Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Pathogens ; 12(12)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38133292

ABSTRACT

As part of the non-clinical safety package characterizing bamlanivimab (SARS-CoV-2 neutralizing monoclonal antibody), the risk profile for antibody-dependent enhancement of infection (ADE) was evaluated in vitro and in an African green monkey (AGM) model of COVID-19. In vitro ADE assays in primary human macrophage, Raji, or THP-1 cells were used to evaluate enhancement of viral infection. Bamlanivimab binding to C1q, FcR, and cell-based effector activity was also assessed. In AGMs, the impact of bamlanivimab pretreatment on viral loads and clinical and histological pathology was assessed to evaluate enhanced SARS-CoV-2 replication or pathology. Bamlanivimab did not increase viral replication in vitro, despite a demonstrated effector function. In vivo, no significant differences were found among the AGM groups for weight, temperature, or food intake. Treatment with bamlanivimab reduced viral loads in nasal and oral swabs and BAL fluid relative to control groups. Viral antigen was not detected in lung tissue from animals treated with the highest dose of bamlanivimab. Bamlanivimab did not induce ADE of SARS-CoV-2 infection in vitro or in an AGM model of infection at any dose evaluated. The findings suggest that high-affinity monoclonal antibodies pose a low risk of mediating ADE in patients and support their safety profile as a treatment of COVID-19 disease.

2.
JCI Insight ; 8(13)2023 07 10.
Article in English | MEDLINE | ID: mdl-37427592

ABSTRACT

B cells contribute to multiple aspects of autoimmune disorders, and B cell-targeting therapies, including B cell depletion, have been proven to be efficacious in treatment of multiple autoimmune diseases. However, the development of novel therapies targeting B cells with higher efficacy and a nondepleting mechanism of action is highly desirable. Here we describe a nondepleting, high-affinity anti-human CD19 antibody LY3541860 that exhibits potent B cell inhibitory activities. LY3541860 inhibits B cell activation, proliferation, and differentiation of primary human B cells with high potency. LY3541860 also inhibits human B cell activities in vivo in humanized mice. Similarly, our potent anti-mCD19 antibody also demonstrates improved efficacy over CD20 B cell depletion therapy in multiple B cell-dependent autoimmune disease models. Our data indicate that anti-CD19 antibody is a highly potent B cell inhibitor that may have potential to demonstrate improved efficacy over currently available B cell-targeting therapies in treatment of autoimmune conditions without causing B cell depletion.


Subject(s)
Autoimmune Diseases , B-Lymphocytes , Mice , Animals , Antigens, CD19 , Autoimmune Diseases/drug therapy
3.
MAbs ; 12(1): 1831880, 2020.
Article in English | MEDLINE | ID: mdl-33183151

ABSTRACT

CXCR1 and CXCR2 signaling play a critical role in neutrophil migration, angiogenesis, and tumorigenesis and are therefore an attractive signaling axis to target in a variety of indications. In human, a total of seven chemokines signal through these receptors and comprise the ELR+CXC chemokine family, so named because of the conserved ELRCXC N-terminal motif. To fully antagonize CXCR1 and CXCR2 signaling, an effective therapeutic should block either both receptors or all seven ligands, yet neither approach has been fully realized clinically. In this work, we describe the generation and characterization of LY3041658, a humanized monoclonal antibody that binds and neutralizes all seven human and cynomolgus monkey ELR+CXC chemokines and three of five mouse and rat ELR+CXC chemokines with high affinity. LY3041658 is able to block ELR+CXC chemokine-induced Ca2+ mobilization, CXCR2 internalization, and chemotaxis in vitro as well as neutrophil mobilization in vivo without affecting other neutrophil functions. In addition to the in vitro and in vivo activity, we characterized the epitope and structural basis for binding in detail through alanine scanning, crystallography, and mutagenesis. Together, these data provide a robust preclinical characterization of LY3041658 for which the efficacy and safety is being evaluated in human clinical trials for neutrophilic skin diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Antibody Affinity , Chemotaxis, Leukocyte/immunology , Humans , Macaca fascicularis , Mice , Neutrophils/immunology , Rats
4.
MAbs ; 12(1): 1770028, 2020 01 01.
Article in English | MEDLINE | ID: mdl-32486889

ABSTRACT

Many therapeutic monoclonal antibodies (mAbs) were initially developed for intravenous (IV) administration. As a means to improve mAb drug-ability and the patient experience, subcutaneous (SC) administration is an increasingly important delivery route for mAbs. Unlike IV administration, bioavailability limitations for antibodies have been reported following SC injection and can dictate whether a mAb is administered via this parenteral route. The SC bioavailability of antibodies has been difficult to predict, and it can be variable and partial, with values ranging from ~50% to 100%. The mechanisms leading to the incomplete bioavailability of some mAbs relative to others are not well understood. There are some limited data that suggest the physiochemical properties inherent to a mAb can contribute to its SC absorption, bioavailability, and in vivo fate. In this study, we evaluated the integrated influence of multiple mAb physiochemical factors on the SC absorption and bioavailability of six humanized mAbs in both rats and cynomolgus monkeys. We demonstrate the physiochemical properties of mAbs are critical to their rate and extent of SC absorption. The combination of high positive charge and hydrophobic interaction significantly reduced the rate of the evaluated mAb's SC absorption and bioavailability. Reduction or balancing of both these attributes via re-engineering the mAbs restored desirable properties of the molecules assessed. This included reduced association with SC tissue, improvements in mAb absorption from the SC space and overall SC bioavailability. Our findings point to the importance of evaluating the relative balance between various physiochemical factors, including charge, hydrophobicity, and stability, to improve the SC drug-ability of mAbs for selecting or engineering mAbs with enhanced in vivo absorption and bioavailability following SC administration.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Chemistry, Physical/methods , Animals , Antibodies, Monoclonal, Humanized/chemistry , Bioengineering , Biological Availability , Drug Development , Humans , Hydrophobic and Hydrophilic Interactions , Injections, Subcutaneous , Macaca fascicularis , Protein Binding , Protein Stability , Rats , Subcutaneous Absorption
5.
Biopharm Drug Dispos ; 41(3): 111-125, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32080869

ABSTRACT

The immunogenicity of biotherapeutics presents a major challenge during the clinical development of new protein drugs including monoclonal antibodies. To address this, multiple humanization and de-immunization techniques that employ in silico algorithms and in vitro test systems have been proposed and implemented. However, the success of these approaches has been variable and to date, the ability of these techniques to predict immunogenicity has not been systematically tested in humans or other primates. This study tested whether antibody humanization and de-immunization strategies reduce the risk of anti-drug antibody (ADA) development using cynomolgus macaque as a surrogate for human. First human-cyno chimeric antibodies were constructed by grafting the variable domains of the adalimumab and golimumab monoclonal antibodies onto cynomolgus macaque IgG1 and Igκ constant domains followed by framework germlining to cyno to reduce the xenogenic content. Next, B and T cell epitopes and aggregation-prone regions were identified using common in silico methods to select domains with an ADA risk for additional modification. The resultant engineered antibodies had a comparable affinity for TNFα, demonstrated similar biophysical properties, and exhibited significantly reduced ADA levels in cynomolgus macaque compared with the parental antibodies, with a corresponding improvement in the pharmacokinetic profile. Notably, plasma concentrations of the engineered antibodies were quantifiable through 504 hours (chimeric) and 840 hours (germlined/de-immunized), compared with only 336 hours (adalimumab) or 336-672 hours (golimumab). The results point to the significant value in the investment in these engineering strategies as an important guide for monoclonal antibody optimization that can contribute to improved clinical outcomes.


Subject(s)
Adalimumab/immunology , Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Animals , Female , Humans , Immunization , Macaca fascicularis , Male , Tumor Necrosis Factor-alpha/immunology
6.
Br J Clin Pharmacol ; 85(5): 935-948, 2019 05.
Article in English | MEDLINE | ID: mdl-30677788

ABSTRACT

AIMS: Erythropoiesis-stimulating agents used to treat anaemia in patients with chronic kidney disease (CKD) have been associated with cardiovascular adverse events. Hepcidin production, controlled by bone morphogenic protein 6 (BMP6), regulates iron homeostasis via interactions with the iron transporter, ferroportin. High hepcidin levels are thought to contribute to increased iron sequestration and subsequent anaemia in CKD patients. To investigate alternative therapies to erythropoiesis-stimulating agents for CKD patients, monoclonal antibodies, LY3113593 and LY2928057, targeting BMP6 and ferroportin respectively, were tested in CKD patients. METHODS: Preclinical in vitro/vivo data and clinical data in healthy subjects and CKD patients were used to illustrate the translation of pharmacological properties of LY3113593 and LY2928057, highlighting the novelty of targeting these nodes within the hepcidin-ferroportin pathway. RESULTS: LY2928057 bound ferroportin and blocked interactions with hepcidin, allowing iron efflux, leading to increased serum iron and transferrin saturation levels and increased hepcidin in monkeys and humans. In CKD patients, LY2928057 led to slower haemoglobin decline and reduction in ferritin (compared to placebo). Serum iron increase was (mean [90% confidence interval]) 1.98 [1.46-2.68] and 1.36 [1.22-1.51] fold-relative to baseline following LY2928057 600 mg and LY311593 150 mg respectively in CKD patients. LY3113593 specifically blocked BMP6 binding to its receptor and produced increases in iron and transferrin saturation and decreases in hepcidin preclinically and clinically. In CKD patients, LY3113593 produced an increase in haemoglobin and reduction in ferritin (compared to placebo). CONCLUSION: LY3113593 and LY2928057 pharmacological effects (serum iron and ferritin) were translated from preclinical-to-clinical development. Such interventions may lead to new CKD anaemia treatments.


Subject(s)
Anemia/drug therapy , Hematologic Agents/pharmacology , Hepcidins/metabolism , Renal Insufficiency, Chronic/complications , Signal Transduction/drug effects , Adult , Anemia/blood , Anemia/etiology , Anemia/metabolism , Animals , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Ferritins/blood , Ferritins/metabolism , Healthy Volunteers , Hematologic Agents/therapeutic use , Hemoglobins/analysis , Humans , Iron/blood , Iron/metabolism , Macaca fascicularis , Male , Mice , Middle Aged , Rats , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/metabolism , Treatment Outcome , Young Adult
7.
Biotechnol J ; 14(3): e1800007, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29802766

ABSTRACT

There is a rapidly growing reinvigoration of the investigation of small proteins, cyclic peptides, and mAb derived domains as biotherapies. The drugability of these structures are challenged by fast peripheral clearance properties that can reduce their potential to be realized as medicines. Engineering strategies have been of limited value because mechanistically the half-life benefit is manifested by increasing the molecular weight and/or the hydrodyanimc radius which slows the molecule's renal elimination, but can result in the inherent loss of activity and target accessibility. The present work evaluated an alternative approach using smaller peptide sequences which bind to the neonatal Fc receptor (FcRn). Results revealed, small linear and cyclic FcRn binding peptides (FcRnBPs) fused to a combination of the N- and C-termini of a Fab can significantly improve the pharmacokinetics of the protein in cynomolgus monkeys relative to the parental Fab. The linear and cyclic conformations, as well as, the number of FcRnBPs fused to the Fab both influence the clearance and the extent of pharmacokinetic benefit. FcRnBP fusion protein kinetics were also affected by a combination of post-translation modifications and non-specific binding properties. The results in this report lay some foundation in fostering the advent of newer technologies toward successfully improving the pharmacokinetics of proteins, peptides, and mAb-derived domains. Additional work in the integration of a variety of factors including the intended site of action, tissue disposition, metabolism, toxicity and pharmacokinetic, and pharmacodynamics relationship of the intended therapeutic modality are key areas for advancement of these approaches.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin Fab Fragments/metabolism , Peptides/metabolism , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal/metabolism , Caco-2 Cells , Cell Line , Cell Line, Tumor , HEK293 Cells , Half-Life , Humans , Kinetics , Macaca fascicularis , Male , Protein Binding/physiology
8.
MAbs ; 10(6): 913-921, 2018.
Article in English | MEDLINE | ID: mdl-29953319

ABSTRACT

Immunomodulatory monoclonal IgG1 antibodies developed for cancer and autoimmune disease have an inherent risk of systemic release of pro-inflammatory cytokines. In vitro cytokine release assays are currently used to predict cytokine release syndrome (CRS) risk, but the validation of these preclinical tools suffers from the limited number of characterized CRS-inducing IgG1 antibodies and the poor understanding of the mechanisms regulating cytokine release. Here, we incubated human whole blood from naïve healthy volunteers with four monoclonal IgG1 antibodies with different proven or predicted capacity to elicit CRS in clinic and measured cytokine release using a multiplex assay. We found that, in contrast to anti-CD52 antibodies (Campath-1H homolog) that elicited high level of multiple inflammatory cytokines from human blood cells in vitro, other IgG1 antibodies with CRS-inducing potential consistently induced release of a single tested cytokine, interferon (IFN)-γ, with a smaller magnitude than Campath. IFN-γ expression was observed as early as 2-4 h after incubation, mediated by natural killer cells, and dependent upon tumor necrosis factor and FcγRIII. Importantly, the magnitude of the IFN-γ response elicited by IgG1 antibodies with CRS-inducing potential was determined by donor FcγRIIIa-V158F polymorphism. Overall, our results highlight the importance of FcγRIIIa-dependent IFN-γ release in preclinical cytokine release assay for the prediction of CRS risk associated with therapeutic IgG1 antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Interferon-gamma/immunology , Receptors, IgG/immunology , Alemtuzumab/immunology , Alemtuzumab/therapeutic use , Antibodies, Monoclonal/therapeutic use , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Humans , Immunoassay/methods , Immunoglobulin G/therapeutic use , Interferon-gamma/blood , Interferon-gamma/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Polymorphism, Genetic/immunology , Prognosis , Receptors, IgG/genetics , Syndrome
9.
Biotechnol Bioeng ; 115(3): 705-718, 2018 03.
Article in English | MEDLINE | ID: mdl-29150961

ABSTRACT

Cross-linking of the Fcγ receptors expressed on the surface of hematopoietic cells by IgG immune complexes triggers the activation of key immune effector mechanisms, including antibody-dependent cell mediated cytotoxicity (ADCC). A conserved N-glycan positioned at the N-terminal region of the IgG CH 2 domain is critical in maintaining the quaternary structure of the molecule for Fcγ receptor engagement. The removal of a single core fucose residue from the N-glycan results in a considerable increase in affinity for FcγRIIIa leading to an enhanced receptor-mediated immunoeffector function. The enhanced potency of the molecule translates into a number of distinct advantages in the development of IgG antibodies for cancer therapy. In an effort to significantly increase the potency of an anti-CD20, IgG1 molecule, we selectively targeted the de novo GDP-fucose biosynthesis pathway of the host CHO cell line to generate >80% afucosylated IgG1 resulting in enhanced FcγRIIIa binding (13-fold) and in vitro ADCC cell-based activity (11-fold). In addition, this effective glycoengineering strategy also allowed for the utilization of the alternate GDP-fucose salvage pathway to provide a fast and efficient mechanism to manipulate the N-glycan fucosylation level to modulate IgG immune effector function.


Subject(s)
Cricetulus/metabolism , Immunoglobulin G/biosynthesis , Protein Engineering , Rituximab/biosynthesis , Animals , Cricetulus/genetics , Glycosylation , Immunoglobulin G/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Rituximab/genetics
10.
Am J Physiol Renal Physiol ; 312(6): F951-F962, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28249836

ABSTRACT

Transforming growth factor-alpha (TGFA) has been shown to play a role in experimental chronic kidney disease associated with nephron reduction, while its role in diabetic kidney disease (DKD) is unknown. We show here that intrarenal TGFA mRNA expression, as well as urine and serum TGFA, are increased in human DKD. We used a TGFA neutralizing antibody to determine the role of TGFA in two models of renal disease, the remnant surgical reduction model and the uninephrectomized (uniNx) db/db DKD model. In addition, the contribution of TGFA to DKD progression was examined using an adeno-associated virus approach to increase circulating TGFA in experimental DKD. In vivo blockade of TGFA attenuated kidney disease progression in both nondiabetic 129S6 nephron reduction and Type 2 diabetic uniNx db/db models, whereas overexpression of TGFA in uniNx db/db model accelerated renal disease. Therapeutic activity of the TGFA antibody was enhanced with renin angiotensin system inhibition with further improvement in renal parameters. These findings suggest a pathologic contribution of TGFA in DKD and support the possibility that therapeutic administration of neutralizing antibodies could provide a novel treatment for the disease.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney/metabolism , Transforming Growth Factor alpha/metabolism , Aged , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Blood Pressure , Cells, Cultured , Dependovirus/genetics , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/physiopathology , Disease Models, Animal , Disease Progression , ErbB Receptors/metabolism , Extracellular Matrix/metabolism , Female , Gene Transfer Techniques , Genetic Vectors , Glomerular Filtration Rate , Humans , Hypertension/complications , Hypertension/physiopathology , Kidney/drug effects , Kidney/physiopathology , Kidney/surgery , Male , Mice, 129 Strain , Mice, Knockout , Middle Aged , Nephrectomy , Phosphorylation , Renin-Angiotensin System , Signal Transduction , Time Factors , Transforming Growth Factor alpha/antagonists & inhibitors , Transforming Growth Factor alpha/deficiency , Transforming Growth Factor alpha/genetics
11.
Protein Sci ; 25(11): 2028-2036, 2016 11.
Article in English | MEDLINE | ID: mdl-27543934

ABSTRACT

Recent studies have implicated a role of the epidermal growth factor receptor (EGFR) pathway in kidney disease. Skin toxicity associated with therapeutics which completely block the EGFR pathway precludes their use in chronic dosing. Therefore, we developed antibodies which specifically neutralize the EGFR ligands TGFα (transforming growth factor-alpha) and epiregulin but not EGF (epidermal growth factor), amphiregulin, betacellulin, HB-EGF (heparin-binding epidermal growth factor), or epigen. The epitope of one such neutralizing antibody, LY3016859, was characterized in detail to elucidate the structural basis for ligand specificity. Here we report a crystal structure of the LY3016859 Fab fragment in complex with soluble human TGFα. Our data demonstrate a conformational epitope located primarily within the C-terminal subdomain of the ligand. In addition, point mutagenesis experiments were used to highlight specific amino acids which are critical for both antigen binding and neutralization, most notably Ala41 , Glu44 , and His45 . These results illustrate the structural basis for the ligand specificity/selectivity of LY3016859 and could also provide insight into further engineering to alter specificity and/or affinity of LY3016859.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibody Specificity , Epiregulin/chemistry , Epitopes/chemistry , Immunoglobulin Fab Fragments/chemistry , Transforming Growth Factor alpha , Animals , Humans , Mice , Transforming Growth Factor alpha/antagonists & inhibitors , Transforming Growth Factor alpha/chemistry
12.
MAbs ; 7(6): 1084-93, 2015.
Article in English | MEDLINE | ID: mdl-26337808

ABSTRACT

The application of protein engineering technologies toward successfully improving antibody pharmacokinetics has been challenging due to the multiplicity of biochemical factors that influence monoclonal antibody (mAb) disposition in vivo. Physiological factors including interactions with the neonatal Fc receptor (FcRn) and specific antigen binding properties of mAbs, along with biophysical properties of the mAbs themselves play a critical role. It has become evident that applying an integrated approach to understand the relative contribution of these factors is critical to rationally guide and apply engineering strategies to optimize mAb pharmacokinetics. The study presented here evaluated the influence of unintended non-specific interactions on the disposition of mAbs whose clearance rates are governed predominantly by either non-specific (FcRn) or target-mediated processes. The pharmacokinetics of 8 mAbs representing a diverse range of these properties was evaluated in cynomolgus monkeys. Results revealed complementarity-determining region (CDR) charge patch engineering to decrease charge-related non-specific binding can have a significant impact on improving the clearance. In contrast, the influence of enhanced in vitro FcRn binding was mixed, and related to both the strength of charge interaction and the general mechanism predominant in governing the clearance of the particular mAb. Overall, improved pharmacokinetics through enhanced FcRn interactions were apparent for a CDR charge-patch normalized mAb which was affected by non-specific clearance. The findings in this report are an important demonstration that mAb pharmacokinetics requires optimization on a case-by-case basis to improve the design of molecules with increased therapeutic application.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Complementarity Determining Regions/metabolism , Histocompatibility Antigens Class I/metabolism , Protein Engineering/methods , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibody Affinity/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Macaca fascicularis , Metabolic Clearance Rate , Mice , Protein Binding/immunology , Receptors, Fc/immunology
13.
MAbs ; 7(3): 483-93, 2015.
Article in English | MEDLINE | ID: mdl-25695748

ABSTRACT

Lowering the isoelectric point (pI) through engineering the variable region or framework of an IgG can improve its exposure and half-life via a reduction in clearance mediated through non-specific interactions. As such, net charge is a potentially important property to consider in developing therapeutic IgG molecules having favorable pharmaceutical characteristics. Frequently, it may not be possible to shift the pI of monoclonal antibodies (mAbs) dramatically without the introduction of other liabilities such as increased off-target interactions or reduced on-target binding properties. In this report, we explored the influence of more subtle modifications of molecular charge on the in vivo properties of an IgG1 and IgG4 monoclonal antibody. Molecular surface modeling was used to direct residue substitutions in the complementarity-determining regions (CDRs) to disrupt positive charge patch regions, resulting in a reduction in net positive charge without affecting the overall pI of the mAbs. The effect of balancing the net positive charge on non-specific binding was more significant for the IgG4 versus the IgG1 molecule that we examined. This differential effect was connected to the degree of influence on cellular degradation in vitro and in vivo clearance, distribution and metabolism in mice. In the more extreme case of the IgG4, balancing the charge yielded an ∼7-fold improvement in peripheral exposure, as well as significantly reduced tissue catabolism and subsequent excretion of proteolyzed products in urine. Balancing charge on the IgG1 molecule had a more subtle influence on non-specific binding and yielded only a modest alteration in clearance, distribution and elimination. These results suggest that balancing CDR charge without affecting the pI can lead to improved mAb pharmacokinetics, the magnitude of which is likely dependent on the relative influence of charge imbalance and other factors affecting the molecule's disposition.


Subject(s)
Antibodies, Monoclonal, Humanized , Antibody Specificity/genetics , Complementarity Determining Regions , Immunoglobulin G , Models, Molecular , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Complementarity Determining Regions/pharmacology , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Isoelectric Point , Mice
14.
J Inflamm Res ; 7: 121-31, 2014.
Article in English | MEDLINE | ID: mdl-25258549

ABSTRACT

B-cell activating factor (BAFF) is a B-cell survival factor with a key role in B-cell homeostasis and tolerance. Dysregulated BAFF expression may contribute to autoimmune diseases or B-cell malignancies via effects on abnormal B-lymphocyte activation, proliferation, survival, and immunoglobulin secretion. Monoclonal antibodies were generated against human BAFF, characterized for species specificity and affinity, and screened for the ability to neutralize both membrane-bound and soluble BAFF. In addition, studies were undertaken to determine the relative potency of membrane-bound and soluble BAFF. Tabalumab has a high affinity for human, cynomolgus monkey, and rabbit BAFF. No binding to mouse BAFF was detected. Tabalumab was able to neutralize soluble human, cynomolgus monkey, or rabbit BAFF with equal potency. Our data demonstrate that membrane-bound BAFF can be a more potent stimulus for B-cells than soluble BAFF, and tabalumab also neutralized membrane-bound BAFF. Tabalumab prevented BAFF from binding to BAFF receptors and demonstrated pharmacodynamic effects in human BAFF transgenic mice. Tabalumab is a high-affinity human antibody with neutralizing activity against membrane-bound and soluble BAFF. Given our findings that membrane-bound BAFF can have greater in vitro potency than soluble BAFF, neutralization of both forms of BAFF is likely to be important for optimal therapeutic effect.

15.
Haematologica ; 99(9): 1516-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24895335

ABSTRACT

Anemia of chronic disease is a multifactorial disorder, resulting mainly from inflammation-driven reticuloendothelial iron retention, impaired erythropoiesis, and reduced biological activity of erythropoietin. Erythropoiesis-stimulating agents have been used for the treatment of anemia of chronic disease, although with varying response rates and potential adverse effects. Serum concentrations of hepcidin, a key regulator of iron homeostasis, are increased in patients with anemia of chronic disease and linked to the pathogenesis of this disease, because hepcidin blocks cellular iron egress, thus limiting availability of iron for erythropoiesis. We tested whether serum hepcidin levels can predict and affect the therapeutic efficacy of erythropoiesis-stimulating agent treatment using a well-established rat model of anemia of chronic disease. We found that high pre-treatment hepcidin levels correlated with an impaired hematologic response to an erythropoiesis-stimulating agent in rats with anemia of chronic disease. Combined treatment with an erythropoiesis-stimulating agent and an inhibitor of hepcidin expression, LDN-193189, significantly reduced serum hepcidin levels, mobilized iron from tissue stores, increased serum iron levels and improved hemoglobin levels more effectively than did the erythropoiesis-stimulating agent or LDN-193189 monotherapy. In parallel, both the erythropoiesis-stimulating agent and erythropoiesis-stimulating agent/LDN-193189 combined reduced the expression of cytokines known to inhibit erythropoiesis. We conclude that serum hepcidin levels can predict the hematologic responsiveness to erythropoiesis-stimulating agent therapy in anemia of chronic disease. Pharmacological inhibition of hepcidin formation improves the erythropoiesis-stimulating agent's therapeutic efficacy, which may favor a reduction of erythropoiesis-stimulating agent dosages, costs and side effects.


Subject(s)
Anemia/drug therapy , Erythropoietin/pharmacology , Hematinics/pharmacology , Hepcidins/genetics , Iron/blood , RNA, Messenger/genetics , Anemia/blood , Anemia/chemically induced , Anemia/pathology , Animals , Biomarkers/blood , Chronic Disease , Drug Combinations , Drug Synergism , Erythropoiesis/drug effects , Female , Gene Expression , Hepcidins/antagonists & inhibitors , Hepcidins/blood , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Iron/agonists , Polysaccharides, Bacterial , Prognosis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/blood , Rats , Rats, Inbred Lew , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
16.
Gut ; 63(12): 1951-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24598129

ABSTRACT

OBJECTIVE: Hypoxia affects body iron homeostasis; however, the underlying mechanisms are incompletely understood. DESIGN: Using a standardised hypoxia chamber, 23 healthy volunteers were subjected to hypoxic conditions, equivalent to an altitude of 5600 m, for 6 h. Subsequent experiments were performed in C57BL/6 mice, CREB-H knockout mice, primary hepatocytes and HepG2 cells. RESULTS: Exposure of subjects to hypoxia resulted in a significant decrease of serum levels of the master regulator of iron homeostasis hepcidin and elevated concentrations of platelet derived growth factor (PDGF)-BB. Using correlation analysis, we identified PDGF-BB to be associated with hypoxia mediated hepcidin repression in humans. We then exposed mice to hypoxia using a standardised chamber and observed downregulation of hepatic hepcidin mRNA expression that was paralleled by elevated serum PDGF-BB protein concentrations and higher serum iron levels as compared with mice housed under normoxic conditions. PDGF-BB treatment in vitro and in vivo resulted in suppression of both steady state and BMP6 inducible hepcidin expression. Mechanistically, PDGF-BB inhibits hepcidin transcription by downregulating the protein expression of the transcription factors CREB and CREB-H, and pharmacological blockade or genetic ablation of these pathways abrogated the effects of PDGF-BB toward hepcidin expression. CONCLUSIONS: Hypoxia decreases hepatic hepcidin expression by a novel regulatory pathway exerted via PDGF-BB, leading to increased availability of circulating iron that can be used for erythropoiesis.


Subject(s)
Hepcidins/metabolism , Hypoxia/metabolism , Iron/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Adult , Animals , Becaplermin , Disease Models, Animal , Down-Regulation , Erythropoiesis/physiology , Female , Healthy Volunteers , Hematologic Agents/pharmacology , Hep G2 Cells , Humans , Hypoxia/etiology , Male , Mice , Mice, Inbred C57BL
17.
J Pharmacol Exp Ther ; 349(2): 330-43, 2014 May.
Article in English | MEDLINE | ID: mdl-24518034

ABSTRACT

At least seven distinct epidermal growth factor (EGF) ligands bind to and activate the EGF receptor (EGFR). This activation plays an important role in the embryo and in the maintenance of adult tissues. Importantly, pharmacologic EGFR inhibition also plays a critical role in the pathophysiology of diverse disease states, especially cancer. The roles of specific EGFR ligands are poorly defined in these disease states. Accumulating evidence suggests a role for transforming growth factor α (TGFα) in skin, lung, and kidney disease. To explore the role of Tgfa, we generated a monoclonal antibody (mAb41) that binds to and neutralizes human Tgfa with high affinity (KD = 36.5 pM). The antibody also binds human epiregulin (Ereg) (KD = 346.6 pM) and inhibits ligand induced myofibroblast cell proliferation (IC50 values of 0.52 and 1.12 nM for human Tgfa and Ereg, respectively). In vivo, a single administration of the antibody to pregnant mice (30 mg/kg s.c. at day 14 after plug) or weekly administration to neonate mice (20 mg/kg s.c. for 4 weeks) phenocopy Tgfa knockout mice with curly whiskers, stunted growth, and expansion of the hypertrophic zone of growth plate cartilage. Humanization of this monoclonal antibody to a human IgG4 antibody (LY3016859) enables clinical development. Importantly, administration of the humanized antibody to cynomolgus monkeys is absent of the skin toxicity observed with current EGFR inhibitors used clinically and no other pathologies were noted, indicating that neutralization of Tgfa could provide a relatively safe profile as it advances in clinical development.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Transforming Growth Factor alpha/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Neutralizing/metabolism , Antibodies, Neutralizing/pharmacology , Cell Line , Cell Proliferation/drug effects , Epiregulin , Humans , Immunoglobulin G/immunology , Macaca fascicularis , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myofibroblasts/cytology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Protein Binding , Transforming Growth Factor alpha/genetics
18.
Thorax ; 67(11): 985-92, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22735687

ABSTRACT

BACKGROUND: Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. METHODS: Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. RESULTS: During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. CONCLUSIONS: The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.


Subject(s)
Fas Ligand Protein/metabolism , Neutrophils/immunology , Phagocytes/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Tumor Necrosis Factor, Member 6b/pharmacology , Animals , Disease Models, Animal , Fas Ligand Protein/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neutrophils/drug effects , Phagocytes/drug effects , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/therapy , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/prevention & control , Signal Transduction/drug effects , Streptococcus pneumoniae/immunology
19.
Drug Metab Dispos ; 40(8): 1545-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584253

ABSTRACT

The pH-dependent binding of IgGs to the neonatal Fc receptor (FcRn) plays a critical role in regulating IgG homeostasis in vivo. Enhancing interactions between Fc and FcRn via protein engineering has been successfully used as an approach for improving the pharmacokinetics of monoclonal antibodies (mAbs). Although the quantitative translatability of the in vitro FcRn affinity enhancement to an in vivo pharmacokinetic benefit has been supported by several studies, there are also published reports indicating a disconnect in this relation. The body of literature suggests there are likely additional biochemical and biophysical properties of the mAbs along with their FcRn affinity that influence the in vivo pharmacokinetics. Herein, we more broadly evaluate the in vitro Fc-FcRn interactions and biochemical properties of five humanized IgG4 antibodies each with two Fc variant sequences (T250Q/M428L and V308P) and their corresponding pharmacokinetics in cynomolgus monkeys. Our findings indicate that the FcRn affinity-pharmacokinetic relationship does not show a direct correlation either across different IgGs or between the two variant sequences within a platform. Other parameters that have been suggested to contribute to mAb pharmacokinetic properties, such as the pH-dependent dissociation of the FcRn-IgG complexes, mAb biophysical properties, and nonspecific/charge binding characteristics of the mAbs, also did not independently explain the differing pharmacokinetic behaviors. Our results suggest that there is likely not a single in vitro parameter that readily predicts in vivo pharmacokinetics, but that the relative contribution and interplay of several factors along with the FcRn binding affinity are important determinants of mAb pharmacokinetic properties.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Animals , Cell Line , Humans , Immunoglobulin G/chemistry , In Vitro Techniques , Macaca fascicularis , Protein Binding
20.
MAbs ; 4(2): 267-73, 2012.
Article in English | MEDLINE | ID: mdl-22377715

ABSTRACT

Engineering monoclonal antibodies (mAbs) with improved binding to the neonatal Fc receptor (FcRn) is a strategy that can extend their in vivo half-life and slow their systemic clearance. Published reports have predominantly characterized the pharmacokinetics of mAbs after intravenous administration. Recently, studies in mice suggest FcRn may also play a role in affecting the subcutaneous bioavailability of mAbs. Herein, we examined whether five mAbs engineered with the T250Q/M428L Fc mutations that improved their FcRn interactions, and subsequently their in vivo pharmacokinetics after intravenous administration, had improved subcutaneous bioavailability compared with their wild-type counterparts in cynomolgus monkeys. Similar to the intravenous administration findings, the pharmacokinetic profiles of our variant mAbs after subcutaneous injection showed improved half-life or clearance. In contrast, a clear effect was not observed on the subcutaneous bioavailability. We expect that while FcRn may play a role in determining mAb subcutaneous bioavailability, multiple biopharmaceutical and physiological factors are likely to influence the success of engineering strategies aimed at targeting this pathway for improving bioavailability.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Histocompatibility Antigens Class I/immunology , Receptors, Fc/immunology , Amino Acid Substitution , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Monoclonal, Murine-Derived/immunology , Biological Availability , Cell Line , Half-Life , Humans , Infusions, Subcutaneous , Macaca fascicularis , Mice , Mutation, Missense , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...