Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 326
1.
Protein Expr Purif ; 189: 105973, 2022 01.
Article En | MEDLINE | ID: mdl-34560256

The novel anti-PD-L1/TGFBR2-ECD fusion protein (BR102) comprises an anti-PD-L1 antibody (HS636) which is fused at the C terminus of the heavy chain to a TGF-ß1 receptor Ⅱ ectodomain (TGFBR2-ECD), and which can sequester the PD-1/PD-L1 pathway and TGF-ß bioactivity in the immunosuppressive tumor microenvironment. In the expression of TGFBR2-ECD wild-type fused protein (BR102-WT), a 50 kDa clipped species was confirmed to be induced by proteolytic cleavage at a "QKS" site located in the N-terminus of the ectodomain, which resulted in the formation of IgG-like clipping. The matrix metalloproteinase-9 was determined to be associated with BR102-WT digestion. In addition, it was observed that the N-glycosylation modifications of the fusion protein were tightly involved in regulating proteolytic activity and the levels of cleavage could be significantly suppressed by MMP-inhibitors. To avoid proteolytic degradation, eliminating protease-sensitive amino acid motifs and introducing potential glycosylation were performed. Three sensitive motifs were mutated, and the levels of clipping were strongly restrained. The mutant candidates exhibited similar binding affinities to hPD-L1 and hTGF-ß1 as well as highly purified BR102-WT2. Furthermore, the mutants displayed more significant proteolytic resistance than that of BR102-WT2 in the lysate incubation reaction and the plasma stability test. Moreover, the bifunctional candidate Mu3 showed an additive antitumor effect in MC38/hPD-L1 bearing models as compared to that of with anti-PD-L1 antibody alone. In conclusion, in this study, the protease-sensitive features of BR102-WT were well characterized and efficient optimization was performed. The candidate BR102-Mutants exhibited advanced druggability in drug stability and displayed desirable antitumor activity.


Antibodies, Neoplasm/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Colonic Neoplasms/therapy , Protein Processing, Post-Translational , Receptor, Transforming Growth Factor-beta Type II/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CHO Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cricetulus , Female , Glycosylation , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Mice , Mice, Inbred C57BL , Mutation , Protein Domains , Proteolysis , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
2.
Elife ; 102021 10 05.
Article En | MEDLINE | ID: mdl-34608861

Myeloid, T, and NK cells are key players in the elimination phase of cancer immunoediting, also referred to as cancer immunosurveillance. However, the role of B cells and NAbs, which are present prior to the encounter with cognate antigens, has been overlooked. One reason is due to the popular use of a single B cell-deficient mouse model, muMT mice. Cancer models using muMT mice display a similar tumor burden as their wild-type (WT) counterparts. Empirically, we observe what others have previously reported with muMT mice. However, using two other B cell-deficient mouse models (IgHELMD4 and CD19creDTA), we show a three- to fivefold increase in tumor burden relative to WT mice. In addition, using an unconventional, non-cancer-related, immune neoantigen model where hypoxic conditions and cell clustering are absent, we provide evidence that B cells and their innate, natural antibodies (NAbs) are critical for the detection and elimination of neoantigen-expressing cells. Finally, we find that muMT mice display anti-tumor immunity because of an unexpected compensatory mechanism consisting of significantly enhanced type 1 interferon (IFN)-producing plasmacytoid dendritic cells (pDCs), which recruit a substantial number of NK cells to the tumor microenvironment compared to WT mice. Diminishing this compensatory pDC-IFN-NK cell mechanism revealed that muMT mice develop a three- to fivefold increase in tumor burden compared to WT mice. In summary, our findings suggest that NAbs are part of an early defense against not only microorganisms and dying cells, but precancerous cells as well.


Antibodies, Neoplasm/immunology , B-Lymphocytes/immunology , Immunity, Innate/genetics , Animals , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Interferon Type I/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Microenvironment
3.
Front Immunol ; 12: 703574, 2021.
Article En | MEDLINE | ID: mdl-34539634

CD38 is the major NAD+-hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background.


ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Heavy Chains/immunology , Membrane Glycoproteins/immunology , Neoplasms/immunology , ADP-ribosyl Cyclase 1/genetics , Animals , Antibodies, Monoclonal, Murine-Derived/genetics , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Humans , Immunoglobulin Heavy Chains/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout
4.
Theranostics ; 10(25): 11404-11415, 2020.
Article En | MEDLINE | ID: mdl-33052222

Through protein engineering and a novel pegylation strategy, a diabody specific to tumor-associated glycoprotein 72 (TAG-72) (PEG-AVP0458) has been created to optimize pharmacokinetics and bioavailability to tumor. We report the preclinical and clinical translation of PEG-AVP0458 to a first-in-human clinical trial of a diabody. Methods: Clinical translation followed characterization of PEG-AVP0458 drug product and preclinical biodistribution and imaging assessments of Iodine-124 trace labeled PEG-AVP0458 (124I-PEG-AVP0458). The primary study objective of the first-in-human study was the safety of a single protein dose of 1.0 or 10 mg/m2 124I-PEG-AVP0458 in patients with TAG-72 positive relapsed/ metastatic prostate or ovarian cancer. Secondary study objectives were evaluation of the biodistribution, tumor uptake, pharmacokinetics and immunogenicity. Patients were infused with a single-dose of 124I labeled PEG-AVP0458 (3-5 mCi (111-185 MBq) for positron emission tomography (PET) imaging, performed sequentially over a one-week period. Safety, pharmacokinetics, biodistribution, and immunogenicity were assessed up to 28 days after infusion. Results: PEG-AVP0458 was radiolabeled with 124I and shown to retain high TAG-72 affinity and excellent targeting of TAG-72 positive xenografts by biodistribution analysis and PET imaging. In the first-in-human trial, no adverse events or toxicity attributable to 124I-PEG-AVP0458 were observed. Imaging was evaluable in 5 patients, with rapid and highly specific targeting of tumor and minimal normal organ uptake, leading to high tumor:blood ratios. Serum concentration values of 124I-PEG-AVP0458 showed consistent values between patients, and there was no significant difference in T½α and T½ß between dose levels with mean (± SD) results of T½α = 5.10 ± 4.58 hours, T½ß = 46.19 ± 13.06 hours. Conclusions: These data demonstrates the safety and feasibility of using pegylated diabodies for selective tumor imaging and potential delivery of therapeutic payloads in cancer patients.


Antibodies, Bispecific/adverse effects , Antigens, Neoplasm/metabolism , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Radiopharmaceuticals/adverse effects , Adult , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacokinetics , Antibodies, Neoplasm/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Line, Tumor , Dose-Response Relationship, Drug , Feasibility Studies , Female , Humans , Infusions, Intravenous , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/adverse effects , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/adverse effects , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
5.
J Clin Invest ; 130(5): 2673-2688, 2020 05 01.
Article En | MEDLINE | ID: mdl-32310221

Tumor-associated peptide-human leukocyte antigen complexes (pHLAs) represent the largest pool of cell surface-expressed cancer-specific epitopes, making them attractive targets for cancer therapies. Soluble bispecific molecules that incorporate an anti-CD3 effector function are being developed to redirect T cells against these targets using 2 different approaches. The first achieves pHLA recognition via affinity-enhanced versions of natural TCRs (e.g., immune-mobilizing monoclonal T cell receptors against cancer [ImmTAC] molecules), whereas the second harnesses an antibody-based format (TCR-mimic antibodies). For both classes of reagent, target specificity is vital, considering the vast universe of potential pHLA molecules that can be presented on healthy cells. Here, we made use of structural, biochemical, and computational approaches to investigate the molecular rules underpinning the reactivity patterns of pHLA-targeting bispecifics. We demonstrate that affinity-enhanced TCRs engage pHLA using a comparatively broad and balanced energetic footprint, with interactions distributed over several HLA and peptide side chains. As ImmTAC molecules, these TCRs also retained a greater degree of pHLA selectivity, with less off-target activity in cellular assays. Conversely, TCR-mimic antibodies tended to exhibit binding modes focused more toward hot spots on the HLA surface and exhibited a greater degree of crossreactivity. Our findings extend our understanding of the basic principles that underpin pHLA selectivity and exemplify a number of molecular approaches that can be used to probe the specificity of pHLA-targeting molecules, aiding the development of future reagents.


HLA Antigens/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Amino Acid Sequence , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibody Specificity , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Line , Cell Line, Tumor , Crystallography, X-Ray , HLA Antigens/chemistry , HLA Antigens/genetics , Humans , Indicators and Reagents , Models, Molecular , Molecular Dynamics Simulation , Molecular Mimicry/genetics , Molecular Mimicry/immunology , Peptides/chemistry , Peptides/genetics , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology
6.
Gynecol Oncol ; 155(2): 324-330, 2019 11.
Article En | MEDLINE | ID: mdl-31477280

OBJECTIVE: To date, The Cancer Genome Atlas (TCGA) has provided the most extensive molecular characterization of invasive cervical cancer (ICC). Analysis of reverse phase protein array (RPPA) data from TCGA samples showed that cervical cancers could be stratified into 3 clusters exhibiting significant differences in survival outcome: hormone, EMT, and PI3K/AKT. The goals of the current study were to: 1) validate the TCGA RPPA results in an independent cohort of ICC patients and 2) to develop and validate an algorithm encompassing a small antibody set for clinical utility. METHODS: Subjects consisted of 2 ICC patient cohorts with accompanying RPPA and clinical-pathologic data: 155 samples from TCGA (TCGA-155) and 61 additional, unique samples (MCW-61). Using data from 173 common RPPA antibodies, we replicated Silhouette clustering analysis in both ICC cohorts. Further, an index score for each patient was calculated from the survival-associated antibodies (SAAs) identified using Random survival forests (RSF) and the Cox proportional hazard regression model. Kaplan-Meier survival analysis and the log-rank test were performed to assess and compare cluster or risk group survival outcome. RESULTS: In addition to validating the prognostic ability of the proteomic clusters reported by TCGA, we developed an algorithm based on 22 unique antibodies (SAAs) that stratified women with ICC into low-, medium-, or high-risk survival groups. CONCLUSIONS: We provide a signature of 22 antibodies which accurately predicted survival outcome in 2 separate groups of ICC patients. Future studies examining these candidate biomarkers in additional ICC cohorts is warranted to fully determine their clinical potential.


Proteomics , Uterine Cervical Neoplasms/mortality , Adult , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Risk Factors , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology
7.
Mol Biol (Mosk) ; 52(3): 380-393, 2018.
Article Ru | MEDLINE | ID: mdl-29989572

Bispecific antibodies capable of simultaneously binding two targets have been studied for many years with a view to their implementation in clinical practice. Unique biological and pharmacological properties, as well as the diversity of their formats, make it possible to consider bispecific antibodies as promising agents for use in various procedures: from visualization of intracellular processes to targeted anticancer therapy. Bispecific antibodies help to determine more precisely the therapeutic target, thereby increasing the efficiency of therapy and reducing the probability of side effects. The present review describes the main formats of bispecific antibodies, methods for their generation, and possibilities for practical application.


Antibodies, Bispecific , Antibodies, Neoplasm , Neoplasms , Animals , Antibodies, Bispecific/genetics , Antibodies, Bispecific/immunology , Antibodies, Bispecific/therapeutic use , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy
8.
Protein Eng Des Sel ; 31(1): 17-28, 2018 01 01.
Article En | MEDLINE | ID: mdl-29301020

We present a strategy to discover recombinant monoclonal antibodies (mAbs) to specific cancers and demonstrate this approach using basal subtype breast cancers. A phage antibody library was depleted of antibodies to common cell surface molecules by incubation with luminal breast cancer cell lines, and then selected on a single basal-like breast cancer cell line (MDA-MB-231) for binding associated receptor-mediated endocytosis. Additional profiling against two luminal and four basal-like cell lines revealed 61 unique basal-specific mAbs from a pool of 1440 phage antibodies. The unique mAbs were further screened on nine basal and seven luminal cell lines to identify those with the greatest affinity, specificity, and internalizing capability for basal-like breast cancer cells. Among the internalizing basal-specific mAbs were those recognizing four transmembrane receptors (EphA2, CD44, CD73 and EGFR), identified by immunoprecipitation-mass spectrometry and yeast-displayed antigen screening. Basal-like breast cancer expression of these four receptors was confirmed using a bioinformatic approach, and expression microarray data on 683 intrinsically subtyped primary breast tumors. This overall approach, which sequentially employs phage display antibody library selection, antigen identification and bioinformatic confirmation of antigen expression by cancer subtypes, offers efficient production of high-affinity mAbs with diagnostic and therapeutic utility against specific cancer subtypes.


Antibodies, Neoplasm , Antibody Affinity , Antibody Specificity , Breast Neoplasms/immunology , Single-Chain Antibodies , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
9.
Protein Eng Des Sel ; 30(9): 673-684, 2017 09 01.
Article En | MEDLINE | ID: mdl-28981915

Bispecific antibodies that redirect the lytic activity of cytotoxic immune effector cells, such as T- and NK cells, onto tumor cells have emerged as a highly attractive and clinically validated treatment modality for hematological malignancies. Advancement of this therapeutic concept into solid tumor indications, however, is hampered by the scarcity of targetable antigens that are surface-expressed on tumor cells but demonstrate only limited expression on healthy tissues. To overcome this limitation, the concept of dual-targeting, i.e. the simultaneous targeting of two tumor-expressed surface antigens with limited co-expression on non-malignant cells, with multispecific antibodies has been proposed to increase tumor selectivity of antibody-induced effector cell cytotoxicity. Here, a novel CD16A (FcγRIIIa)-directed trispecific, tetravalent antibody format, termed aTriFlex, is described, that is capable of redirecting NK cell cytotoxicity to two surface-expressed antigens. Using a BCMA/CD200-based in vitro model system, the potential use of aTriFlex antibodies for dual-targeting and selective induction of NK cell-mediated target cell lysis was investigated. Bivalent bispecific target cell binding was found to result in significant avidity gains and up to 17-fold increased in vitro potency. These data suggest trispecific aTriFlex antibodies may support dual-targeting strategies to redirect NK cell cytotoxicity with increased selectivity to enable targeting of solid tumor antigens.


Antibodies, Bispecific/biosynthesis , Antibodies, Neoplasm/biosynthesis , Cytotoxicity, Immunologic , Immunotherapy/methods , Killer Cells, Natural/immunology , Receptors, IgG/immunology , Animals , Antibodies, Bispecific/genetics , Antibodies, Neoplasm/genetics , Antibody Affinity , Antigens, CD/genetics , Antigens, CD/immunology , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/immunology , CHO Cells , Coculture Techniques , Cricetulus , Gene Expression , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation , Primary Cell Culture , Protein Binding , Receptors, IgG/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology
10.
Mol Ther ; 25(11): 2440-2451, 2017 11 01.
Article En | MEDLINE | ID: mdl-28974431

In solid tumors, chimeric antigen receptor (CAR)-modified T cells must overcome the challenges of the immunosuppressive tumor microenvironment. We hypothesized that pre-treating tumors with our binary oncolytic adenovirus (CAd), which produces local oncolysis and expresses immunostimulatory molecules, would enhance the antitumor activity of HER2-specific CAR T cells, which alone are insufficient to cure solid tumors. We tested multiple cytokines in conjunction with PD-L1-blocking antibody and found that Ad-derived IL-12p70 prevents the loss of HER2.CAR-expressing T cells at the tumor site. Accordingly, we created a construct encoding the PD-L1-blocking antibody and IL-12p70 (CAd12_PDL1). In head and neck squamous cell carcinoma (HNSCC) xenograft models, combining local treatment with CAd12_PDL1 and systemic HER2.CAR T cell infusion improved survival to >100 days compared with approximately 25 days with either approach alone. This combination also controlled both primary and metastasized tumors in an orthotopic model of HNSCC. Overall, our data show that CAd12_PDL1 augments the anti-tumor effects of HER2.CAR T cells, thus controlling the growth of both primary and metastasized tumors.


Adenoviridae/immunology , Carcinoma, Squamous Cell/therapy , Combined Modality Therapy/methods , Head and Neck Neoplasms/therapy , Immunotherapy, Adoptive/methods , Oncolytic Virotherapy/methods , Adenoviridae/genetics , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Female , Gene Expression , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Interleukin-12/genetics , Interleukin-12/immunology , Lymphocyte Transfusion , Mice , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tumor Burden , Xenograft Model Antitumor Assays
11.
Protein Eng Des Sel ; 30(10): 713-721, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-29040754

As a stress-inducible natural killer (NK) cell ligand, B7H6 plays a role in innate tumor immunosurveillance and is a fairly tumor selective marker expressed on a variety of solid and hematologic cancer cells. Here, we describe the isolation and characterization of a new family of single chain fragment variable (scFv) molecules targeting the human B7H6 ligand. Through directed evolution of a yeast surface displayed non-immune human-derived scFv library, eight candidates comprising a single family of clones differing by up to four amino acid mutations and exhibiting nM avidities for soluble B7H6-Ig were isolated. A representative clone re-formatted as an scFv-CH1-Fc molecule demonstrated specific binding to both B7H6-Ig and native membrane-bound B7H6 on tumor cell lines with a binding avidity comparable to the previously characterized B7H6-targeting antibody, TZ47. Furthermore, these clones recognized an epitope distinct from that of TZ47 and the natural NK cell ligand NKp30, and demonstrated specific activity against B7H6-expressing tumor cells when expressed as a chimeric antigen receptor (CAR) in T cells.


Antibodies, Neoplasm/chemistry , B7 Antigens/chemistry , Biomarkers, Tumor/chemistry , Mutant Chimeric Proteins/chemistry , Receptors, Antigen, T-Cell/chemistry , Single-Chain Antibodies/chemistry , Amino Acid Substitution , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , B7 Antigens/genetics , B7 Antigens/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cell Line, Tumor , Cell Surface Display Techniques , Cytotoxicity, Immunologic , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Gene Expression , HEK293 Cells , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Models, Molecular , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/immunology , Mutation , Natural Cytotoxicity Triggering Receptor 3/chemistry , Natural Cytotoxicity Triggering Receptor 3/genetics , Natural Cytotoxicity Triggering Receptor 3/immunology , Protein Binding , Protein Interaction Domains and Motifs , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics
12.
Cell Rep ; 20(5): 1073-1087, 2017 08 01.
Article En | MEDLINE | ID: mdl-28768193

Recent successes in tumor immunotherapies have highlighted the importance of tumor immunity. However, most of the work conducted to date has been on T cell immunity, while the role of B cell immunity in cancer remains more elusive. In this study, immunogenetic repertoire profiling for tumor-infiltrating B and T cells in gastric cancers was carried out to help reveal the architecture of B cell immunity in cancer. Humoral immunity in cancer was shown to involve oligoclonal expansions of tumor-specific and private B cell repertoires. We find that B cell repertoires in cancer are shaped by somatic hypermutation (SHM) either with or without positive selection biases, the latter of which tended to be auto-reactive. Importantly, we identified sulfated glycosaminoglycans (GAGs) as major functional B cell antigens among gastric tumors. Furthermore, natural anti-sulfated GAG antibodies discovered in gastric cancer tissues showed robust growth-suppressive functions against a wide variety of human malignancies of various organs.


Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Somatic Hypermutation, Immunoglobulin , Stomach Neoplasms/immunology , Antibodies, Neoplasm/genetics , B-Lymphocytes/pathology , Hep G2 Cells , Humans , Stomach Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
13.
PLoS One ; 12(6): e0179239, 2017.
Article En | MEDLINE | ID: mdl-28617827

Immunodeficient mice transplanted with human peripheral blood mononuclear cells (PBMCs) are promising tools to evaluate human immune responses to vaccines. However, these mice usually develop severe graft-versus-host disease (GVHD), which makes estimation of antigen-specific IgG production after antigen immunization difficult. To evaluate antigen-specific IgG responses in PBMC-transplanted immunodeficient mice, we developed a novel NOD/Shi-scid-IL2rγnull (NOG) mouse strain that systemically expresses the human IL-4 gene (NOG-hIL-4-Tg). After human PBMC transplantation, GVHD symptoms were significantly suppressed in NOG-hIL-4-Tg compared to conventional NOG mice. In kinetic analyses of human leukocytes, long-term engraftment of human T cells has been observed in peripheral blood of NOG-hIL-4-Tg, followed by dominant CD4+ T rather than CD8+ T cell proliferation. Furthermore, these CD4+ T cells shifted to type 2 helper (Th2) cells, resulting in long-term suppression of GVHD. Most of the human B cells detected in the transplanted mice had a plasmablast phenotype. Vaccination with HER2 multiple antigen peptide (CH401MAP) or keyhole limpet hemocyanin (KLH) successfully induced antigen-specific IgG production in PBMC-transplanted NOG-hIL-4-Tg. The HLA haplotype of donor PBMCs might not be relevant to the antibody secretion ability after immunization. These results suggest that the human PBMC-transplanted NOG-hIL-4-Tg mouse is an effective tool to evaluate the production of antigen-specific IgG antibodies.


Antibodies, Neoplasm/immunology , Antibody Formation , Carrier Proteins/immunology , Immunization , Immunoglobulin G/immunology , Interleukin-4/immunology , Peptides/pharmacology , Receptor, ErbB-2/pharmacology , Animals , Antibodies, Neoplasm/genetics , B-Lymphocytes/immunology , B-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Carrier Proteins/genetics , Heterografts , Humans , Immunoglobulin G/genetics , Interleukin-4/genetics , Mice , Mice, SCID , Mice, Transgenic , Peptides/immunology , Receptor, ErbB-2/immunology , Th2 Cells/immunology , Th2 Cells/transplantation
14.
Monoclon Antib Immunodiagn Immunother ; 36(3): 104-112, 2017 Jun.
Article En | MEDLINE | ID: mdl-28504613

Podoplanin is expressed in many cancers, including oral cancers and brain tumors. The interaction between podoplanin and its receptor C-type lectin-like receptor 2 (CLEC-2) has been reported to be involved in cancer metastasis and tumor malignancy. We previously established many monoclonal antibodies (mAbs) against human podoplanin using the cancer-specific mAb (CasMab) technology. LpMab-23 (IgG1, kappa), one of the mouse anti-podoplanin mAbs, was shown to be a CasMab. However, we have not shown the usefulness of LpMab-23 for antibody therapy against podoplanin-expressing cancers. In this study, we first determined the minimum epitope of LpMab-23 and revealed that Gly54-Leu64 peptide, especially Gly54, Thr55, Ser56, Glu57, Asp58, Arg59, Tyr60, and Leu64 of podoplanin, is a critical epitope of LpMab-23. We further produced human-mouse chimeric LpMab-23 (chLpMab-23) and investigated whether chLpMab-23 exerts antibody-dependent cellular cytotoxicity (ADCC) and antitumor activity. In flow cytometry, chLpMab-23 showed high sensitivity against a podoplanin-expressing glioblastoma cell line, LN319, and an oral cancer cell line, HSC-2. chLpMab-23 also showed ADCC activity against podoplanin-expressing CHO cells (CHO/podoplanin). In xenograft models with HSC-2 and CHO/podoplanin, chLpMab-23 exerts antitumor activity using human natural killer cells, indicating that chLpMab-23 could be useful for antibody therapy against podoplanin-expressing cancers.


Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Membrane Glycoproteins/antagonists & inhibitors , Mouth Neoplasms/drug therapy , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CHO Cells , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cricetulus , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Gene Expression , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mouth Neoplasms/genetics , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Protein Eng Des Sel ; 30(4): 291-301, 2017 04 01.
Article En | MEDLINE | ID: mdl-28062646

Bispecific antibodies (bsAbs) pave the way for novel therapeutic modes of action along with potential benefits in several clinical applications. However, their generation remains challenging due to the necessity of correct pairings of two different heavy and light chains and related manufacturability issues. We describe a generic approach for the generation of fully human IgG-like bsAbs. For this, heavy chain repertoires from immunized transgenic rats were combined with either a randomly chosen common light chain or a light chain of an existing therapeutic antibody and screened for binders against tumor-related targets CEACAM5 and CEACAM6 by yeast surface display. bsAbs with subnanomolar affinities were identified, wherein each separate binding arm mediated specific binding to the respective antigen. Altogether, the described strategy represents a combination of in vivo immunization with an in vitro selection method, which allows for the integration of existing therapeutic antibodies into a bispecific format.


Antibodies, Bispecific , Antibodies, Neoplasm , Single-Chain Antibodies , Animals , Antibodies, Bispecific/biosynthesis , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/genetics , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antigens, CD , Carcinoembryonic Antigen , Cell Adhesion Molecules/antagonists & inhibitors , GPI-Linked Proteins/antagonists & inhibitors , Humans , Mice , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics
16.
Mol Immunol ; 72: 49-56, 2016 Apr.
Article En | MEDLINE | ID: mdl-26943931

TH2-biased immunity to parasites and allergens is often associated with increased levels of antigen-specific and high affinity IgE. The role in reacting against minute amounts of target structures and to provoke severe anaphylactic reactions renders IgE a mechanistically outstanding isotype. IgE represents the least abundant serum antibody isotype and exhibits a variety of peculiarities including structure, extensive glycosylation and effector functions. Despite large progress in antibody technologies, however, the recombinant access to isotypes beyond IgG such as IgE still is scarce. The capacity of expression systems has to meet the complex structural conformations and the extensive posttranslational modifications that are indispensable for biological activity. In order to provide alternatives to mammalian expression systems with often low yield and a more complex glycosylation pattern we established the recombinant production of the highly complex IgE isotype in insect cells. Recombinant IgE (rIgE) was efficiently assembled and secreted into the supernatant in yields of >30 mg/L. Purification from serum free medium using different downstream processing methods provided large amounts of rIgE. This exhibited a highly specific interaction with its antigen, therapeutic anti-IgE and its high affinity receptor, the FcεRI. Lectins and glyco-proteomic analyses proved the presence of prototypic insect type N-glycans on the epsilon heavy chain. Mediator release assays demonstrated a biological activity of the rIgE comparable to IgE derived from mammalian cells. In summary the expression in insect cells provides rIgE with variant glycosylation pattern, but retained characteristics and biological activity. Therefore our data contribute to the understanding of functional and structural aspects and potential use of the IgE isotype.


Cloning, Molecular/methods , Immunoglobulin E/biosynthesis , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin E/isolation & purification , Polysaccharides/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sf9 Cells , Spodoptera , Surface Plasmon Resonance
17.
MAbs ; 8(2): 414-25, 2016.
Article En | MEDLINE | ID: mdl-26785809

L1 cell adhesion molecule (L1CAM) is aberrantly expressed in malignant tumors and plays important roles in tumor progression. Thus, L1CAM could serve as a therapeutic target and anti-L1CAM antibodies may have potential as anticancer agents. However, L1CAM is expressed in neural cells and the druggability of anti-L1AM antibody must be validated at the earliest stages of preclinical study. Here, we generated a human monoclonal antibody that is cross-reactive with mouse L1CAM and evaluated its pharmacokinetic properties and anti-tumor efficacy in rodent models. First, we selected an antibody (Ab4) that binds human and mouse L1CAM from the human naïve Fab library using phage display, then increased its affinity 45-fold through mutation of 3 residues in the complementarity-determining regions (CDRs) to generate Ab4M. Next, the affinity of Ab4M was increased 1.8-fold by yeast display of single-chain variable fragment containing randomly mutated light chain CDR3 to generate Ab417. The affinities (KD) of Ab417 for human and mouse L1CAM were 0.24 nM and 79.16 pM, respectively. Ab417 specifically bound the Ig5 domain of L1CAM and did not exhibit off-target activity, but bound to the peripheral nerves embedded in normal human tissues as expected in immunohistochemical analysis. In a pharmacokinetics study, the mean half-life of Ab417 was 114.49 h when a single dose (10 mg/kg) was intravenously injected into SD rats. Ab417 significantly inhibited tumor growth in a human cholangiocarcinoma xenograft nude mouse model and did not induce any adverse effect in in vivo studies. Thus, Ab417 may have potential as an anticancer agent.


Antibodies, Neoplasm , Antibody Specificity/immunology , Neoplasms, Experimental/drug therapy , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Single-Chain Antibodies , Animals , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , CHO Cells , Cricetinae , Cricetulus , Cross Reactions/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neural Cell Adhesion Molecule L1/immunology , PC12 Cells , Rats , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , Xenograft Model Antitumor Assays
18.
Clin Cancer Res ; 22(5): 1055-8, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26463707

Immunotoxins are targeted anticancer therapeutics that kill cancer cells using a cytotoxic bacterial toxin payload. Their development for use in solid tumor malignancies was delayed due to issues with their immunogenicity and limited therapeutic window. However, new research has rejuvenated the field. Coadministration with a lymphocyte-depleting regimen of pentostatin and cyclophosphamide can delay antidrug antibody formation, increasing the number of treatment cycles that patients can receive and resulting in durable responses in heavily pretreated patients. In addition, a new generation of immunotoxin molecules with reduced immunogenicity and nonspecific toxicity has been developed through protein engineering techniques, and one has recently entered the clinic. In preclinical studies in mouse models, these new agents are effective against many tumor types as single agents, and also produce synergistic antitumor responses in combination with chemotherapy. These new immunotoxins have renewed excitement in the field and may prove a promising addition to the targeted therapy repertoire.


Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Immunotherapy , Mesothelioma/drug therapy , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , ADP Ribose Transferases/therapeutic use , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , Cyclophosphamide/therapeutic use , Disease Models, Animal , Exotoxins/genetics , Exotoxins/immunology , Exotoxins/therapeutic use , Humans , Lymphocytes/drug effects , Mesothelioma/genetics , Mesothelioma/pathology , Mice , Pentostatin/therapeutic use , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/therapeutic use , Pseudomonas aeruginosa Exotoxin A
19.
J Immunol ; 195(10): 5077-87, 2015 Nov 15.
Article En | MEDLINE | ID: mdl-26475927

Complement-dependent cytotoxicity (CDC) has been suggested to be an important mechanism of action of tumor-targeting Abs. However, single unmodified epidermal growth factor receptor (EGFR)-targeting IgG1 Abs fail to trigger efficient CDC. For the current study, we generated a CDC-optimized variant of the EGFR Ab matuzumab (H425 wt) by introducing amino acid substitutions K326A/E333A (H425 mt). This Ab was then used to elucidate the impact of complement activation on the capacity of effector cells such as mononuclear cells (MNC) and polymorphonuclear cells (PMN) to exert Ab-dependent cell-mediated cytotoxicity (ADCC). H425 mt, but not H425 wt, significantly induced complement deposition, release of anaphylatoxins, and CDC against distinct tumor cell lines, whereas no differences in ADCC by MNC or PMN were detected. Notably, stronger cytotoxicity was induced by H425 mt than by H425 wt in whole blood assays and in experiments in which MNC or PMN were combined with serum. Although MNC-ADCC was not affected by C5 cleavage, the cytotoxic activity of PMN in the presence of serum strongly depended on C5 cleavage, pointing to a direct interaction between complement and PMN. Strong cell surface expression of C5a receptors was detected on PMN, whereas NK cells completely lacked expression. Stimulation of PMN with C5a led to upregulation of activated complement receptor 3, resulting in enhanced complement receptor 3-dependent PMN-ADCC against tumor cells. In conclusion, complement-optimized EGFR Abs may constitute a promising strategy to improve tumor cell killing by enhancing the interaction between humoral and cellular effector functions in Ab-based tumor therapy.


Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neoplasm/pharmacology , Complement C5a/immunology , ErbB Receptors/antagonists & inhibitors , Leukocytes/immunology , Neoplasms/drug therapy , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/immunology , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptor, Anaphylatoxin C5a/immunology
20.
J Control Release ; 216: 37-46, 2015 Oct 28.
Article En | MEDLINE | ID: mdl-26151293

Dendritic cells (DC) are attractive targets for cancer immunotherapy as they initiate strong and long-lived tumour-specific T cell responses. DC can be effectively targeted in vivo with tumour antigens by using nanocarriers such as liposomes. Cross-presentation of tumour antigens is enhanced with strong adjuvants such as TLR ligands. However, often these adjuvants have off-target effects, and would benefit from a DC-specific targeting strategy, similar to the tumour antigen. The goal of this study was to develop a strategy for specifically targeting DC with tumour antigen and adjuvant by using glycoliposomes. We have generated liposomes containing the glycan Lewis(Le)(X) which is highly specific for the C-type lectin receptor DC-SIGN expressed by DC. Le(X)-modified liposomes were taken up by human monocyte-derived DC in a DC-SIGN-specific manner. As adjuvants we incorporated the TLR ligands Pam3CySK4, Poly I:C, MPLA and R848 into liposomes and compared their adjuvant capacity on DC. Incorporation of the TLR4 ligand MPLA into glycoliposomes induced DC maturation and production of pro-inflammatory cytokines, in a DC-SIGN-specific manner, and DC activation was comparable to administration of soluble MPLA. Incorporation of MPLA into glycoliposomes significantly enhanced antigen cross-presentation of the melanoma tumour antigen gp100280-288 peptide to CD8(+) T cells compared to non-glycosylated MPLA liposomes. Importantly, antigen cross-presentation of the gp100280-288 peptide was significantly higher using MPLA glycoliposomes compared to the co-administration of soluble MPLA with glycoliposomes. Taken together, our data demonstrates that specific targeting of a gp100 tumour antigen and the adjuvant MPLA to DC-SIGN-expressing DC enhances the uptake of peptide-containing liposomes, the activation of DC, and induces tumour antigen-specific CD8(+) T cell responses. These data demonstrate that adjuvant-containing glycoliposome-based vaccines targeting DC-SIGN(+) DC represent a powerful new approach for CD8(+) T cell activation.


Dendritic Cells/drug effects , Liposomes/chemistry , Nucleic Acid Amplification Techniques/methods , T-Lymphocytes, Cytotoxic/drug effects , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/genetics , Antigen Presentation/drug effects , Antigens, Neoplasm/chemistry , CD8-Positive T-Lymphocytes/drug effects , Cytokines/biosynthesis , Drug Delivery Systems , Humans , Macrophages/drug effects , Melanoma, Experimental/genetics , Toll-Like Receptor 4/drug effects , gp100 Melanoma Antigen/drug effects
...