: All authors are paid employees of Genentech, Inc., which is the funder of the reported studies. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2018
- Full Text
- View/download PDF
35. A targeted boost-and-sort immunization strategy using Escherichia coli BamA identifies rare growth inhibitory antibodies.
- Author
-
Vij R, Lin Z, Chiang N, Vernes JM, Storek KM, Park S, Chan J, Meng YG, Comps-Agrar L, Luan P, Lee S, Schneider K, Bevers J 3rd, Zilberleyb I, Tam C, Koth CM, Xu M, Gill A, Auerbach MR, Smith PA, Rutherford ST, Nakamura G, Seshasayee D, Payandeh J, and Koerber JT
- Subjects
- Antibodies, Monoclonal genetics, Antibodies, Monoclonal isolation & purification, Bacterial Outer Membrane Proteins immunology, Escherichia coli immunology, Escherichia coli Proteins immunology, Immunization, Protein Conformation, Protein Folding, Protein Transport genetics, Protein Transport immunology, Vaccination, Antibodies, Monoclonal immunology, Bacterial Outer Membrane Proteins genetics, Escherichia coli genetics, Escherichia coli Proteins genetics
- Abstract
Outer membrane proteins (OMPs) in Gram-negative bacteria are essential for a number of cellular functions including nutrient transport and drug efflux. Escherichia coli BamA is an essential component of the OMP β-barrel assembly machinery and a potential novel antibacterial target that has been proposed to undergo large (~15 Å) conformational changes. Here, we explored methods to isolate anti-BamA monoclonal antibodies (mAbs) that might alter the function of this OMP and ultimately lead to bacterial growth inhibition. We first optimized traditional immunization approaches but failed to identify mAbs that altered cell growth after screening >3000 hybridomas. We then developed a "targeted boost-and-sort" strategy that combines bacterial cell immunizations, purified BamA protein boosts, and single hybridoma cell sorting using amphipol-reconstituted BamA antigen. This unique workflow improves the discovery efficiency of FACS + mAbs by >600-fold and enabled the identification of rare anti-BamA mAbs with bacterial growth inhibitory activity in the presence of a truncated lipopolysaccharide layer. These mAbs represent novel tools for dissecting the BamA-mediated mechanism of β-barrel folding and our workflow establishes a new template for the efficient discovery of novel mAbs against other highly dynamic membrane proteins.
- Published
- 2018
- Full Text
- View/download PDF
36. Monoclonal antibody targeting the β-barrel assembly machine of Escherichia coli is bactericidal.
- Author
-
Storek KM, Auerbach MR, Shi H, Garcia NK, Sun D, Nickerson NN, Vij R, Lin Z, Chiang N, Schneider K, Wecksler AT, Skippington E, Nakamura G, Seshasayee D, Koerber JT, Payandeh J, Smith PA, and Rutherford ST
- Subjects
- Bacterial Outer Membrane Proteins immunology, Bacterial Outer Membrane Proteins metabolism, Cell Membrane drug effects, Cell Membrane immunology, Cell Membrane metabolism, Escherichia coli immunology, Escherichia coli metabolism, Escherichia coli Proteins immunology, Escherichia coli Proteins metabolism, Models, Molecular, Protein Conformation, Protein Folding, Anti-Bacterial Agents pharmacology, Antibodies, Bacterial pharmacology, Antibodies, Monoclonal pharmacology, Bacterial Outer Membrane Proteins antagonists & inhibitors, Escherichia coli drug effects, Escherichia coli Proteins antagonists & inhibitors, Membrane Fluidity drug effects
- Abstract
The folding and insertion of integral β-barrel membrane proteins into the outer membrane of Gram-negative bacteria is required for viability and bacterial pathogenesis. Unfortunately, the lack of selective and potent modulators to dissect β-barrel folding in vivo has hampered our understanding of this fundamental biological process. Here, we characterize a monoclonal antibody that selectively inhibits an essential component of the Escherichia coli β-barrel assembly machine, BamA. In the absence of complement or other immune factors, the unmodified antibody MAB1 demonstrates bactericidal activity against an E. coli strain with truncated LPS. Direct binding of MAB1 to an extracellular BamA epitope inhibits its β-barrel folding activity, induces periplasmic stress, disrupts outer membrane integrity, and kills bacteria. Notably, resistance to MAB1-mediated killing reveals a link between outer membrane fluidity and protein folding by BamA in vivo, underscoring the utility of this antibody for studying β-barrel membrane protein folding within a living cell. Identification of this BamA antagonist highlights the potential for new mechanisms of antibiotics to inhibit Gram-negative bacterial growth by targeting extracellular epitopes., Competing Interests: Conflict of interest statement: All authors are employees of Genentech, a member of the Roche Group, and are shareholders in Roche. Study was supported by internal Genentech funds, and the funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
37. An improved single-chain Fab platform for efficient display and recombinant expression.
- Author
-
Koerber JT, Hornsby MJ, and Wells JA
- Subjects
- Amino Acid Sequence, Animals, Bacteria genetics, Bacteriophages genetics, Complementarity Determining Regions chemistry, Complementarity Determining Regions genetics, Enzyme-Linked Immunosorbent Assay, Genetic Vectors, HEK293 Cells, Humans, Immunoglobulin Fab Fragments genetics, Mammals genetics, Molecular Sequence Data, Promoter Regions, Genetic, Protein Conformation, Protein Processing, Post-Translational, Recombinant Proteins genetics, Sequence Analysis, DNA, Single-Chain Antibodies genetics, Computer Simulation, Gene Expression, Immunoglobulin Fab Fragments chemistry, Recombinant Proteins chemistry, Single-Chain Antibodies chemistry
- Abstract
Antibody phage display libraries combined with high-throughput selections have recently demonstrated tremendous promise to create the next generation of renewable, recombinant antibodies to study proteins and their many post-translational modification states; however, many challenges still remain, such as optimized antibody scaffolds. Recently, a single-chain fragment antigen binding (Fab) (scFab) format, in which the carboxy-terminus of the light chain is linked to the amino-terminus of the heavy chain, was described to potentially combine the high display levels of a single-chain fragment variable with the high stability of purified Fabs. However, this format required removal of the interchain disulfide bond to achieve modest display levels and subsequent bacterial expression resulted in high levels of aggregated scFab, hindering further use of scFabs. Here, we developed an improved scFab format that retains the interchain disulfide bond by increasing the linker length between the light and heavy chains to improve display and bacterial expression levels to 1-3 mg/L. Furthermore, rerouting of the scFab to the co-translational signal recognition particle pathway combined with reengineering of the signal peptide sequence results in display levels 24-fold above the original scFab format and 3-fold above parent Fab levels. This optimized scFab scaffold can be easily reformatted in a single step for expression in a bacterial or mammalian host to produce stable (Tm of 81 °C), predominantly monomeric (>90%) antibodies at a high yield. Ultimately, this new scFab format will advance high-throughput antibody generation platforms to discover the next generation of research and therapeutic antibodies., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
38. Quantitative proteomics reveal a feedforward mechanism for mitochondrial PARKIN translocation and ubiquitin chain synthesis.
- Author
-
Ordureau A, Sarraf SA, Duda DM, Heo JM, Jedrychowski MP, Sviderskiy VO, Olszewski JL, Koerber JT, Xie T, Beausoleil SA, Wells JA, Gygi SP, Schulman BA, and Harper JW
- Subjects
- Feedback, Physiological, HeLa Cells, Humans, Membrane Potential, Mitochondrial, Mutation, Missense, Parkinson Disease enzymology, Phosphorylation, Protein Kinases metabolism, Protein Multimerization, Protein Transport, Proteomics, Ubiquitin-Protein Ligases genetics, Mitochondria enzymology, Polyubiquitin biosynthesis, Ubiquitin-Protein Ligases metabolism, Ubiquitination
- Abstract
Phosphorylation is often used to promote protein ubiquitylation, yet we rarely understand quantitatively how ligase activation and ubiquitin (UB) chain assembly are integrated with phosphoregulation. Here we employ quantitative proteomics and live-cell imaging to dissect individual steps in the PINK1 kinase-PARKIN UB ligase mitochondrial control pathway disrupted in Parkinson's disease. PINK1 plays a dual role by phosphorylating PARKIN on its UB-like domain and poly-UB chains on mitochondria. PARKIN activation by PINK1 produces canonical and noncanonical UB chains on mitochondria, and PARKIN-dependent chain assembly is required for accumulation of poly-phospho-UB (poly-p-UB) on mitochondria. In vitro, PINK1 directly activates PARKIN's ability to assemble canonical and noncanonical UB chains and promotes association of PARKIN with both p-UB and poly-p-UB. Our data reveal a feedforward mechanism that explains how PINK1 phosphorylation of both PARKIN and poly-UB chains synthesized by PARKIN drives a program of PARKIN recruitment and mitochondrial ubiquitylation in response to mitochondrial damage., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
39. Nature-inspired design of motif-specific antibody scaffolds.
- Author
-
Koerber JT, Thomsen ND, Hannigan BT, Degrado WF, and Wells JA
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Antibodies, Phospho-Specific chemistry, Crystallography, X-Ray, Humans, Models, Molecular, Molecular Sequence Data, Phosphopeptides chemistry, Phosphopeptides immunology, Single-Chain Antibodies immunology, Antibodies, Phospho-Specific biosynthesis, Antibody Specificity immunology, Protein Engineering
- Abstract
Aberrant changes in post-translational modifications (PTMs) such as phosphate groups underlie a majority of human diseases. However, detection and quantification of PTMs for diagnostic or biomarker applications often require PTM-specific monoclonal antibodies (mAbs), which are challenging to generate using traditional antibody-selection methods. Here we outline a general strategy for producing synthetic, PTM-specific mAbs by engineering a motif-specific 'hot spot' into an antibody scaffold. Inspired by a natural phosphate-binding motif, we designed and selected mAb scaffolds with hot spots specific for phosphoserine, phosphothreonine or phosphotyrosine. Crystal structures of the phospho-specific mAbs revealed two distinct modes of phosphoresidue recognition. Our data suggest that each hot spot functions independently of the surrounding scaffold, as phage display antibody libraries using these scaffolds yielded >50 phospho- and target-specific mAbs against 70% of target peptides. Our motif-specific scaffold strategy may provide a general solution for rapid, robust development of anti-PTM mAbs for signaling, diagnostic and therapeutic applications.
- Published
- 2013
- Full Text
- View/download PDF
40. Structural snapshots reveal distinct mechanisms of procaspase-3 and -7 activation.
- Author
-
Thomsen ND, Koerber JT, and Wells JA
- Subjects
- Apoptosis physiology, Caspase 3 genetics, Caspase 3 metabolism, Caspase 7 genetics, Caspase 7 metabolism, Enzyme Activation physiology, Enzyme Precursors genetics, Enzyme Precursors metabolism, Humans, Protein Structure, Quaternary, Protein Structure, Tertiary, Structure-Activity Relationship, Caspase 3 chemistry, Caspase 7 chemistry, Enzyme Precursors chemistry, Protein Multimerization physiology
- Abstract
Procaspase-3 (P3) and procaspase-7 (P7) are activated through proteolytic maturation to form caspase-3 (C3) and caspase-7 (C7), respectively, which serve overlapping but nonredundant roles as the executioners of apoptosis in humans. However, it is unclear if differences in P3 and P7 maturation mechanisms underlie their unique biological functions, as the structure of P3 remains unknown. Here, we report structures of P3 in a catalytically inactive conformation, structures of P3 and P7 bound to covalent peptide inhibitors that reveal the active conformation of the zymogens, and the structure of a partially matured C7:P7 heterodimer. Along with a biochemical analysis, we show that P3 is catalytically inactive and matures through a symmetric all-or-nothing process. In contrast, P7 contains latent catalytic activity and matures through an asymmetric and tiered mechanism, suggesting a lower threshold for activation. Finally, we use our structures to design a selection strategy for conformation specific antibody fragments that stimulate procaspase activity, showing that executioner procaspase conformational equilibrium can be rationally modulated. Our studies provide a structural framework that may help to explain the unique roles of these important proapoptotic enzymes, and suggest general strategies for the discovery of proenzyme activators.
- Published
- 2013
- Full Text
- View/download PDF
41. Enhanced sialic acid-dependent endocytosis explains the increased efficiency of infection of airway epithelia by a novel adeno-associated virus.
- Author
-
Dickey DD, Excoffon KJ, Koerber JT, Bergen J, Steines B, Klesney-Tait J, Schaffer DV, and Zabner J
- Subjects
- Animals, Cell Line, Cricetinae, Cricetulus, Epithelial Cells virology, Humans, Mutation, Missense, Recombination, Genetic, Swine, Dependovirus pathogenicity, Endocytosis, N-Acetylneuraminic Acid metabolism, Receptors, Virus metabolism, Virus Internalization
- Abstract
We previously used directed evolution in human airway epithelia to create adeno-associated virus 2.5T (AAV2.5T), a highly infectious chimera of AAV2 and AAV5 with one point mutation (A581T). We hypothesized that the mechanism for its increased infection may be a higher binding affinity to the surface of airway epithelia than its parent AAV5. Here, we show that, like AAV5, AAV2.5T, uses 2,3N-linked sialic acid as its primary receptor; however, AAV2.5T binds to the apical surface of human airway epithelia at higher levels and has more receptors than AAV5. Furthermore, its binding affinity is similar to that of AAV5. An alternative hypothesis is that AAV2.5T interaction with 2,3N-linked sialic acid may instead be required for cellular internalization. Consistent with this, AAV2.5T binds but fails to be internalized by CHO cells that lack surface expression of sialic acid. Moreover, whereas AAV2.5T binds similarly to human (rich in 2,3N-linked sialic acid) and pig airway epithelia (2,6N-linked sialic acid), significantly more virus was internalized by human airway. Subsequent transduction correlated with the level of internalized rather than surface-bound virus. We also found that human airway epithelia internalized significantly more AAV2.5T than AAV5. These data suggest that AAV2.5T has evolved to utilize specific 2,3N-linked sialic acid residues on the surface of airway epithelia that mediate rapid internalization and subsequent infection. Thus, sialic acid serves as not just an attachment factor but is also required for AAV2.5T internalization, possibly representing an important rate-limiting step for other viruses that use sialic acids.
- Published
- 2011
- Full Text
- View/download PDF
42. An evolved adeno-associated viral variant enhances gene delivery and gene targeting in neural stem cells.
- Author
-
Jang JH, Koerber JT, Kim JS, Asuri P, Vazin T, Bartel M, Keung A, Kwon I, Park KI, and Schaffer DV
- Subjects
- Animals, Cells, Cultured, Chromatography, Female, Fluorescent Antibody Technique, Gene Transfer Techniques, Polymerase Chain Reaction, Rats, Rats, Inbred F344, Dependovirus genetics, Neural Stem Cells virology
- Abstract
Gene delivery to, and gene targeting in, stem cells would be a highly enabling technology for basic science and biomedical application. Adeno-associated viral (AAV) vectors have demonstrated the capacity for efficient delivery to numerous cells, but their application to stem cells has been limited by low transduction efficiency. Due to their considerable advantages, however, engineering AAV delivery systems to enhance gene delivery to stem cells may have an impact in stem cell biology and therapy. Therefore, using several diverse AAV capsid libraries-including randomly mutagenized, DNA shuffled, and random peptide insertion variants-we applied directed evolution to create a "designer" AAV vector with enhanced delivery efficiency for neural stem cells (NSCs). A novel AAV variant, carrying an insertion of a selected peptide sequence on the surface of the threefold spike within the heparin-binding site, emerged from this evolution. Importantly, this evolved AAV variant mediated efficient gene delivery to rat, mouse, and human NSCs, as well as efficient gene targeting within adult NSCs, and it is thus promising for applications ranging from basic stem cell biology to clinical translation.
- Published
- 2011
- Full Text
- View/download PDF
43. Molecular evolution of adeno-associated virus for enhanced glial gene delivery.
- Author
-
Koerber JT, Klimczak R, Jang JH, Dalkara D, Flannery JG, and Schaffer DV
- Subjects
- Animals, Astrocytes metabolism, Cells, Cultured, Dependovirus growth & development, Directed Molecular Evolution, Glioblastoma genetics, Glioblastoma metabolism, Green Fluorescent Proteins metabolism, Humans, Neuroblastoma genetics, Neuroblastoma metabolism, Rats, Rats, Sprague-Dawley, Serotyping, Transduction, Genetic, Dependovirus genetics, Drug Delivery Systems, Gene Transfer Techniques, Genetic Vectors, Green Fluorescent Proteins genetics
- Abstract
The natural tropism of most viral vectors, including adeno-associated viral (AAV) vectors, leads to predominant transduction of neurons and epithelia within the central nervous system (CNS) and retina. Despite the clinical relevance of glia for homeostasis in neural tissue, and as causal contributors in genetic disorders such as Alzheimer's and amyotrophic lateral sclerosis, efforts to develop more efficient gene delivery vectors for glia have met with limited success. Recently, viral vector engineering involving high-throughput random diversification and selection has enabled the rapid creation of AAV vectors with valuable new gene delivery properties. We have engineered novel AAV variants capable of efficient glia transduction by employing directed evolution with a panel of four distinct AAV libraries, including a new semi-random peptide replacement strategy. These variants transduced both human and rat astrocytes in vitro up to 15-fold higher than their parent serotypes, and injection into the rat striatum yielded astrocyte transduction levels up to 16% of the total transduced cell population, despite the human astrocyte selection platform. Furthermore, one variant exhibited a substantial shift in tropism toward Müller glia within the retina, further highlighting the general utility of these variants for efficient glia transduction in multiple species within the CNS and retina.
- Published
- 2009
- Full Text
- View/download PDF
44. A novel adeno-associated viral variant for efficient and selective intravitreal transduction of rat Müller cells.
- Author
-
Klimczak RR, Koerber JT, Dalkara D, Flannery JG, and Schaffer DV
- Subjects
- Animals, Astrocytes metabolism, Cell Line, DNA Mutational Analysis, Gene Transfer Techniques, Genetic Therapy, Genetic Vectors, Humans, Neuroglia cytology, Neuroglia metabolism, Rats, Rats, Sprague-Dawley, Retina metabolism, Retinal Neurons metabolism, Dependovirus genetics, Retinal Diseases metabolism, Retinal Diseases therapy
- Abstract
Background: The pathologies of numerous retinal degenerative diseases can be attributed to a multitude of genetic factors, and individualized treatment options for afflicted patients are limited and cost-inefficient. In light of the shared neurodegenerative phenotype among these disorders, a safe and broad-based neuroprotective approach would be desirable to overcome these obstacles. As a result, gene delivery of secretable-neuroprotective factors to Müller cells, a type of retinal glia that contacts all classes of retinal neurons, represents an ideal approach to mediate protection of the entire retina through a simple and innocuous intraocular, or intravitreal, injection of an efficient vehicle such as an adeno-associated viral vector (AAV). Although several naturally occurring AAV variants have been isolated with a variety of tropisms, or cellular specificities, these vectors inefficiently infect Müller cells via intravitreal injection., Methodology/principal Findings: We have previously applied directed evolution to create several novel AAV variants capable of efficient infection of both rat and human astrocytes through iterative selection of a panel of highly diverse AAV libraries. Here, in vivo and in vitro characterization of these isolated variants identifies a previously unreported AAV variant ShH10, closely related to AAV serotype 6 (AAV6), capable of efficient, selective Müller cell infection through intravitreal injection. Importantly, this new variant shows significantly improved transduction relative to AAV2 (>60%) and AAV6., Conclusions/significance: Our findings demonstrate that AAV is a highly versatile vector capable of powerful shifts in tropism from minor sequence changes. This isolated variant represents a new therapeutic vector to treat retinal degenerative diseases through secretion of neuroprotective factors from Müller cells as well as provides new opportunities to study their biological functions in the retina.
- Published
- 2009
- Full Text
- View/download PDF
45. Directed evolution of adeno-associated virus to an infectious respiratory virus.
- Author
-
Excoffon KJ, Koerber JT, Dickey DD, Murtha M, Keshavjee S, Kaspar BK, Zabner J, and Schaffer DV
- Subjects
- Animals, Capsid chemistry, Cell Line, Cell Polarity, Cystic Fibrosis pathology, Cystic Fibrosis virology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Epithelial Cells pathology, Epithelial Cells virology, Gene Library, Genetic Vectors genetics, Humans, Models, Molecular, Mutation genetics, N-Acetylneuraminic Acid metabolism, Phenotype, Selection, Genetic, Transduction, Genetic, Virus Attachment, Dependovirus genetics, Directed Molecular Evolution, Parvoviridae Infections virology
- Abstract
Respiratory viruses evolve to maintain infectivity levels that permit spread yet prevent host and virus extinction, resulting in surprisingly low infection rates. Respiratory viruses harnessed as gene therapy vectors have illustrated this limitation. We used directed evolution in an organotypic human airway model to generate a highly infectious adeno-associated virus. This virus mediated gene transfer more than 100-fold better than parental strains and corrected the cystic fibrosis epithelial Cl(-) transport defect. Thus, under appropriate selective pressures, viruses can evolve to be more infectious than observed in nature, a finding that holds significant implications for designing vectors for gene therapy and for understanding emerging pathogens.
- Published
- 2009
- Full Text
- View/download PDF
46. DNA shuffling of adeno-associated virus yields functionally diverse viral progeny.
- Author
-
Koerber JT, Jang JH, and Schaffer DV
- Subjects
- Animals, Base Sequence, Cell Line, DNA Primers, Dependovirus physiology, Directed Molecular Evolution, Female, Humans, Mice, Mice, Inbred BALB C, Neutralization Tests, Polymerase Chain Reaction, Transduction, Genetic, Virus Assembly, DNA Shuffling, DNA, Viral genetics, Dependovirus genetics
- Abstract
Adeno-associated virus (AAV) vectors are extremely effective gene-delivery vehicles for a broad range of applications. However, the therapeutic efficacy of these and other vectors is currently limited by barriers to safe, efficient gene delivery, including pre-existing antiviral immunity, and infection of off-target cells. Recently, we have implemented directed evolution of AAV, involving the generation of randomly mutagenized viral libraries based on serotype 2 and high-throughput selection, to engineer enhanced viral vectors. Here, we significantly extend this capability by performing high-efficiency in vitro recombination to create a large (10(7)), diverse library of random chimeras of numerous parent AAV serotypes (AAV1, 2, 4-6, 8, and 9). In order to analyze the extent to which such highly chimeric viruses can be viable, we selected the library for efficient viral packaging and infection, and successfully recovered numerous novel chimeras. These new viruses exhibited a broad range of cell tropism both in vitro and in vivo and enhanced resistance to human intravenous immunoglobulin (IVIG), highlighting numerous functional differences between these chimeras and their parent serotypes. Thus, directed evolution can potentially yield unlimited numbers of new AAV variants with novel gene-delivery properties, and subsequent analysis of these variants can further extend basic knowledge of AAV biology.
- Published
- 2008
- Full Text
- View/download PDF
47. Transposon-based mutagenesis generates diverse adeno-associated viral libraries with novel gene delivery properties.
- Author
-
Koerber JT and Schaffer DV
- Subjects
- Genetic Therapy methods, Plasmids, Capsid Proteins genetics, Dependovirus genetics, Gene Library, Gene Transfer Techniques, Genetic Vectors genetics, Mutagenesis, Insertional genetics, Retroelements genetics
- Abstract
The engineering of novel properties and functions into viral vectors for improved gene delivery remains a barrier to the development of efficient, customized gene delivery vehicles. Rational methods for designing improved viral vectors are often experimentally challenging and laborious, particularly when knowledge of viral structure-function relationships is limited. As an alternative, high-throughput libraries may be rapidly and efficiently selected for viral variants with a desired function. Here we describe a transposon-based insertional mutagenesis approach to generate large diverse adeno- associated viral (AAV) libraries containing a randomly located peptide. Briefly, a selectable marker is randomly inserted throughout the AAV2 cap gene and the resulting "bookmarked' AAV cap gene is cloned into an AAV packaging vector. The selectable marker is then replaced with a defined oligonucleotide, and the final AAV library is used to package a diverse pool of AAV virions, which can used for functional selection.
- Published
- 2008
- Full Text
- View/download PDF
48. Molecular engineering of viral gene delivery vehicles.
- Author
-
Schaffer DV, Koerber JT, and Lim KI
- Subjects
- Gene Targeting trends, Genetic Engineering trends, Genetic Therapy trends, Genetic Vectors genetics, Transfection trends, Viruses genetics
- Abstract
Viruses can be engineered to efficiently deliver exogenous genes, but their natural gene delivery properties often fail to meet human therapeutic needs. Therefore, engineering viral vectors with new properties, including enhanced targeting abilities and resistance to immune responses, is a growing area of research. This review discusses protein engineering approaches to generate viral vectors with novel gene delivery capabilities. Rational design of viral vectors has yielded successful advances in vitro, and to an extent in vivo. However, there is often insufficient knowledge of viral structure-function relationships to reengineer existing functions or create new capabilities, such as virus-cell interactions, whose molecular basis is distributed throughout the primary sequence of the viral proteins. Therefore, high-throughput library and directed evolution methods offer alternative approaches to engineer viral vectors with desired properties. Parallel and integrated efforts in rational and library-based design promise to aid the translation of engineered viral vectors toward the clinic.
- Published
- 2008
- Full Text
- View/download PDF
49. Engineering adeno-associated virus for one-step purification via immobilized metal affinity chromatography.
- Author
-
Koerber JT, Jang JH, Yu JH, Kane RS, and Schaffer DV
- Subjects
- Amino Acid Sequence, Animals, Capsid chemistry, Capsid Proteins chemistry, Capsid Proteins genetics, Cells, Cultured, Dependovirus chemistry, Female, Genetic Vectors chemistry, Humans, Molecular Sequence Data, Mutation, Nickel chemistry, Protein Conformation, Rats, Rats, Inbred F344, Chromatography, Affinity methods, Dependovirus genetics, Genetic Vectors genetics, Genetic Vectors isolation & purification, Protein Engineering
- Abstract
Adeno-associated virus (AAV) is a promising vehicle for gene therapy, which will rely on the generation of high-titer, high-purity recombinant vectors. However, numerous purification protocols can involve challenging optimization or scalability issues, and most AAV serotypes do not bind heparin or sialic acid, used for AAV2/3 or AAV4/5 purification, requiring the development of new chromatography strategies. Immobilized metal affinity chromatography (IMAC) allows for robust protein purification via affinity tags such as the hexahistidine (His(6)) sequence. Through the combination of a diverse AAV2 library and rational peptide insertions, we have located an optimal His(6) tag insertion site within the viral capsid. This mutant and a related AAV8 variant can be purified from clarified cell lysate in a single gravity column step at infectious particle yields exceeding 90%. Furthermore, injection of IMAC-purified vector into the brain demonstrates that it mediates high-efficiency gene delivery in vivo, equivalent to that of wild-type capsid, with minimal immune cell activation. This affinity chromatography method may offer advantages in ease of purification, final vector purity, and process scalability. Moreover, a combined rational design and high-throughput library selection approach can aid in the design of enhanced viral gene delivery vectors.
- Published
- 2007
- Full Text
- View/download PDF
50. Directed evolution of adeno-associated virus yields enhanced gene delivery vectors.
- Author
-
Maheshri N, Koerber JT, Kaspar BK, and Schaffer DV
- Subjects
- Adenoviridae genetics, Directed Molecular Evolution methods, Drug Delivery Systems methods, Gene Targeting methods, Genetic Enhancement methods, Genetic Therapy methods, Genetic Vectors genetics, Transfection methods
- Abstract
Adeno-associated viral vectors are highly safe and efficient gene delivery vehicles. However, numerous challenges in vector design remain, including neutralizing antibody responses, tissue transport and infection of resistant cell types. Changes must be made to the viral capsid to overcome these problems; however, very often insufficient information is available for rational design of improvements. We therefore applied a directed evolution approach involving the generation of large mutant capsid libraries and selection of adeno-associated virus (AAV) 2 variants with enhanced properties. High-throughput selection processes were designed to isolate mutants within the library with altered affinities for heparin or the ability to evade antibody neutralization and deliver genes more efficiently than wild-type capsid in the presence of anti-AAV serum. This approach, which can be extended to additional gene delivery challenges and serotypes, directs viral evolution to generate 'designer' gene delivery vectors with specified, enhanced properties.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.