Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
J Pharm Sci ; 112(11): 2910-2920, 2023 Nov.
Article En | MEDLINE | ID: mdl-37429356

MTBT1466A is a high-affinity TGFß3-specific humanized IgG1 monoclonal antibody with reduced Fc effector function, currently under investigation in clinical trials as a potential anti-fibrotic therapy. Here, we characterized the pharmacokinetics (PK) and pharmacodynamics (PD) of MTBT1466A in mice and monkeys and predicted the PK/PD of MTBT1466A in humans to guide the selection of the first-in-human (FIH) starting dose. MTBT1466A demonstrated a typical IgG1-like biphasic PK profile in monkeys, and the predicted human clearance of 2.69 mL/day/kg and t1/2 of 20.4 days are consistent with those expected for a human IgG1 antibody. In a mouse model of bleomycin-induced lung fibrosis, changes in expression of TGFß3-related genes, serpine1, fibronectin-1, and collagen 1A1 were used as PD biomarkers to determine the minimum pharmacologically active dose of 1 mg/kg. Unlike in the fibrosis mouse model, evidence of target engagement in healthy monkeys was only observed at higher doses. Using a PKPD-guided approach, the recommended FIH dose of 50 mg, IV, provided exposures that were shown to be safe and well tolerated in healthy volunteers. MTBT1466A PK in healthy volunteers was predicted reasonably well using a PK model with allometric scaling of PK parameters from monkey data. Taken together, this work provides insights into the PK/PD behavior of MTBT1466A in preclinical species, and supports the translatability of the preclinical data into the clinic.

2.
Bioanalysis ; 14(1): 7-18, 2022 Jan.
Article En | MEDLINE | ID: mdl-34789009

Aim: Development of recombinant fusion proteins as drugs poses unique challenges for bioanalysis. This paper describes a case study of a glycosylated fusion protein, where variable glycosylation, matrix interference and high sensitivity needs posed unique challenges. Results: Six different assay configurations, across four different platforms were evaluated for measurement of drug concentrations. Two platforms that achieved the assay requirements were Simoa HD-1 and immune-capture LC-MS/MS-based assay. Conclusion: Both, Simoa HD-1 and the mass spectrometry-based methods were able to detect total drug by providing the adequate matrix tolerance, required sensitivity and detection of all the various glycosylated fusion proteins to support clinical sample analysis. The mass spectrometry-based method was selected due to robustness and ease of method transfer.


Enzyme-Linked Immunosorbent Assay/methods , N-Acetylneuraminic Acid/pharmacokinetics , Glycosylation , Humans
3.
Sci Transl Med ; 13(605)2021 08 04.
Article En | MEDLINE | ID: mdl-34349032

Transforming growth factor-ß (TGFß) is a key driver of fibrogenesis. Three TGFß isoforms (TGFß1, TGFß2, and TGFß3) in mammals have distinct functions in embryonic development; however, the postnatal pathological roles and activation mechanisms of TGFß2 and TGFß3 have not been well characterized. Here, we show that the latent forms of TGFß2 and TGFß3 can be activated by integrin-independent mechanisms and have lower activation thresholds compared to TGFß1. Unlike TGFB1, TGFB2 and TGFB3 expression is increased in human lung and liver fibrotic tissues compared to healthy control tissues. Thus, TGFß2 and TGFß3 may play a pathological role in fibrosis. Inducible conditional knockout mice and anti-TGFß isoform-selective antibodies demonstrated that TGFß2 and TGFß3 are independently involved in mouse fibrosis models in vivo, and selective TGFß2 and TGFß3 inhibition does not lead to the increased inflammation observed with pan-TGFß isoform inhibition. A cocrystal structure of a TGFß2-anti-TGFß2/3 antibody complex reveals an allosteric isoform-selective inhibitory mechanism. Therefore, inhibiting TGFß2 and/or TGFß3 while sparing TGFß1 may alleviate fibrosis without toxicity concerns associated with pan-TGFß blockade.


Transforming Growth Factor beta2 , Transforming Growth Factor beta3 , Animals , Disease Models, Animal , Female , Fibrosis , Humans , Mice , Protein Isoforms/metabolism , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta3/metabolism
4.
Bioanalysis ; 13(9): 679-691, 2021 May.
Article En | MEDLINE | ID: mdl-33900106

Background: Low volume sampling technologies have come a long way; however, their uptake has been slow due to logistical and perceived implementation challenges. Additional studies are needed to overcome these barriers. Materials & methods/results: Here we present two studies where different sampling technologies were evaluated to determine the feasibility of their implementation. First, we evaluated pharmacokinetic profiles for anti-gD in rats using three tail bleed sampling methods, glass capillary tubes, Shimadzu MSW2® and the Neoteryx Mitra® microsampler. Second, we evaluated two low volume-sampling methods to measure drug levels from patients treated with anti-A therapeutic. This evaluation used whole blood finger pricks for Neoteryx Mitra and plasma from capillary blood using TASSO OnDemand technology to compare results to established venipuncture collection method. Conclusion: These studies evaluate the feasibility and considerations for implementation of different low volume sampling technologies.


Antibodies, Monoclonal/blood , Blood Specimen Collection , Animals , Enzyme-Linked Immunosorbent Assay , Humans , Phlebotomy , Rats
6.
Cell ; 179(2): 417-431.e19, 2019 Oct 03.
Article En | MEDLINE | ID: mdl-31585081

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 ß-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 ß-tryptase, which dissociates active tetramers into inactive monomers. A 2.15 Å crystal structure of a ß-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.


Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/therapy , Mast Cells/enzymology , Mast Cells/immunology , Tryptases/antagonists & inhibitors , Tryptases/immunology , Adolescent , Allosteric Regulation/immunology , Animals , Cell Line , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Rabbits
7.
Pharmacol Res Perspect ; 6(6): e00434, 2018 12.
Article En | MEDLINE | ID: mdl-30464842

Although Interleukin-22 (IL-22) is produced by various leukocytes, it preferentially targets cells with epithelial origins. IL-22 exerts essential roles in modulating various tissue epithelial functions, such as innate host defense against extracellular pathogens, barrier integrity, regeneration, and wound healing. Therefore, IL-22 is thought to have therapeutic potential in treating diseases associated with infection, tissue injury or chronic tissue damage. A number of in vitro and in vivo nonclinical studies were conducted to characterize the pharmacological activity and safety parameters of UTTR1147A, an IL-22 recombinant fusion protein that links the human cytokine IL-22 with the Fc portion of a human immunoglobulin. To assess the pharmacological activity of UTTR1147A, STAT3 activation was evaluated in primary hepatocytes isolated from human, cynomolgus monkey, minipig, rat, and mouse after incubation with UTTR1147A. UTTR1147A activated STAT3 in all species evaluated, demonstrating that all were appropriate nonclinical species for toxicology studies. The nonclinical safety profile of UTTR1147A was evaluated in rats, minipigs, and cynomolgus monkeys to establish a safe clinical starting dose for humans in Phase I trials and to support clinical intravenous, subcutaneous and/or topical administration treatment regimen. Results demonstrate the cross-species translatability of the biological response in activating the IL-22 pathway as well as the translatability of findings from in vitro to in vivo systems. UTTR1147A was well tolerated in all species tested and induced the expected pharmacologic effects of epidermal hyperplasia and a transient increase in on-target acute phase proteins. These effects were all considered to be clinically predictable, manageable, monitorable, and reversible.


Hepatocytes/drug effects , Immunologic Factors/pharmacology , Interleukins/toxicity , Recombinant Fusion Proteins/toxicity , Animals , Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical , Female , Hepatocytes/metabolism , Humans , Interleukins/administration & dosage , Macaca fascicularis , Male , Mice , Primary Cell Culture , Rats , Recombinant Fusion Proteins/administration & dosage , STAT3 Transcription Factor/metabolism , Swine , Swine, Miniature , Interleukin-22
8.
Biochem Pharmacol ; 152: 224-235, 2018 06.
Article En | MEDLINE | ID: mdl-29608910

Interleukin (IL)-22 plays protective roles in infections and in inflammatory diseases that have been linked to its meditation of innate immunity via multiple mechanisms. IL-22 binds specifically to its heterodimeric receptor, which is expressed on a variety of epithelial tissues. UTTR1147A is a recombinant fusion protein that links the human cytokine IL-22 with the Fc portion of human immunoglobulin (Ig) G4. Here, we report extensive in vitro and in vivo nonclinical studies that were conducted to characterize the pharmacological activity of UTTR1147A. The in vitro activity and potency of UTTR1147A were analyzed using primary human hepatocytes and human colonic epithelial cell lines. Assessment of in vivo efficacy was performed in a mouse colitis model and by measuring relevant pharmacodynamic biomarkers, including antimicrobial peptides REG3A/ß, serum amyloid protein A (SAA) and lipopolysaccharide binding protein (LBP). The pharmacokinetic and pharmacodynamic characteristics of UTTR1147A were assessed in healthy mice, rats and cynomolgus monkeys. UTTR1147A induced STAT3 activation through binding to IL-22 receptor expressed in primary human hepatocytes and human colon cell lines. In both, activation occurred in a concentration-dependent manner with similar potencies. In the mouse colitis model, murine IL-22Fc- (muIL-22Fc) treated groups at doses of 1.25 µg and above had statistically lower average histologic colitis scores compared to the control treated group. Administration of muIL-22Fc or UTTR1147A was associated with a dose-dependent induction of PD markers REG3ß and SAA in rodents as well as REG3A, SAA and LBP in cynomolgus monkeys. The combined data confirm pharmacological activity of IL-22Fc and support potential regenerative and protective mechanisms in epithelial tissues.


Immunoglobulin G/metabolism , Interleukins/metabolism , Animals , Area Under Curve , Cell Line , Colitis/chemically induced , Colitis/therapy , Cytokines , Female , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Recombinant Proteins , Interleukin-22
9.
MAbs ; 8(5): 991-7, 2016 07.
Article En | MEDLINE | ID: mdl-27031797

MHAA4549A is a human immunoglobulin G1 (IgG1) monoclonal antibody that binds to a highly conserved epitope on the stalk of influenza A hemagglutinin and blocks the hemagglutinin-mediated membrane fusion in the endosome, neutralizing all known human influenza A strains. Pharmacokinetics (PK) of MHAA4549A and its related antibodies were determined in DBA/2J and Balb-c mice at 5 mg/kg and in cynomolgus monkeys at 5 and 100 mg/kg as a single intravenous dose. Serum samples were analyzed for antibody concentrations using an ELISA and the PK was evaluated using WinNonlin software. Human PK profiles were projected based on the PK in monkeys using species-invariant time method. The human efficacious dose projection was based on in vivo nonclinical pharmacological active doses, exposure in mouse infection models and expected human PK. The PK profiles of MHAA4549A and its related antibody showed a linear bi-exponential disposition in mice and cynomolgus monkeys. In mice, clearance and half-life ranged from 5.77 to 9.98 mL/day/kg and 10.2 to 5.76 days, respectively. In cynomolgus monkeys, clearance and half-life ranged from 4.33 to 4.34 mL/day/kg and 11.3 to 11.9 days, respectively. The predicted clearance in humans was ∼2.60 mL/day/kg. A single intravenous dose ranging from 15 to 45 mg/kg was predicted to achieve efficacious exposure in humans. In conclusion, the PK of MHAA4549A was as expected for a human IgG1 monoclonal antibody that lacks known endogenous host targets. The predicted clearance and projected efficacious doses in humans for MHAA4549A have been verified in a Phase 1 study and Phase 2a study, respectively.


Antibodies, Monoclonal/pharmacokinetics , Antibodies, Viral/pharmacology , Influenza, Human/drug therapy , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Models, Theoretical
10.
J Immunol Methods ; 424: 91-9, 2015 Sep.
Article En | MEDLINE | ID: mdl-26009247

During drug development, measurement of suitable pharmacodynamic biomarkers is key to establishing in vivo drug activity. Binding of monoclonal antibody (mAb) therapeutics to soluble target proteins often results in elevated serum levels of their target antigen, and measuring total (free and bound) concentration of the target antigen can be an important means of demonstrating that the mAb has reached its specific target. However, accurately measuring soluble circulating antigen in preclinical or clinical samples in the presence of a therapeutic mAb presents a bioanalytical challenge. Particularly in the case of low molecular weight and/or multimeric targets, epitopes for capture and detection of the target by reagent antibodies can be obscured by bound therapeutic mAb. Lymphotoxin-alpha (LTα) is a cytokine in the TNF superfamily that has been implicated in the pathophysiology of autoimmune disease, and is a therapeutic target for neutralizing mAb. During preclinical safety studies in cynomolgus macaques, we encountered difficulties in measuring total LTα in serum of dosed animals. When serum LTα trimer was saturated with the anti-LTα mAb, binding of two reagent antibodies, as required for a classic sandwich ELISA, was not feasible, and dissociation methods were also found to be unsuitable. We therefore developed an approach in which excess anti-LTα mAb was added to the in vitro assay system to fully saturate all binding sites, and an anti-idiotypic antibody was used to detect bound therapeutic antibody. Using this method, total LTα could be accurately measured in cynomolgus macaque serum, and was observed to increase with increasing anti-LTα therapeutic mAb dose. Additional in vitro studies demonstrated that the method worked equally well in human serum. This assay strategy will be useful for quantifying total concentrations of other small and/or multimeric target proteins in the presence of a therapeutic antibody.


Antibodies, Monoclonal/pharmacokinetics , Immunoassay/methods , Lymphotoxin-alpha/blood , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Humans , Ligands , Lymphotoxin-alpha/immunology , Macaca fascicularis , Reproducibility of Results , Sensitivity and Specificity
11.
PLoS One ; 8(12): e83958, 2013.
Article En | MEDLINE | ID: mdl-24409221

BACKGROUND: Macrophage stimulating protein (MSP) is a serum growth factor that binds to and activates the receptor tyrosine kinase, Recepteur d'Origine Nantais (RON). A non-synonymous coding variant in MSP (689C) has been associated with genetic susceptibility to both Crohn's disease and ulcerative colitis, two major types of inflammatory bowel disease (IBD) characterized by chronic inflammation of the digestive tract. We investigated the consequences of this polymorphism for MSP-RON pathway activity and IBD pathogenesis. METHODS: RON expression patterns were examined on mouse and human cells and tissues under normal and disease conditions to identify cell types regulated by MSP-RON. Recombinant MSP variants were tested for their ability to bind and stimulate RON and undergo proteolytic activation. MSP concentrations were quantified in the serum of individuals carrying the MSP 689R and 689C alleles. RESULTS: In intestinal tissue, RON was primarily expressed by epithelial cells under normal and disease conditions. The 689C polymorphism had no impact on the ability of MSP to bind to or signal through RON. In a cohort of normal individuals and IBD patients, carriers of the 689C polymorphism had lower concentrations of MSP in their serum. CONCLUSIONS: By reducing the quantities of circulating MSP, the 689C polymorphism, or a variant in linkage disequilibrium with this polymorphism, may impact RON ligand availability and thus receptor activity. Given the known functions of RON in regulating wound healing and our analysis of RON expression patterns in human intestinal tissue, these data suggest that decreased RON activity may impact the efficiency of epithelial repair and thus underlie the increased IBD susceptibility associated with the MSP 689C allele.


Alleles , Genetic Predisposition to Disease , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Polymorphism, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Hepatocyte Growth Factor/blood , Humans , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Mice , Models, Molecular , Protein Conformation , Proteolysis , Proto-Oncogene Proteins/blood , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
12.
MAbs ; 4(4): 475-87, 2012.
Article En | MEDLINE | ID: mdl-22699308

The effector functions of therapeutic antibodies are strongly affected by the specific glycans added to the Fc domain during post-translational processing. Antibodies bearing high levels of N-linked mannose-5 glycan (Man5) have been reported to exhibit enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) compared with antibodies with fucosylated complex or hybrid glycans. To better understand the relationship between antibodies with high levels of Man5 and their biological activity in vivo, we developed an approach to generate substantially homogeneous antibodies bearing the Man5 glycoform. A mannosidase inhibitor, kifunensine, was first incorporated in the cell culture process to generate antibodies with a distribution of high mannose glycoforms. Antibodies were then purified and treated with a mannosidase for trimming to Man5 in vitro. This 2-step approach can consistently generate antibodies with > 99% Man5 glycan. Antibodies bearing varying levels of Man5 were studied to compare ADCC and Fcγ receptor binding, and they showed enhanced ADCC activity and increased binding affinity to the FcγRIIIA. In addition, the clearance rate of antibodies bearing Man8/9 and Man5 glycans was determined in a pharmacokinetics study in mice. When compared with historical data, the antibodies bearing the high mannose glycoform exhibited faster clearance rate compared with antibodies bearing the fucosylated complex glycoform, while the pharmacokinetic properties of antibodies with Man8/9 and Man5 glycoforms appeared similar. In addition, we identified the presence of a mannosidase in mouse serum that converted most Man8/9 to Man6 after 24 h.


Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Mannose/immunology , Polysaccharides/immunology , Alkaloids/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Area Under Curve , Binding, Competitive , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Mannose/metabolism , Mannosidases/metabolism , Metabolic Clearance Rate , Mice , Mice, Nude , Polysaccharides/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Spectrometry, Mass, Electrospray Ionization
13.
MAbs ; 4(1): 101-9, 2012.
Article En | MEDLINE | ID: mdl-22327433

The neonatal Fc receptor (FcRn) plays an important and well-known role in immunoglobulin G (IgG) catabolism; however, its role in the disposition of IgG after subcutaneous (SC) administration, including bioavailability, is relatively unknown. To examine the potential effect of FcRn on IgG SC bioavailability, we engineered three anti-amyloid ß monoclonal antibody (mAb) reverse chimeric mouse IgG2a (mIgG2a) Fc variants (I253A.H435A, N434H and N434Y) with different binding affinities to mouse FcRn (mFcRn) and compared their SC bioavailability to that of the wild-type (WT) mAb in mice. Our results indicated that the SC bioavailability of mIgG2a was affected by mFcRn-binding affinity. Variant I253A.H435A, which did not bind to mFcRn at either pH 6.0 or pH 7.4, had the lowest bioavailability (41.8%). Variant N434Y, which had the greatest increase in binding affinity at both pH 6.0 and pH 7.4, had comparable bioavailability to the WT antibody (86.1% vs. 76.3%), whereas Variant N434H, which had modestly increased binding affinity at pH 6.0 to mFcRn and affinity comparable to the WT antibody at pH 7.4, had the highest bioavailability (94.7%). A semi-mechanism-based pharmacokinetic model, which described well the observed data with the WT antibody and variant I253A.H435A, is consistent with the hypothesis that the decreased bioavailability of variant I253A.H435A was due to loss of the FcRn-mediated protection from catabolism at the absorption site. Together, these data demonstrate that FcRn plays an important role in SC bioavailability of therapeutic IgG antibodies.


Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibody Affinity/immunology , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Biological Availability , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Injections, Subcutaneous , Mice , Mice, SCID , Protein Binding
14.
Science ; 335(6064): 89-92, 2012 Jan 06.
Article En | MEDLINE | ID: mdl-22174130

Lysosomal storage diseases (LSDs) are a group of heterogeneous disorders caused by defects in lysosomal enzymes or transporters, resulting in accumulation of undegraded macromolecules or metabolites. Macrophage numbers are expanded in several LSDs, leading to histiocytosis of unknown pathophysiology. Here, we found that mice lacking the equilibrative nucleoside transporter 3 (ENT3) developed a spontaneous and progressive macrophage-dominated histiocytosis. In the absence of ENT3, defective apoptotic cell clearance led to lysosomal nucleoside buildup, elevated intralysosomal pH, and altered macrophage function. The macrophage accumulation was partly due to increased macrophage colony-stimulating factor and receptor expression and signaling secondary to the lysosomal defects. These studies suggest a cellular and molecular basis for the development of histiocytosis in several human syndromes associated with ENT3 mutations and potentially other LSDs.


Histiocytosis/physiopathology , Homeostasis , Lysosomes/physiology , Macrophages/physiology , Nucleoside Transport Proteins/physiology , Adenosine/metabolism , Animals , Apoptosis , Cell Count , Cell Proliferation , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Listeriosis/immunology , Listeriosis/microbiology , Lysosomal Storage Diseases/physiopathology , Lysosomes/ultrastructure , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis , Nucleoside Transport Proteins/genetics , Phagocytosis , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Thymocytes/immunology , Thymocytes/physiology
15.
MAbs ; 4(6): 753-60, 2012.
Article En | MEDLINE | ID: mdl-23778268

A majority of human therapeutic antibody candidates show pharmacokinetic properties suitable for clinical use, but an unexpectedly fast antibody clearance is sometimes observed that may limit the clinical utility. Pharmacokinetic data in cynomolgus monkeys collected for a panel of 52 antibodies showed broad distribution of target-independent clearance values (2.4-61.3 mL/day/kg), with 15 (29%) having clearance > 10 mL/day/kg. Alteration in the interaction with the recycling FcRn receptor did not account for the faster than expected clearance observed for the antibodies; off-target binding was presumed to account for the fast clearance. We developed an assay based on ELISA detection of non-specific binding to baculovirus particles that can identify antibodies having increased risk for fast clearance. This assay can be used during lead generation or optimization to identify antibodies with increased risk of having fast clearance in both humans and cynomolgus monkeys, and thus increase the likelihood of obtaining a suitable drug candidate.


Antibodies, Monoclonal/metabolism , Metabolic Clearance Rate , Animals , Baculoviridae/immunology , Drug Discovery , Enzyme-Linked Immunosorbent Assay , Humans , Macaca fascicularis , Protein Binding , Risk Adjustment , Virion/metabolism
...