Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cancer Immunol Immunother ; 67(2): 247-259, 2018 02.
Article in English | MEDLINE | ID: mdl-29067496

ABSTRACT

Strong evidence exists supporting the important role T cells play in the immune response against tumors. Still, the ability to initiate tumor-specific immune responses remains a challenge. Recent clinical trials suggest that bispecific antibody-mediated retargeted T cells are a promising therapeutic approach to eliminate hematopoietic tumors. However, this approach has not been validated in solid tumors. PF-06671008 is a dual-affinity retargeting (DART®)-bispecific protein engineered with enhanced pharmacokinetic properties to extend in vivo half-life, and designed to engage and activate endogenous polyclonal T cell populations via the CD3 complex in the presence of solid tumors expressing P-cadherin. This bispecific molecule elicited potent P-cadherin expression-dependent cytotoxic T cell activity across a range of tumor indications in vitro, and in vivo in tumor-bearing mice. Regression of established tumors in vivo was observed in both cell line and patient-derived xenograft models engrafted with circulating human T lymphocytes. Measurement of in vivo pharmacodynamic markers demonstrates PF-06671008-mediated T cell activation, infiltration and killing as the mechanism of tumor inhibition.


Subject(s)
Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Cadherins/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , CD3 Complex/immunology , Cell Line, Tumor , Cricetinae , Cricetulus , Female , HCT116 Cells , HT29 Cells , Humans , Mice , Xenograft Model Antitumor Assays
2.
Proc Natl Acad Sci U S A ; 112(50): 15354-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621728

ABSTRACT

Although humanized antibodies have been highly successful in the clinic, all current humanization techniques have potential limitations, such as: reliance on rodent hosts, immunogenicity due to high non-germ-line amino acid content, v-domain destabilization, expression and formulation issues. This study presents a technology that generates stable, soluble, ultrahumanized antibodies via single-step complementarity-determining region (CDR) germ-lining. For three antibodies from three separate key immune host species, binary substitution CDR cassettes were inserted into preferred human frameworks to form libraries in which only the parental or human germ-line destination residue was encoded at each position. The CDR-H3 in each case was also augmented with 1 ± 1 random substitution per clone. Each library was then screened for clones with restored antigen binding capacity. Lead ultrahumanized clones demonstrated high stability, with affinity and specificity equivalent to, or better than, the parental IgG. Critically, this was mainly achieved on germ-line frameworks by simultaneously subtracting up to 19 redundant non-germ-line residues in the CDRs. This process significantly lowered non-germ-line sequence content, minimized immunogenicity risk in the final molecules and provided a heat map for the essential non-germ-line CDR residue content of each antibody. The ABS technology therefore fully optimizes the clinical potential of antibodies from rodents and alternative immune hosts, rendering them indistinguishable from fully human in a simple, single-pass process.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Complementarity Determining Regions/immunology , Germ Cells/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Specificity/immunology , Clone Cells , Complementarity Determining Regions/chemistry , Computer Simulation , Enzyme-Linked Immunosorbent Assay , Epitopes, T-Lymphocyte/immunology , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Peptide Library , Protein Stability , Protein Structure, Tertiary , Rats , Sequence Alignment , Sequence Analysis, Protein , tau Proteins/chemistry , tau Proteins/immunology
3.
Proc Natl Acad Sci U S A ; 111(5): 1766-71, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24443552

ABSTRACT

Using an expanded genetic code, antibodies with site-specifically incorporated nonnative amino acids were produced in stable cell lines derived from a CHO cell line with titers over 1 g/L. Using anti-5T4 and anti-Her2 antibodies as model systems, site-specific antibody drug conjugates (NDCs) were produced, via oxime bond formation between ketones on the side chain of the incorporated nonnative amino acid and hydroxylamine functionalized monomethyl auristatin D with either protease-cleavable or noncleavable linkers. When noncleavable linkers were used, these conjugates were highly stable and displayed improved in vitro efficacy as well as in vivo efficacy and pharmacokinetic stability in rodent models relative to conventional antibody drug conjugates conjugated through either engineered surface-exposed or reduced interchain disulfide bond cysteine residues. The advantages of the oxime-bonded, site-specific NDCs were even more apparent when low-antigen-expressing (2+) target cell lines were used in the comparative studies. NDCs generated with protease-cleavable linkers demonstrated that the site of conjugation had a significant impact on the stability of these rationally designed prodrug linkers. In a single-dose rat toxicology study, a site-specific anti-Her2 NDC was well tolerated at dose levels up to 90 mg/kg. These experiments support the notion that chemically defined antibody conjugates can be synthesized in commercially relevant yields and can lead to antibody drug conjugates with improved properties relative to the heterogeneous conjugates formed by nonspecific chemical modification.


Subject(s)
Antibodies/metabolism , Immunoconjugates/metabolism , Pharmaceutical Preparations/chemical synthesis , Protein Engineering/methods , Animals , Antibodies/blood , Antibodies/chemistry , Antibodies/toxicity , Batch Cell Culture Techniques , CHO Cells , Cell Death/drug effects , Cell Line , Cricetinae , Cricetulus , Cysteine/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/toxicity , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemistry , Protein Stability/drug effects , Rats
4.
J Biol Chem ; 288(24): 17408-19, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23632026

ABSTRACT

The immunoglobulin new antigen receptors (IgNARs) are a class of Ig-like molecules of the shark immune system that exist as heavy chain-only homodimers and bind antigens by their single domain variable regions (V-NARs). Following shark immunization and/or in vitro selection, V-NARs can be generated as soluble, stable, and specific high affinity monomeric binding proteins of ∼12 kDa. We have previously isolated a V-NAR from an immunized spiny dogfish shark, named E06, that binds specifically and with high affinity to human, mouse, and rat serum albumins. Humanization of E06 was carried out by converting over 60% of non-complementarity-determining region residues to those of a human germ line Vκ1 sequence, DPK9. The resulting huE06 molecules have largely retained the specificity and affinity of antigen binding of the parental V-NAR. Crystal structures of the shark E06 and its humanized variant (huE06 v1.1) in complex with human serum albumin (HSA) were determined at 3- and 2.3-Å resolution, respectively. The huE06 v1.1 molecule retained all but one amino acid residues involved in the binding site for HSA. Structural analysis of these V-NARs has revealed an unusual variable domain-antigen interaction. E06 interacts with HSA in an atypical mode that utilizes extensive framework contacts in addition to complementarity-determining regions that has not been seen previously in V-NARs. On the basis of the structure, the roles of various elements of the molecule are described with respect to antigen binding and V-NAR stability. This information broadens the general understanding of antigen recognition and provides a framework for further design and humanization of shark IgNARs.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Sharks/immunology , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Crystallography, X-Ray , Fish Proteins , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Engineering , Protein Structure, Quaternary , Protein Structure, Secondary , Rats , Sequence Homology, Amino Acid , Serum Albumin/chemistry
5.
Immunology ; 143(3): 416-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24831554

ABSTRACT

Interleukin-4 (IL-4) and IL-13 are critical drivers of immune activation and inflammation in ulcerative colitis, asthma and other diseases. Because these cytokines may have redundant function, dual targeting holds promise for achieving greater efficacy. We have recently described a bifunctional therapeutic targeting IL-4 and IL-13 developed on a novel protein scaffold, generated by combining specific binding domains in an optimal configuration using appropriate linker regions. In the current study, the bifunctional IL-4/IL-13 antagonist was evaluated in the murine oxazolone-induced colitis model, which produces disease with features of ulcerative colitis. The bifunctional IL-4/IL-13 antagonist reduced body weight loss throughout the 7-day course of the model, and ameliorated the increased colon weight and decreased colon length that accompany disease. Colon tissue gene expression was modulated in accordance with the treatment effect. Concentrations of serum amyloid P were elevated in proportion to disease severity, making it an effective biomarker. Serum concentrations of the bifunctional IL-4/IL-13 antagonist were inversely proportional to disease severity, colon tissue expression of pro-inflammatory genes, and serum amyloid P concentration. Taken together, these results define a panel of biomarkers signifying engagement of the IL-4/IL-13 pathway, confirm the T helper type 2 nature of disease in this model, and demonstrate the effectiveness of dual cytokine blockade.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colitis, Ulcerative/metabolism , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Mice , Oxazolone/adverse effects , Recombinant Fusion Proteins/administration & dosage , Serum Amyloid A Protein/metabolism , Serum Amyloid P-Component/metabolism , Severity of Illness Index
6.
J Immunol ; 188(1): 322-33, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22131336

ABSTRACT

Examination of 1269 unique naive chicken V(H) sequences showed that the majority of positions in the framework (FW) regions were maintained as germline, with high mutation rates observed in the CDRs. Many FW mutations could be clearly related to the modulation of CDR structure or the V(H)-V(L) interface. CDRs 1 and 2 of the V(H) exhibited frequent mutation in solvent-exposed positions, but conservation of common structural residues also found in human CDRs at the same positions. In comparison with humans and mice, the chicken CDR3 repertoire was skewed toward longer sequences, was dominated by small amino acids (G/S/A/C/T), and had higher cysteine (chicken, 9.4%; human, 1.6%; and mouse, 0.25%) but lower tyrosine content (chicken, 9.2%; human, 16.8%; and mouse 26.4%). A strong correlation (R(2) = 0.97) was observed between increasing CDR3 length and higher cysteine content. This suggests that noncanonical disulfides are strongly favored in chickens, potentially increasing CDR stability and complexity in the topology of the combining site. The probable formation of disulfide bonds between CDR3 and CDR1, FW2, or CDR2 was also observed, as described in camelids. All features of the naive repertoire were fully replicated in the target-selected, phage-displayed repertoire. The isolation of a chicken Fab with four noncanonical cysteines in the V(H) that exhibits 64 nM (K(D)) binding affinity for its target proved these constituents to be part of the humoral response, not artifacts. This study supports the hypothesis that disulfide bond-constrained CDR3s are a structural diversification strategy in the restricted germline v-gene repertoire of chickens.


Subject(s)
Amino Acid Substitution , Chickens/genetics , Complementarity Determining Regions/genetics , Immunoglobulin Heavy Chains/genetics , Mutation , Animals , Antibody Affinity/genetics , Camelus/genetics , Camelus/immunology , Chickens/immunology , Complementarity Determining Regions/immunology , Disulfides/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Mice , Protein Stability , Species Specificity
7.
Am J Respir Cell Mol Biol ; 49(1): 37-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23449738

ABSTRACT

IL-4 and IL-13 comprise promising targets for therapeutic interventions in asthma and other Th2-associated diseases, but agents targeting either IL-4 or IL-13 alone have shown limited efficacy in human clinical studies. Because these cytokines may involve redundant function, dual targeting holds promise for achieving greater efficacy. We describe a bifunctional therapeutic targeting IL-4 and IL-13, developed by a combination of specific binding domains. IL-4-targeted and IL-13-targeted single chain variable fragments were joined in an optimal configuration, using appropriate linker regions on a novel protein scaffold. The bifunctional IL-4/IL-13 antagonist displayed high affinity for both cytokines. It was a potent and efficient neutralizer of both murine IL-4 and murine IL-13 bioactivity in cytokine-responsive Ba/F3 cells, and exhibited a half-life of approximately 4.7 days in mice. In a murine model of ovalbumin-induced ear swelling, the bifunctional molecule blocked both the IL-4/IL-13-dependent early-phase response and the IL-4-dependent late-phase response. In the ovalbumin-induced lung inflammation model, the bifunctional IL-4/IL-13 antagonist reduced the IL-4-dependent rise in serum IgE titers, and reduced IL-13-dependent airway hyperresponsiveness, lung inflammation, mucin gene expression, and serum chitinase responses. Taken together, these findings demonstrate the effective dual blockade of IL-4 and IL-13 with a single agent, which resulted in the modulation of a more extensive range of endpoints than could be achieved by targeting either cytokine alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunoglobulin E/immunology , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Pneumonia/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/immunology , Binding Sites , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , CHO Cells , Cricetinae , Cross-Linking Reagents/chemistry , Ear/physiopathology , Female , Half-Life , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Mice , Mice, Inbred C57BL , Molecular Conformation , Neutralization Tests , Ovalbumin/adverse effects , Ovalbumin/immunology , Pneumonia/immunology , Protein Binding , Protein Interaction Domains and Motifs , Single-Chain Antibodies/metabolism
8.
J Immunol ; 187(1): 561-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622864

ABSTRACT

Human studies using Abs to two different, nonoverlapping epitopes of IL-13 suggested that epitope specificity can have a clinically significant impact on clearance of IL-13. We propose that Ab modulation of IL-13 interaction with IL-13Rα2 underlies this effect. Two Abs were administered to healthy subjects and mild asthmatics in separate dose-ranging studies and allergen-challenge studies. IMA-638 allows IL-13 interaction with IL-13Rα1 or IL-13Rα2 but blocks recruitment of IL-4Rα to the IL-13/IL-13Rα1 complex, whereas IMA-026 competes with IL-13 interaction with IL-13Rα1 and IL-13Rα2. We found ∼10-fold higher circulating titer of captured IL-13 in subjects treated with IMA-026 compared with those administered IMA-638. To understand how this difference could be related to epitope, we asked whether either Ab affects IL-13 internalization through cell surface IL-13Rα2. Humans inducibly express cell surface IL-13Rα2 but lack the soluble form that regulates IL-13 responses in mice. Cells with high IL-13Rα2 expression rapidly and efficiently depleted extracellular IL-13, and this activity persisted in the presence of IMA-638 but not IMA-026. The potency and efficiency of this clearance pathway suggest that cell surface IL-13Rα2 acts as a scavenger for IL-13. These findings could have important implications for the design and characterization of IL-13 antagonists.


Subject(s)
Interleukin-13 Receptor alpha2 Subunit/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Isoantibodies/physiology , Receptors, Scavenger/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Dose-Response Relationship, Immunologic , Drug Delivery Systems , Extracellular Space/immunology , Extracellular Space/metabolism , HT29 Cells , Humans , Interleukin-13/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/biosynthesis , Macaca fascicularis , Mice , Mice, Inbred BALB C , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/physiology
9.
Sci Rep ; 12(1): 7262, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508689

ABSTRACT

Next-generation site-specific cysteine-based antibody-drug-conjugates (ADCs) broaden therapeutic index by precise drug-antibody attachments. However, manufacturing such ADCs for clinical validation requires complex full reduction and reoxidation processes, impacting product quality. To overcome this technical challenge, we developed a novel antibody manufacturing process through cysteine (Cys) metabolic engineering in Chinese hamster ovary cells implementing a unique cysteine-capping technology. This development enabled a direct conjugation of drugs after chemoselective-reduction with mild reductant tris(3-sulfonatophenyl)phosphine. This innovative platform produces clinical ADC products with superior quality through a simplified manufacturing process. This technology also has the potential to integrate Cys-based site-specific conjugation with other site-specific conjugation methodologies to develop multi-drug ADCs and exploit multi-mechanisms of action for effective cancer treatments.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Animals , Antibodies , Antineoplastic Agents/therapeutic use , CHO Cells , Cricetinae , Cricetulus , Cysteine , Disulfides , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Metabolic Engineering
10.
Rheumatology (Oxford) ; 50(6): 1033-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21258049

ABSTRACT

OBJECTIVES: To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. METHODS: Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10 mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. RESULTS: 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. CONCLUSIONS: SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD20/drug effects , Antigens, CD20/immunology , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites , Cells, Cultured , Disease Models, Animal , Humans , Immunologic Factors/pharmacology , In Vitro Techniques , Linear Models , Macaca fascicularis , Random Allocation , Rituximab , Sensitivity and Specificity
11.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095881

ABSTRACT

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Immunoconjugates/pharmacology , Receptor, Notch3/metabolism , Cell Line, Tumor/drug effects , Humans , Immunoconjugates/metabolism , Oncogenes/drug effects , Receptor, Notch3/immunology , Receptors, Notch/antagonists & inhibitors , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 27(2): 622-631, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33148666

ABSTRACT

PURPOSE: Mortality due to acute myeloid leukemia (AML) remains high, and the management of relapsed or refractory AML continues to be therapeutically challenging. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has provided a proof of concept for an ADC-based therapeutic for AML. Several other ADCs have since entered clinical development of AML, but have met with limited success. We sought to develop a next-generation ADC for AML with a wide therapeutic index (TI) that overcomes the shortcomings of previous generations of ADCs. EXPERIMENTAL DESIGN: We compared the TI of our novel CD33-targeted ADC platform with other currently available CD33-targeted ADCs in preclinical models of AML. Next, using this next-generation ADC platform, we performed a head-to-head comparison of two attractive AML antigens, CD33 and CD123. RESULTS: Our novel ADC platform offered improved safety and TI when compared with certain currently available ADC platforms in preclinical models of AML. Differentiation between the CD33- and CD123-targeted ADCs was observed in safety studies conducted in cynomolgus monkeys. The CD33-targeted ADC produced severe hematologic toxicity, whereas minimal hematologic toxicity was observed with the CD123-targeted ADC at the same doses and exposures. The improved toxicity profile of an ADC targeting CD123 over CD33 was consistent with the more restricted expression of CD123 in normal tissues. CONCLUSIONS: We optimized all components of ADC design (i.e., leukemia antigen, antibody, and linker-payload) to develop an ADC that has the potential to translate into an effective new therapy against AML.


Subject(s)
Gemtuzumab/therapeutic use , Immunoconjugates/therapeutic use , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Area Under Curve , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gemtuzumab/immunology , Gemtuzumab/pharmacokinetics , HL-60 Cells , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacokinetics , Interleukin-3 Receptor alpha Subunit/immunology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , Macaca fascicularis , Mice , Sialic Acid Binding Ig-like Lectin 3/immunology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods
13.
MAbs ; 13(1): 1850395, 2021.
Article in English | MEDLINE | ID: mdl-33459147

ABSTRACT

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Subject(s)
Antibodies, Bispecific/immunology , CD3 Complex/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Enterotoxin/immunology , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Bispecific/therapeutic use , Cell Line, Tumor , Female , Humans , Hybridomas , Macaca fascicularis/immunology , Macaca fascicularis/metabolism , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/metabolism , Protein Engineering/methods , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacokinetics , Single-Chain Antibodies/therapeutic use , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Mol Cancer Ther ; 19(10): 2068-2078, 2020 10.
Article in English | MEDLINE | ID: mdl-32747418

ABSTRACT

The approval of ado-trastuzumab emtansine (T-DM1) in HER2+ metastatic breast cancer validated HER2 as a target for HER2-specific antibody-drug conjugates (ADC). Despite its demonstrated clinical efficacy, certain inherent properties within T-DM1 hamper this compound from achieving the full potential of targeting HER2-expressing solid tumors with ADCs. Here, we detail the discovery of PF-06804103, an anti-HER2 ADC designed to have a widened therapeutic window compared with T-DM1. We utilized an empirical conjugation site screening campaign to identify the engineered ĸkK183C and K290C residues as those that maximized in vivo ADC stability, efficacy, and safety for a four drug-antibody ratio (DAR) ADC with this linker-payload combination. PF-06804103 incorporates the following novel design elements: (i) a new auristatin payload with optimized pharmacodynamic properties, (ii) a cleavable linker for optimized payload release and enhanced antitumor efficacy, and (iii) an engineered cysteine site-specific conjugation approach that overcomes the traditional safety liabilities of conventional conjugates and generates a homogenous drug product with a DAR of 4. PF-06804103 shows (i) an enhanced efficacy against low HER2-expressing breast, gastric, and lung tumor models, (ii) overcomes in vitro- and in vivo-acquired T-DM1 resistance, and (iii) an improved safety profile by enhancing ADC stability, pharmacokinetic parameters, and reducing off-target toxicities. Herein, we showcase our platform approach in optimizing ADC design, resulting in the generation of the anti-HER2 ADC, PF-06804103. The design elements of identifying novel sites of conjugation employed in this study serve as a platform for developing optimized ADCs against other tumor-specific targets.


Subject(s)
Breast Neoplasms/drug therapy , Immunoconjugates/therapeutic use , Lung Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Animals , Breast Neoplasms/pathology , Female , Humans , Immunoconjugates/pharmacology , Lung Neoplasms/pathology , Mice , Mice, Nude , Stomach Neoplasms/pathology
15.
Clin Cancer Res ; 26(9): 2188-2202, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31996389

ABSTRACT

PURPOSE: Gastrointestinal cancers remain areas of high unmet need despite advances in targeted and immunotherapies. Here, we demonstrate potent, tumor-selective efficacy with PF-07062119, a T-cell engaging CD3 bispecific targeting tumors expressing Guanylyl Cyclase C (GUCY2C), which is expressed widely across colorectal cancer and other gastrointestinal malignancies. In addition, to address immune evasion mechanisms, we explore combinations with immune checkpoint blockade agents and with antiangiogenesis therapy. EXPERIMENTAL DESIGN: PF-07062119 activity was evaluated in vitro in multiple tumor cell lines, and in vivo in established subcutaneous and orthotopic human colorectal cancer xenograft tumors with adoptive transfer of human T cells. Efficacy was also evaluated in mouse syngeneic tumors using human CD3ε transgenic mice. IHC and mass cytometry were performed to demonstrate drug biodistribution, recruitment of activated T cells, and to identify markers of immune evasion. Combination studies were performed with anti-PD-1/PD-L1 and anti-VEGF antibodies. Toxicity and pharmacokinetic studies were done in cynomolgus macaque. RESULTS: We demonstrate that GUCY2C-positive tumors can be targeted with an anti-GUCY2C/anti-CD3ε bispecific, with selective drug biodistribution to tumors. PF-07062119 showed potent T-cell-mediated in vitro activity and in vivo efficacy in multiple colorectal cancer human xenograft tumor models, including KRAS- and BRAF-mutant tumors, as well as in the immunocompetent mouse syngeneic tumor model. PF-07062119 activity was further enhanced when combined with anti-PD-1/PD-L1 treatment or in combination with antiangiogenic therapy. Toxicity studies in cynomolgus indicated a monitorable and manageable toxicity profile. CONCLUSIONS: These data highlight the potential for PF-07062119 to demonstrate efficacy and improve patient outcomes in colorectal cancer and other gastrointestinal malignancies.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/immunology , Colorectal Neoplasms/therapy , Gastrointestinal Neoplasms/therapy , Immunotherapy/methods , Receptors, Enterotoxin/immunology , T-Lymphocytes/immunology , Adoptive Transfer/methods , Animals , Antibodies, Bispecific/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Disease Models, Animal , Female , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Tissue Distribution
16.
Mol Cell Endocrinol ; 302(1): 26-32, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19356623

ABSTRACT

UNLABELLED: Methodological problems, including binding of myostatin to plasma proteins and cross-reactivity of assay reagents with other proteins, have confounded myostatin measurements. Here we describe development of an accurate assay for measuring myostatin concentrations in humans. Monoclonal antibodies that bind to distinct regions of myostatin served as capture and detector antibodies in a sandwich ELISA that used acid treatment to dissociate myostatin from binding proteins. Serum from myostatin-deficient Belgian Blue cattle was used as matrix and recombinant human myostatin as standard. The quantitative range was 0.15-37.50 ng/mL. Intra- and inter-assay CVs in low, mid, and high range were 4.1%, 4.7%, and 7.2%, and 3.9%, 1.6%, and 5.2%, respectively. Myostatin protein was undetectable in sera of Belgian Blue cattle and myostatin knockout mice. Recovery in spiked sera approximated 100%. ActRIIB-Fc or anti-myostatin antibody MYO-029 had no effect on myostatin measurements when assayed at pH 2.5. Myostatin levels were higher in young than older men (mean+/-S.E.M. 8.0+/-0.3 ng/mL vs. 7.0+/-0.4 ng/mL, P=0.03). In men treated with graded doses of testosterone, myostatin levels were significantly higher on day 56 than baseline in both young and older men; changes in myostatin levels were significantly correlated with changes in total and free testosterone in young men. Myostatin levels were not significantly associated with lean body mass in either young or older men. CONCLUSION: Myostatin ELISA has the characteristics of a valid assay: nearly 100% recovery, excellent precision, accuracy, and sufficient sensitivity to enable measurement of myostatin concentrations in men and women.


Subject(s)
Androgens/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Myostatin/blood , Testosterone/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Androgens/administration & dosage , Animals , Cattle , Female , Humans , Male , Middle Aged , Reproducibility of Results , Sensitivity and Specificity , Testosterone/administration & dosage , Young Adult
17.
Mucosal Immunol ; 12(5): 1174-1186, 2019 09.
Article in English | MEDLINE | ID: mdl-31308480

ABSTRACT

There continues to be a major need for more effective inflammatory bowel disease (IBD) therapies. IL-13Rα2 is a decoy receptor that binds the cytokine IL-13 with high affinity and diminishes its STAT6-mediated effector functions. Previously, we found that IL-13Rα2 was necessary for IBD in mice deficient in the anti-inflammatory cytokine IL-10. Here, we tested for the first time a therapeutic antibody specifically targeting IL-13Rα2. We also used the antibody and Il13ra2-/- mice to dissect the role of IL-13Rα2 in IBD pathogenesis and recovery. Il13ra2-/- mice were modestly protected from induction of dextran sodium sulfate (DSS)-induced colitis. Following a 7-day recovery period, Il13ra2-/- mice or wild-type mice administered the IL-13Rα2-neutralizing antibody had significantly improved colon health compared to control mice. Neutralizing IL-13Rα2 to increase IL-13 bioavailability promoted resolution of IBD even if neutralization occurred only during recovery. To link our observations in mice to a large human cohort, we conducted a phenome-wide association study of a more active variant of IL-13 (R130Q) that has reduced affinity for IL-13Rα2. Human subjects carrying R130Q reported a lower risk for Crohn's disease. Our findings endorse moving anti-IL-13Rα2 into preclinical drug development with the goal of accelerating recovery and maintaining remission in Crohn's disease patients.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Inflammatory Bowel Diseases/metabolism , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/metabolism , Animals , Crohn Disease/etiology , Crohn Disease/metabolism , Crohn Disease/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Disease Susceptibility , Eosinophils/immunology , Eosinophils/metabolism , Gain of Function Mutation , Genetic Variation , Humans , Immunity , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/pathology , Interleukin-13 Receptor alpha2 Subunit/genetics , Mice , Odds Ratio
18.
J Pharmacol Exp Ther ; 325(3): 882-92, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18337474

ABSTRACT

Interleukin (IL)-13 is a key cytokine driving allergic and asthmatic responses and contributes to airway inflammation in cynomolgus monkeys after segmental challenge with Ascaris suum antigen. IL-13 bioactivity is mediated by a heterodimeric receptor (IL-13Ralpha1/IL-4Ralpha) and can be inhibited in vitro by targeting IL-13 interaction with either chain. However, in cytokine systems, in vitro neutralization activity may not always predict inhibitory function in vivo. To address the efficacy of two different IL-13 neutralization mechanisms in a primate model of atopic disease, two humanized monoclonal antibodies to IL-13 were generated, with highly homologous properties, differing in epitope recognition. Ab01 blocks IL-13 interaction with IL-4Ralpha, and Ab02 blocks IL-13 interaction with IL-13Ralpha1. In a cynomolgus monkey model of IgE responses to A. suum antigen, both Ab01 and Ab02 effectively reduced serum titers of Ascaris-specific IgE and diminished ex vivo Ascaris-triggered basophil histamine release, assayed 8 weeks after a single administration of antibody. The two antibodies also produced comparable reductions in pulmonary inflammation after lung segmental challenge with Ascaris antigen. Increased serum levels of IL-13, lacking demonstrable biological activity, were seen postchallenge in animals given either anti-IL-13 antibody but not in control animals given human IgG of irrelevant specificity. These findings demonstrate a potent effect of IL-13 neutralization on IgE-mediated atopic responses in a primate system and show that IL-13 can be efficiently neutralized by targeting either the IL-4Ralpha-binding epitope or the IL-13Ralpha1-binding epitope.


Subject(s)
Antigens, Helminth/immunology , Ascaris/immunology , Immunoglobulin E/blood , Immunoglobulin G/immunology , Inflammation/immunology , Interleukin-13/immunology , Lung/immunology , Receptors, Interleukin-13/immunology , Animals , Antibodies, Helminth/immunology , Basophils/immunology , Bronchoalveolar Lavage Fluid/immunology , Epitopes/immunology , Histamine Release/immunology , Humans , Macaca fascicularis , Male
19.
Int Immunopharmacol ; 8(3): 477-83, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18279802

ABSTRACT

Numerous animal and in vitro studies suggest that neutralization of IL-13 is an attractive approach for therapeutic intervention in asthma. In this paper we describe preclinical pharmacokinetics (PK), interspecies scaling, and biodistribution of two humanized anti-IL-13 IgG1 monoclonal antibodies, Ab-01 and Ab-02, with different IL-13 neutralization mechanisms. PK parameters of Ab-01 and Ab-02 following IV or SC dosage to mouse, rat, cynomolgus monkey, and sheep, were similar. After IV administration, the elimination of anti-IL-13 antibodies was slow in all species tested and the serum clearance ranged from 0.13 mL/h/kg in monkeys to 0.81 mL/h/kg in mice. Both anti-IL-13 antibodies appeared to be confined primarily to the vascular space, as volume of distribution was relatively small (<120 mL/kg) in all species and tissue-to-serum concentration ratios (in mice and rats) were low (<0.5) in the tissues examined. The elimination half-life ranged from 3-6 days in mice to 14-17 days in monkey and sheep. In monkeys, PK parameters appeared to be approximately linear in the 1-100 mg/kg dose range. Following SC administration, the bioavailability of anti-IL-13 antibodies was 60-90% in all species tested. PK profile of Ab-02 in the model of acute airway inflammation (induced by Ascaris challenge) was, in general, similar to that in unchallenged monkeys; however, volume of distribution and clearance tended to decrease in Ascaris-challenged animals. Allometric scaling suggested that anti-IL-13 antibodies would likely to have a favorable PK profile, such as slow clearance and long terminal half-life, following IV or SC administration to humans.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Interleukin-13/immunology , Animals , Half-Life , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Rats , Rats, Sprague-Dawley , Sheep , Species Specificity , Tissue Distribution
20.
MAbs ; 10(1): 81-94, 2018 01.
Article in English | MEDLINE | ID: mdl-28991504

ABSTRACT

A large body of data exists demonstrating that neonatal Fc receptor (FcRn) binding of an IgG via its Fc CH2-CH3 interface trends with the pharmacokinetics (PK) of IgG. We have observed that PK of IgG molecules vary widely, even when they share identical Fc domains. This led us to hypothesize that domains distal from the Fc could contribute to FcRn binding and affect PK. In this study, we explored the role of these IgG domains in altering the affinity between IgG and FcRn. Using a surface plasmon resonance-based assay developed to examine the steady-state binding affinity (KD) of IgG molecules to FcRn, we dissected the contributions of IgG domains in modulating the affinity between FcRn and IgG. Through analysis of a broad collection of therapeutic antibodies containing more than 50 unique IgG molecules, we demonstrated that variable domains, and in particular complementarity-determining regions (CDRs), significantly alter binding affinity to FcRn in vitro. Furthermore, a panel of IgG molecules differing only by 1-5 mutations in CDRs altered binding affinity to FcRn in vitro, by up to 79-fold, and the affinity values correlated with calculated isoelectric point values of both variable domains and CDR-L3. In addition, tighter affinity values trend with faster in vivo clearance of a set of IgG molecules differing only by 1-3 mutations in human FcRn transgenic mice. Understanding the role of CDRs in modulation of IgG affinity to FcRn in vitro and their effect on PK of IgG may have far-reaching implications in the optimization of IgG therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Complementarity Determining Regions/metabolism , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibody Specificity , Binding Sites, Antibody , CHO Cells , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Cricetulus , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Male , Mice, Transgenic , Mutation , Pharmacogenomic Variants , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Fc/genetics , Receptors, Fc/immunology
SELECTION OF CITATIONS
SEARCH DETAIL