Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
MAbs ; 16(1): 2362789, 2024.
Article En | MEDLINE | ID: mdl-38845069

Bispecific antibodies, including bispecific IgG, are emerging as an important new class of antibody therapeutics. As a result, we, as well as others, have developed engineering strategies designed to facilitate the efficient production of bispecific IgG for clinical development. For example, we have extensively used knobs-into-holes (KIH) mutations to facilitate the heterodimerization of antibody heavy chains and more recently Fab mutations to promote cognate heavy/light chain pairing for efficient in vivo assembly of bispecific IgG in single host cells. A panel of related monospecific and bispecific IgG1 antibodies was constructed and assessed for immunogenicity risk by comparison with benchmark antibodies with known low (Avastin and Herceptin) or high (bococizumab and ATR-107) clinical incidence of anti-drug antibodies. Assay methods used include dendritic cell internalization, T cell proliferation, and T cell epitope identification by in silico prediction and MHC-associated peptide proteomics. Data from each method were considered independently and then together for an overall integrated immunogenicity risk assessment. In toto, these data suggest that the KIH mutations and in vitro assembly of half antibodies do not represent a major risk for immunogenicity of bispecific IgG1, nor do the Fab mutations used for efficient in vivo assembly of bispecifics in single host cells. Comparable or slightly higher immunogenicity risk assessment data were obtained for research-grade preparations of trastuzumab and bevacizumab versus Herceptin and Avastin, respectively. These data provide experimental support for the common practice of using research-grade preparations of IgG1 as surrogates for immunogenicity risk assessment of their corresponding pharmaceutical counterparts.


Antibodies, Bispecific , Immunoglobulin G , Antibodies, Bispecific/immunology , Antibodies, Bispecific/genetics , Humans , Immunoglobulin G/immunology , Immunoglobulin G/genetics , Risk Assessment , Trastuzumab/immunology , Trastuzumab/genetics , Animals , Bevacizumab/immunology , Bevacizumab/genetics , Mutation
2.
Angiogenesis ; 26(2): 279-293, 2023 05.
Article En | MEDLINE | ID: mdl-36459240

PURPOSE: Ongoing angiogenesis renders the tumor endothelium unresponsive to inflammatory cytokines and interferes with adhesion of leukocytes, resulting in escape from immunity. This process is referred to as tumor endothelial cell anergy. We aimed to investigate whether anti-angiogenic agents can overcome endothelial cell anergy and provide pro-inflammatory conditions. EXPERIMENTAL DESIGN: Tissues of renal cell carcinoma (RCC) patients treated with VEGF pathway-targeted drugs and control tissues were subject to RNAseq and immunohistochemical profiling of the leukocyte infiltrate. Analysis of adhesion molecule regulation in cultured endothelial cells, in a preclinical model and in human tissues was performed and correlated to leukocyte infiltration. RESULTS: It is shown that treatment of RCC patients with the drugs sunitinib or bevacizumab overcomes tumor endothelial cell anergy. This treatment resulted in an augmented inflammatory state of the tumor, characterized by enhanced infiltration of all major leukocyte subsets, including T cells, regulatory T cells, macrophages of both M1- and M2-like phenotypes and activated dendritic cells. In vitro, exposure of angiogenic endothelial cells to anti-angiogenic drugs normalized ICAM-1 expression. In addition, a panel of tyrosine kinase inhibitors was shown to increase transendothelial migration of both non-adherent and monocytic leukocytes. In primary tumors of RCC patients, ICAM-1 expression was found to be significantly increased in both the sunitinib and bevacizumab-treated groups. Genomic analysis confirmed the correlation between increased immune cell infiltration and ICAM-1 expression upon VEGF-targeted treatment. CONCLUSION: The results support the emerging concept that anti-angiogenic therapy can boost immunity and show how immunotherapy approaches can benefit from combination with anti-angiogenic compounds.


Angiogenesis Inhibitors , Carcinoma, Renal Cell , Endothelial Cells , Kidney Neoplasms , Neovascularization, Pathologic , Humans , Bevacizumab/immunology , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelium/drug effects , Endothelium/immunology , Endothelium/pathology , Intercellular Adhesion Molecule-1/immunology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Sunitinib/immunology , Sunitinib/pharmacology , Sunitinib/therapeutic use , Vascular Endothelial Growth Factor A/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Neoplasm Invasiveness/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use
3.
Sci Rep ; 11(1): 6328, 2021 03 18.
Article En | MEDLINE | ID: mdl-33737682

Although paclitaxel plus bevacizumab (PB) therapy is an effective chemotherapeutic regimen for HER2-negative advanced breast cancer (ABC), predictive markers for its effectiveness remain undefined. We investigated the usefulness of systemic immunity markers associated with lymphocytes as predictive markers for PB therapy in patients with HER2-negative ABC. We retrospectively reviewed data from 114 patients with HER2-negative ABC who underwent PB therapy from November 2011 to December 2019. We calculated the absolute lymphocyte count (ALC), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) as representative systemic immunity markers. The time to treatment failure (TTF) and overall survival (OS) of the patients with high ALC, low NLR, and high LMR were significantly longer compared with those of the patients with low ALC, high NLR, and low LMR. A multivariable analysis revealed that high ALC, low NLR, and low PLR were independent predictors for TTF and high ALC, low NLR, and high LMR were independent predictors for OS. Systemic immunity markers were significantly associated with longer TTF and OS in patients who underwent PB therapy and may represent predictive markers for PB therapy in patients with HER2-negative ABC.


Bevacizumab/genetics , Breast Neoplasms/immunology , Immunity/genetics , Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Bevacizumab/immunology , Bevacizumab/therapeutic use , Blood Cell Count , Blood Platelets/immunology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Immunity/immunology , Lymphocytes/pathology , Middle Aged , Monocytes/immunology , Neutrophils/immunology , Paclitaxel/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology
4.
Biochemistry ; 59(30): 2776-2781, 2020 08 04.
Article En | MEDLINE | ID: mdl-32672953

The success of bevacizumab (Avastin), a monoclonal antibody (mAb) anticancer drug targeting vascular endothelial growth factor A (VEGF-A), has motivated the development of biosimilars. Establishing target epitope similarity using epitope mapping is a critical step in preclinical mAb biosimilar development. Here we use time-resolved electrospray ionization hydrogen-deuterium exchange (HDX) mass spectrometry to rapidly compare the epitopes of commercial Avastin and a biosimilar in preclinical development (ApoBev) on an extended construct of VEGF-A. The Avastin and ApoBev epitopes determined in our experiments agree with each other and with the known epitope derived from the Avastin Fab domain/truncated VEGF co-crystal structure. However, subtly different allosteric effects observed exclusively at short (millisecond) HDX labeling times may reflect a slightly different binding mode for ApoBev.


Bevacizumab/immunology , Biosimilar Pharmaceuticals/pharmacology , Epitope Mapping , Hydrogen Deuterium Exchange-Mass Spectrometry , Vascular Endothelial Growth Factor A/chemistry , Humans , Kinetics , Microfluidics , Models, Molecular
5.
Nucleic Acids Res ; 47(W1): W482-W489, 2019 07 02.
Article En | MEDLINE | ID: mdl-31069385

The study of contact residues and interfacial waters of antibody-antigen (Ab-Ag) structures could help in understanding the principles of antibody-antigen interactions as well as provide guidance for designing antibodies with improved affinities. Given the rapid pace with which new antibody-antigen structures are deposited in the protein databank (PDB), it is crucial to have computational tools to analyze contact residues and interfacial waters, and investigate them at different levels. In this study, we have developed AppA, a web server that can be used to analyze and compare 3D structures of contact residues and interfacial waters of antibody-antigen complexes. To the best of our knowledge, this is the first web server for antibody-antigen structures equipped with the capability for dissecting the contributions of interfacial water molecules, hydrogen bonds, hydrophobic interactions, van der Waals interactions and ionic interactions at the antibody-antigen interface, and for comparing the structures and conformations of contact residues. Various examples showcase the utility of AppA for such analyses and comparisons that could help in the understanding of antibody-antigen interactions and suggest mutations of contact residues to improve affinities of antibodies. The AppA web server is freely accessible at http://mspc.bii.a-star.edu.sg/minhn/appa.html.


Antigen-Antibody Complex/chemistry , Software , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigen-Antibody Complex/immunology , Bevacizumab/chemistry , Bevacizumab/immunology , Computer Graphics , Internet , Models, Molecular , Ranibizumab/chemistry , Ranibizumab/immunology , Trastuzumab/chemistry , Trastuzumab/immunology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/immunology , Water/chemistry
6.
J Immunol Methods ; 469: 42-46, 2019 06.
Article En | MEDLINE | ID: mdl-30943379

The development of Bevacizumab (Avastin) biosimilar products has grown rapidly over the last ten years as the original Avastin's patent will expire soon. The approval of Avastin biosimilars requires the demonstration of similarity between the biosimilar and the reference product. To support pre-clinical and clinical studies, pharmacokinetic (PK) assays are required to measure the biosimilar and Avastin with comparable precision and accuracy. The PK assay of Avastin employed by Genentech was a Sandwich ELISA which could detect the total drug concentration. However, it was developed in-house and not commercially available. Therefore, in most of the Avastin biosimilar pre-clinical studies, the antibody drug concentrations were measured using an indirect ELISA against coated VEGF, which could only measure the free instead of the total antibody drugs. It failed the essential requirement to develop the biosimilars. In this study, we reported the generation of mouse monoclonal antibodies (mAbs) that specifically recognize Avastin in a VEGF non-competitive manner. Using a pair of non-VEGF competing anti-Avastin mAbs, a Sandwich ELISA was developed with a lower limit of quantitation (LLOQ) at 400 ng/mL and upper limit of quantitation (ULOQ) at 12800 ng/mL. The assay validation was carried out with serum samples from monkey treated with Avastin biosimilar at seven different time points. Our data showed that the Sandwich ELISA kit we developed is sensitive, simple, reproducible and ready for use in human clinical trials.


Angiogenesis Inhibitors/blood , Antibodies, Monoclonal/immunology , Bevacizumab/blood , Biosimilar Pharmaceuticals/blood , Drug Monitoring/methods , Enzyme-Linked Immunosorbent Assay , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antibody Specificity , Bevacizumab/immunology , Bevacizumab/pharmacokinetics , Biosimilar Pharmaceuticals/pharmacokinetics , Female , Haplorhini , Humans , Limit of Detection , Mice, Inbred BALB C , Predictive Value of Tests , Reproducibility of Results
7.
J Pharm Sci ; 108(7): 2358-2366, 2019 07.
Article En | MEDLINE | ID: mdl-30797781

The immunogenicity of protein aggregates has been investigated in numerous studies. Nevertheless, it is still unknown which kind of protein aggregates enhance immunogenicity the most. The ability of the currently used in vitro and in vivo systems regarding their predictability of immunogenicity in humans is often questionable, and results are partially contradictive. In this study, we used a 2D in vitro assay and a complex 3D human artificial lymph node model to predict the immunogenicity of protein aggregates of bevacizumab and adalimumab. The monoclonal antibodies were exposed to different stress conditions such as light, heat, and mechanical stress to trigger the formation of protein aggregates and particles, and samples were analyzed thoroughly. Cells and culture supernatants were harvested and analyzed for dendritic cell marker and cytokines. Our study in the artificial lymph node model revealed that bevacizumab after exposure to heat triggered a TH1- and proinflammatory immune response, whereas no trend of immune responses was seen for adalimumab after exposure to different stress conditions. The human artificial lymph node model represents a new test model for testing the immunogenicity of protein aggregates combining the relevance of a 3D human system with the rather easy handling of an in vitro setup.


Antibody Formation/immunology , Lymph Nodes/immunology , Protein Aggregates/immunology , Adalimumab/immunology , Antibodies, Monoclonal/immunology , Bevacizumab/immunology , Cells, Cultured , Cytokines/immunology , Dendritic Cells/immunology , Humans , Inflammation/immunology , Th1 Cells/immunology
8.
Talanta ; 192: 331-338, 2019 Jan 15.
Article En | MEDLINE | ID: mdl-30348399

This study describes, for the first time, the development of an automated sensitive flow fluorescent noncompetitive immunoassay based on kinetic-exclusion analysis (KinExA) for the quantitative determination of human plasma levels of monoclonal antibodies (mAbs) used for cancer immunotherapy. The assay was adapted on KinExA™ 3200 biosensor and optimized and validated for bevacizumab (BEV) and cetuximab (CET), as representative examples of the mAbs, using their specific antigens. These antigens were the human vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) for BEV and CET, respectively. The limits of detection were 1.28 and 52.64 ng mL-1 for BEV and CET, respectively. The accuracy of the assay was demonstrated with analytical recovery of analytes from spiked plasma at 96.2-104.3 and 96.8-105.3% for BEV and CET, respectively. The precision of the assay was satisfactory as shown by relative standard deviation (RSD) at 2.2-5.7 and 2.5-6.1% for assay of BEV and CET, respectively. The high sensitivity of the assay allowed the use of very small volumes (~ 1 µL) of plasma sample for analysis. Automated analysis by the proposed KinExA-based assay facilitates the processing of large numbers of mAbs-containing specimens in studies of pharmacokinetics (PK), pharmacodynamics (PD), and therapeutic drug monitoring (TDM) of therapeutic mAbs. The proposed assay can be used to overcome the problems encountered in the existing conventional immunoassays for mAbs.


Antineoplastic Agents, Immunological/blood , Bevacizumab/blood , Biosensing Techniques/methods , Cetuximab/blood , Immunoassay/methods , Antineoplastic Agents, Immunological/immunology , Bevacizumab/immunology , Calibration , Cetuximab/immunology , ErbB Receptors/immunology , Fluorescein-5-isothiocyanate/chemistry , Fluorescence , Fluorescent Antibody Technique, Indirect/methods , Humans , Immunoglobulin G/immunology , Limit of Detection , Vascular Endothelial Growth Factor A/immunology
9.
Lancet Gastroenterol Hepatol ; 3(12): 845-855, 2018 12.
Article En | MEDLINE | ID: mdl-30262136

BACKGROUND: BEVZ92 is a proposed biosimilar to bevacizumab. The two molecules have similar physicochemical and functional properties in in-vitro and preclinical studies. In this clinical study, we compared the pharmacokinetic profile, efficacy, safety, and immunogenicity of BEVZ92 with reference bevacizumab as a first-line treatment in patients with metastatic colorectal cancer. METHODS: We did a randomised, open-label trial at 15 centres in Argentina, Brazil, India, Spain, and Ukraine. Eligible patients were aged 18 years or older, had metastatic colorectal cancer with at least one measurable non-irradiated lesion for which first-line chemotherapy was indicated and Eastern Cooperative Oncology Group (ECOG) performance status of 2 or less, had not received previous treatment for advanced disease, and whose bone marrow, hepatic, renal, and coagulation markers were all within normal ranges. Patients were randomly assigned (1:1) to either BEVZ92 or reference bevacizumab (5 mg/kg on day 1 of each cycle every 2 weeks) in combination with fluorouracil, leucovorin, and oxaliplatin (FOLFOX) or fluorouracil, leucovorin, and irinotecan (FOLFIRI). Randomisation was done via a web service based on a stochastic minimisation algorithm and was stratified by chemotherapy regimen (FOLFOX vs FOLFIRI), previous adjuvant therapy (yes vs no), ECOG performance status (0-1 vs 2), and study site. The primary endpoint was the area under the concentration-versus-time curve after a single infusion (AUC0-336h) and at steady state (AUCss)-ie, at cycle 7-in the assessable population, which comprised all treated patients for whom serum concentration measurements were available during the first seven cycles. Bioequivalence was established if the 90% CIs for the ratio of BEVZ92 to reference bevacizumab of the geometric means for AUC0-336h and AUCss were within the acceptance interval of 80-125%. Secondary endpoints included objective response, clinical benefit, and progression-free survival in the intention-to-treat population and immunogenicity and safety profiles in all treated patients. This trial is registered with ClinicalTrials.gov, number NCT02069704, and is closed to new participants, with follow-up completed. FINDINGS: 142 patients were randomly assigned, 71 to the BEVZ92 group and 71 to the reference bevacizumab group. Two participants assigned to BEVZ92 did not receive treatment (one withdrew consent, the other had a serious intestinal obstruction before starting treatment); therefore, the treated population comprised 69 patients in the BEVZ92 group and 71 in the reference bevacizumab group. The geometric mean ratio of AUC0-336h in the BEVZ92 versus the control group was 99·4% (90% CI 90·5-109·0) and of AUCss was 100·0% (90·2-112·0). Objective response (35 [49%] of 71 vs 40 [56%] of 71), clinical benefit (62 [87%] vs 65 [92%]), and progression-free survival (median 10·8 months [95% CI 7·4-11·5] vs 11·1 months [95% CI 8·0-12·8]) were similar in the BEVZ92 and reference bevacizumab groups. No relevant differences were noted between the safety profiles of the two study treatments. Neutropenia was the most common grade 3 or 4 adverse event reported in the BEVZ92 (14 [20%] of 69 patients) and reference bevacizumab (19 [27%] of 71 patients) groups. Serious adverse events occurred in 19 (28%) patients in the BEVZ92 group and 21 (30%) in the reference bevacizumab group. Two patients died because of bevacizumab-related serious adverse events: a sudden death in the BEVZ92 group and a serious large intestinal perforation in the reference bevacizumab group. The occurrence of anti-drug antibodies was low and similar in both treatment groups (two patients in the BEVZ92 group and one in the reference bevacizumab group). INTERPRETATION: Our results suggest that BEVZ92 and reference bevacizumab are pharmacokinetically bioequivalent and have no appreciable differences in efficacy, immunogenicity, and safety profiles as first-line treatment in combination with FOLFOX or FOLFIRI in patients with metastatic colorectal cancer. FUNDING: mAbxience Research SL.


Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Adult , Aged , Antibodies/blood , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/immunology , Area Under Curve , Bevacizumab/adverse effects , Bevacizumab/immunology , Bevacizumab/pharmacokinetics , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/pharmacokinetics , Camptothecin/therapeutic use , Colorectal Neoplasms/pathology , Female , Fluorouracil/therapeutic use , Humans , Intention to Treat Analysis , Leucovorin/therapeutic use , Male , Middle Aged , Neoplasm Metastasis , Organoplatinum Compounds/therapeutic use , Progression-Free Survival , Therapeutic Equivalency
10.
Sci Rep ; 8(1): 11477, 2018 07 31.
Article En | MEDLINE | ID: mdl-30065306

Therapeutic proteins can induce immune responses that affect their safety and efficacy. Product aggregates and innate immune response modulating impurities (IIRMI) are risk factors of product immunogenicity. In this study, we use Intravenous Immunoglobulin (IVIG), Avastin, and Human Serum Albumin (HSA) to explore whether increased aggregates activate innate immune cells or modify the response to IIRMI. We show that increased aggregates (shaken or stirred) in IVIG and Avastin, but not HSA, induced activation of MAPKs (pp38, pERK and pJNK) and transcription of immune-related genes including IL8, IL6, IL1ß, CSF1, CCL2, CCL7, CCL3, CCL24, CXCL2, IRAK1, EGR2, CEBPß, PPARg and TNFSF15 in human PBMC. The immunomodulatory effect was primarily mediated by FcγR, but not by TLR. Interestingly, increased aggregates in IVIG or Avastin magnified innate immune responses to TLR2/4 agonists, but diminished responses to TLR3/9 agonists. This study shows that IIRMI and aggregates can modify the activity of immune cells potentially modifying the milieu where the products are delivered highlighting the complex interplay of different impurities on product immunogenicity risk. Further, we show that aggregates could modify the sensitivity of PBMC-based assays designed to detect IIRMI. Understanding and managing immunogenicity risk is a critical component of product development and regulation.


Immunity, Innate/immunology , Immunoglobulins, Intravenous/immunology , Serum Albumin, Human/immunology , Animals , Antibody Formation/immunology , Bevacizumab/immunology , Cells, Cultured , HEK293 Cells , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/immunology , Transcription, Genetic/immunology
11.
Talanta ; 188: 562-569, 2018 Oct 01.
Article En | MEDLINE | ID: mdl-30029413

This study describes, for the first time, the development and validation of a highly selective and sensitive heterogeneous fluoroimmunoassay (FIA) for the bioanalysis of two monoclonal antibodies (mAbs) used for cancer immunotherapy: bevacizumab (BEV) and cetuximab (CET). The assay combines reliable non-competitive binding of BEV and CET to their specific cell receptor proteins (human vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR), respectively) with the highly specific fluorescence activity of the fluorescein isothiocyanate labeled anti-human IgG (FITC-IgG) used as label. The limits of detection were 14.14 and 1.27 × 103 ng mL-1 for BEV and CET, respectively. The accuracy and precision of the assay were demonstrated. The assay is simple, convenient, and requires very small volume (~ 5 µL) of plasma sample for analysis. The assay can offer high throughput analysis in clinical settings when modern microplates of multiplies of 96 (up to 6144-wells) are used and/or integrated as a part of automated robotic system. The proposed assay can be used for routine clinical bioanalysis of mAbs with potential application in pharmacokinetics, pharmacodynamics and therapeutic drug monitoring (TDM).


Bevacizumab/analysis , Cetuximab/analysis , Fluoroimmunoassay/methods , Bevacizumab/immunology , Calibration , Cetuximab/immunology , ErbB Receptors/immunology , Fluorescein-5-isothiocyanate/chemistry , Humans , Immunoglobulin G/immunology , Limit of Detection , Vascular Endothelial Growth Factor A/immunology
12.
Cancer Chemother Pharmacol ; 82(2): 339-351, 2018 08.
Article En | MEDLINE | ID: mdl-29905898

PURPOSE: MINT1526A is a monoclonal antibody that blocks the interaction of integrin alpha 5 beta 1 (α5ß1) with its extracellular matrix ligands. This phase I study evaluated the safety and pharmacokinetics of MINT1526A with or without bevacizumab in patients with advanced solid tumors. METHODS: MINT1526A was administered every 3 weeks (Q3W) as monotherapy (arm 1) or in combination with bevacizumab 15 mg/kg, Q3W (arm 2). Each arm included a 3 + 3 dose-escalation stage and a dose-expansion stage. RESULTS: Twenty-four patients were enrolled in arm 1 (dose range 2-30 mg/kg) and 30 patients were enrolled in arm 2 (dose range 3-15 mg/kg). Monocyte α5ß1 receptor occupancy was saturated at a dose of 15 mg/kg. No dose-limiting toxicities were observed, and the maximum tolerated dose was not reached in either arm. The most common adverse events, regardless of causality, included abdominal pain (25%), diarrhea (25%), nausea (21%), vomiting (21%), and fatigue (21%) in arm 1 and nausea (40%), fatigue (33%), vomiting (30%), dehydration (30%), headache (30%), and hypertension (30%) in arm 2. No grade ≥ 3 bleeding events were observed in either arm. No confirmed partial responses (PR) were observed in arm 1. In arm 2, one patient with thymic carcinoma experienced a confirmed PR and two patients with hepatocellular carcinoma (HCC) experienced durable minor radiographic responses. CONCLUSIONS: MINT1526A, with or without bevacizumab, was well-tolerated. Preliminary evidence of combination efficacy, including in patients with HCC, was observed, but cannot be distinguished from bevacizumab monotherapy in this phase I study.


Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/immunology , Antineoplastic Combined Chemotherapy Protocols/immunology , Bevacizumab/administration & dosage , Bevacizumab/immunology , Dose-Response Relationship, Drug , Female , Humans , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/immunology , Male , Middle Aged , Neoplasms/immunology
13.
J Immunol Methods ; 461: 44-52, 2018 10.
Article En | MEDLINE | ID: mdl-29772250

Ranibizumab (Lucentis®), a humanized antigen-binding fragment (Fab) monoclonal antibody that blocks VEGF-A activity, is currently approved for the treatment of several retinal degenerative diseases. The assessment of drug pharmacokinetics (PK) is essential for evaluating exposure as it relates to drug safety and efficacy. For drugs administered intravitreally, systemic drug levels during the course of clinical studies are typically 100 to 1000-fold lower than those of similar therapeutics dosed intravenously, posing a significant bioanalytical challenge for PK measurements. Thus, the development of a highly-sensitive assay for measuring pg/mL levels of ranibizumab in patients' sera after intravitreal administration was needed to support clinical studies. In this report, we describe the development of a novel method that utilizes a high-affinity murine monoclonal anti-ranibizumab-VEGF-complexes antibody (MARA) reagent to measure ranibizumab in human serum. The assay format utilizes a semi-homogeneous solution phase step using a monoclonal antibody (the MARA) that binds specifically to the ranibizumab-VEGF complex, but not to either alone. This unique reagent exhibited low non-specific binding and high selectivity, increasing signal-to-noise readouts and maximizing assay sensitivity. The resulting MARA enzyme-linked immunosorbent assay (ELISA) has a lower limit of quantification of 15 pg/mL in human serum. In the assay, serum samples are incubated overnight with a mixture containing biotinylated-VEGF and MARA, which form a three-molecule complex with ranibizumab in the sample. These complexes are then captured onto streptavidin-coated wells, followed by enzymatic detection using a horseradish peroxidase-labeled-anti-murine antibody reagent and a colorimetric reaction. The assay conditions were optimized to allow for quantitative detection of "total" ranibizumab levels in serum. The assay was fully validated, establishing its high tolerance to sample matrix, as well as its suitable specificity, accuracy, dilution linearity, as well as intra- and inter-assay precision. The MARA ELISA's novel and unique approach has resulted in a considerably more sensitive ranibizumab PK assay compared to earlier versions of this assay. The MARA ELISA has been used for PK measurements in multiple ranibizumab studies, supporting this drug's life-cycle management and related preclinical and clinical-development studies.


Antibodies, Monoclonal, Murine-Derived/chemistry , Bevacizumab/pharmacokinetics , Retinal Degeneration/blood , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Bevacizumab/immunology , Bevacizumab/therapeutic use , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Male , Mice , Mice, Inbred BALB C , Rabbits , Retinal Degeneration/drug therapy , Retinal Degeneration/immunology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/immunology
14.
Clin Exp Pharmacol Physiol ; 45(7): 659-667, 2018 07.
Article En | MEDLINE | ID: mdl-29543385

With the use of multikinase inhibitors (MKIs) having emerged in recent years, skin toxicities such as hand-foot skin reaction (HFSR) are primary side effects, and they lack effective prediction methods. Here, we updated a previous systematic review by establishing a meta-analysis of the risk of developing HFSR among patients receiving MKIs and antivascular endothelial growth factor antibody. Publications from PubMed and abstracts presented at the American Society of Clinical Oncology Annual Meeting up to February 5, 2015, were searched to identify relevant studies, and a total of 236 patients with metastatic tumours in nine trials were included for analysis. In the meta-analysis, the pooled incidence rates of all-grade and high-grade HFSR among patients who received the combination therapy were 56.9% [95% confidence interval (CI), 45%-71.1%] and 14.3% (95% CI, 9%-24.2%), respectively, with significant differences observed with MKI monotherapy (P < .05). Further subgroup analysis demonstrated that increasing the dosages of bevacizumab (77.8% vs 51.1%, P = .04) and MKIs (64.3% vs 52.6%, P = .02) significantly increased HFSR incidence. Moreover, combination with chemotherapy exerted a minimal effect on HFSR risk (61% vs 55.3%, P = .5). This updated review and meta-analysis confirm the increased risk of HFSR incidence due to the use of MKIs and antivascular endothelial growth factor antibody. Thus, using these therapies requires safety standards.


Bevacizumab/pharmacology , Foot , Hand , Protein Kinase Inhibitors/adverse effects , Skin/drug effects , Vascular Endothelial Growth Factor A/immunology , Animals , Bevacizumab/immunology , Humans , Risk
15.
Anal Biochem ; 540-541: 30-37, 2018 01 01.
Article En | MEDLINE | ID: mdl-29128290

Therapeutic monoclonal antibodies (mAbs) are developed for treatment of diverse cancers and autoimmune diseases. For expansion of mAbs approval against unapproved diseases and pharmaceutical development, pharmacokinetics study is very important. Bioanalysis provides one of the most essential index against pharmacokinetics information. So far, we developed useful method for bioanalysis of mAbs in plasma or serum, nSMOL: nano-surface and molecular-orientation limited proteolysis. This method can provide accurate and reproducible value of mAbs content in plasma. Quantification of mAbs using ELISA is strongly influenced by endogenous ligand or anti-drug antibodies. In this report, we exhibited the role of nSMOL proteolysis coupled to LC-MS/MS analysis against quantification of mAbs bound to some binding molecules. The ligands against mAbs do not affect quantification of mAbs concentration in plasma using nSMOL proteolysis. On the other hands, some anti-drug antibodies (ADA), such as idiotypic antibodies, inhibit quantification of mAbs using nSMOL proteolysis. Acid dissociation has some efficacy in accurate value of quantitation of ADA binding mAbs using nSMOL proteolysis coupled to LC-MS/MS analysis. Accordingly, we consider that nSMOL method will contribute to understanding of mAb PK data and therapeutic reference combining with ADA measurements.


Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal/blood , Bevacizumab/blood , Enzyme-Linked Immunosorbent Assay , Trastuzumab/blood , Animals , Antibodies, Anti-Idiotypic/blood , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antigen-Antibody Reactions , Bevacizumab/immunology , Bevacizumab/pharmacokinetics , Chromatography, High Pressure Liquid , Half-Life , Humans , Nanoparticles/chemistry , Proteolysis , Surface Properties , Tandem Mass Spectrometry , Trastuzumab/immunology , Trastuzumab/pharmacokinetics
16.
PLoS One ; 12(10): e0186046, 2017.
Article En | MEDLINE | ID: mdl-29023507

Administration of high dose intravenous immunoglobulin (IVIg) is widely used in the clinic to treat autoimmune and severe inflammatory diseases. However, its mechanisms of action remain poorly understood. We assessed the impact of IVIg on immune cell populations using an in vivo ovalbumin (Ova)-immunization mouse model. High dose IVIg significantly reduced the Ova-specific antibody response. Intriguingly, the results obtained indicate an immediate and massive immune reaction against IVIg, as shown by the activation and expansion of B cells and CD4+ T cells in the spleen and draining lymph nodes and the production of IVIg-specific antibodies. We propose that IVIg competes at the T-cell level with the response against Ova to explain the immunomodulatory properties of IVIg. Two monoclonal antibodies did not succeeded in reproducing the effects of IVIg. This suggests that in addition to the mouse response against human constant domains, the enormous sequence diversity of IVIg may significantly contribute to this massive immune response against IVIg. While correlation of these findings to IVIg-treated patients remains to be explored, our data demonstrate for the first time that IVIg re-directs the immune response towards IVIg and away from a specific antigen response.


Antibody Formation/immunology , Immunoglobulins, Intravenous/immunology , Ovalbumin/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibody Formation/drug effects , Antigens/immunology , B-Lymphocytes/immunology , Bevacizumab/immunology , Bevacizumab/pharmacology , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Epitopes/immunology , Female , Humans , Immunoglobulins, Intravenous/administration & dosage , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice, Inbred C57BL , Spleen/drug effects , Spleen/immunology , Thymus Gland/drug effects , Thymus Gland/immunology
17.
Clin Cancer Res ; 23(22): 7059-7071, 2017 Nov 15.
Article En | MEDLINE | ID: mdl-28912141

Purpose: Bevacizumab, a humanized monoclonal antibody to VEGF, is used routinely in the treatment of patients with recurrent glioblastoma (GBM). However, very little is known regarding the effects of bevacizumab on the cells in the perivascular space in tumors.Experimental Design: Established orthotopic xenograft and syngeneic models of GBM were used to determine entry of monoclonal anti-VEGF-A into, and uptake by cells in, the perivascular space. Based on the results, we examined CD133+ cells derived from GBM tumors in vitro Bevacizumab internalization, trafficking, and effects on cell survival were analyzed using multilabel confocal microscopy, immunoblotting, and cytotoxicity assays in the presence/absence of inhibitors.Results: In the GBM mouse models, administered anti-mouse-VEGF-A entered the perivascular tumor niche and was internalized by Sox2+/CD44+ tumor cells. In the perivascular tumor cells, bevacizumab was detected in the recycling compartment or the lysosomes, and increased autophagy was found. Bevacizumab was internalized rapidly by CD133+/Sox2+-GBM cells in vitro through macropinocytosis with a fraction being trafficked to a recycling compartment, independent of FcRn, and a fraction to lysosomes. Bevacizumab treatment of CD133+ GBM cells depleted VEGF-A and induced autophagy thereby improving cell survival. An inhibitor of lysosomal acidification decreased bevacizumab-induced autophagy and increased cell death. Inhibition of macropinocytosis increased cell death, suggesting macropinocytosis of bevacizumab promotes CD133+ cell survival.Conclusions: We demonstrate that bevacizumab is internalized by Sox2+/CD44+-GBM tumor cells residing in the perivascular tumor niche. Macropinocytosis of bevacizumab and trafficking to the lysosomes promotes CD133+ cell survival, as does the autophagy induced by bevacizumab depletion of VEGF-A. Clin Cancer Res; 23(22); 7059-71. ©2017 AACR.


Antineoplastic Agents, Immunological/pharmacology , Bevacizumab/pharmacology , Glioblastoma/immunology , Glioblastoma/metabolism , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/metabolism , Pinocytosis/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Actins/metabolism , Animals , Antineoplastic Agents, Immunological/immunology , Autophagy/drug effects , Bevacizumab/immunology , Biomarkers , Cell Line, Tumor , Cell Membrane/metabolism , Disease Models, Animal , Endosomes/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Immunophenotyping , Lysosomes/metabolism , Mice , Xenograft Model Antitumor Assays
18.
J Immunol Methods ; 451: 28-36, 2017 12.
Article En | MEDLINE | ID: mdl-28827189

Next generation sequencing (NGS) is widely applied in immunological research, but has yet to become common in antibody epitope mapping. A method utilizing a 12-mer random peptide library expressed in bacteria coupled with magnetic-based cell sorting and NGS correctly identified >75% of epitope residues on the antigens of two monoclonal antibodies (trastuzumab and bevacizumab). PepSurf, a web-based computational method designed for structural epitope mapping was utilized to compare peptides in libraries enriched for monoclonal antibody (mAb) binders to antigen surfaces (HER2 and VEGF-A). Compared to mimotopes recovered from Sanger sequencing of plated colonies from the same sorting protocol, motifs derived from sets of the NGS data improved epitope prediction as defined by sensitivity and precision, from 18% to 82% and 0.27 to 0.51 for trastuzumab and 47% to 76% and 0.19 to 0.27 for bevacizumab. Specificity was similar for Sanger and NGS, 99% and 97% for trastuzumab and 66% and 67% for bevacizumab. These results indicate that combining peptide library screening with NGS yields epitope motifs that can improve prediction of structural epitopes.


Antibodies, Monoclonal/metabolism , Antineoplastic Agents, Immunological/metabolism , Bevacizumab/metabolism , Epitope Mapping/methods , Epitopes , High-Throughput Nucleotide Sequencing , Peptide Library , Receptor, ErbB-2/genetics , Trastuzumab/metabolism , Vascular Endothelial Growth Factor A/genetics , Algorithms , Amino Acid Motifs , Antibodies, Monoclonal/immunology , Antibody Specificity , Antineoplastic Agents, Immunological/immunology , Bevacizumab/immunology , Binding Sites, Antibody , Computational Biology , Databases, Genetic , Immunomagnetic Separation , Models, Chemical , Protein Binding , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Structure-Activity Relationship , Trastuzumab/immunology , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism
20.
Proc Natl Acad Sci U S A ; 113(44): 12532-12537, 2016 11 01.
Article En | MEDLINE | ID: mdl-27791128

Therapeutic targeting of the VEGF signaling axis by the VEGF-neutralizing monoclonal antibody bevacizumab has clearly demonstrated clinical benefit in cancer patients. To improve this strategy using a polyclonal approach, we developed a vaccine targeting VEGF using 3D-structured peptides that mimic the bevacizumab binding site. An in-depth study on peptide optimization showed that the antigen's 3D structure is essential to achieve neutralizing antibody responses. Peptide 1 adopts a clear secondary, native-like structure, including the typical cysteine-knot fold, as evidenced by CD spectroscopy. Binding and competition studies with bevacizumab in ELISA and surface plasmon resonance analysis revealed that peptide 1 represents the complete bevacizumab binding site, including the hairpin loop (ß5-turn-ß6) and the structure-supporting ß2-α2-ß3 loop. Vaccination with peptide 1 elicited high titers of cross-reactive antibodies to VEGF, with potent neutralizing activity. Moreover, vaccination-induced antisera displayed strong angiostatic and tumor-growth-inhibiting properties in a preclinical mouse model for colorectal carcinoma, whereas antibodies raised with peptides exclusively encompassing the ß5-turn-ß6 loop (peptides 15 and 20) did not. Immunization with peptide 1 or 7 (murine analog of 1) in combination with the potent adjuvant raffinose fatty acid sulfate ester (RFASE) showed significant inhibition of tumor growth in the B16F10 murine melanoma model. Based on these data, we conclude that this vaccination technology, which is currently being investigated in a phase I clinical trial (NCT02237638), can potentially outperform currently applied anti-VEGF therapeutics.


Bevacizumab/therapeutic use , Colonic Neoplasms/drug therapy , Peptides/therapeutic use , Vaccination/methods , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Bevacizumab/immunology , Binding Sites/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cross Reactions/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Targeted Therapy/methods , Peptides/chemistry , Peptides/immunology , Rats, Wistar , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
...