Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 823
Filter
1.
Prostate ; 80(11): 811-823, 2020 08.
Article in English | MEDLINE | ID: mdl-32427375

ABSTRACT

BACKGROUND: Exosomes secreted by tumor cells can be regarded as carriers of tumor-associated antigens and have potential value in tumor immunotherapy. The aim of this study was to evaluate the antitumor efficacy of a novel exosomal vaccine (interferon-γ [IFN-γ]-modified exosomal vaccine) in prostate cancer. METHODS: Prostate cancer cell-derived exosomes were used to prepare the exosomal vaccine using our protein-anchoring technique. The immunogenicity and therapeutic efficacy of the exosomes was evaluated by measuring the effects of the exosomal vaccine on M1 macrophage differentiation, the ability of macrophages to engulf the exosomes, the production of antibodies against exosomes, and tumor angiogenesis and metastasis, and tumor growth. RESULTS: The IFN-γ fusion protein was efficiently anchored on the surface of prostate cancer cell-derived exosomes and retained its bioactivity. The IFN-γ-exosomal vaccine increased the number of M1 macrophages, enhanced the ability of M1 macrophages to engulf RM-1 cell-derived exosomes, and induced the production of specific antibodies against exosomes. The exosomal vaccine downregulated the expression of vascular endothelial growth factor receptor 2 and attenuated the effect of exosomes in promoting tumor metastasis. The proportions of CD4+ , CD8+ , and IFN-γ+ CD8+ T cells in the exosomal vaccine group were the highest among the four groups. Interestingly, the IFN-γ-exosomal vaccine decreased the percentage of Tregs and downregulated the expression of programed death-ligand 1 and indoleamine 2, 3-dioxygenase 1 in the tumor environment. The exosomal vaccine significantly inhibited tumor growth and prolonged the survival time of mice with prostate cancer. The exosomal and tumor cell vaccines had a good synergistic effect in promoting tumor immunity. CONCLUSIONS: The novel exosomal vaccine induced an immune response that cleared prostate cancer cell-derived exosomes, thereby eliminating the regulatory effect of the exosomes. This study may provide experimental evidence for the use of exosomes as a therapeutic tool or target in immunotherapy for human prostate cancer.


Subject(s)
Cancer Vaccines/pharmacology , Exosomes/immunology , Interferon-gamma/pharmacology , Prostatic Neoplasms/therapy , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/immunology , Cancer Vaccines/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Line, Tumor , Cytokines/blood , Cytokines/immunology , Immunotherapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/immunology , Random Allocation , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
2.
Protein Eng Des Sel ; 30(9): 673-684, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28981915

ABSTRACT

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.


Subject(s)
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
3.
Protein Eng Des Sel ; 30(10): 713-721, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-29040754

ABSTRACT

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.


Subject(s)
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
4.
PLoS One ; 12(7): e0180786, 2017.
Article in English | MEDLINE | ID: mdl-28686661

ABSTRACT

In a previous study, we developed an E1 monobody specific for the tumor biomarker hEphA2 [PLoS ONE (2015) 10(7): e0132976]. E1 showed potential as a molecular probe for in vitro and in vivo targeting of cancers overexpressing hEphA2. In the present study, we constructed expression vectors for E1 conjugated to optical reporters such as Renilla luciferase variant 8 (Rluc8) or enhanced green fluorescent protein (EGFP) and purified such recombinant proteins by affinity chromatography in E. coli. E1-Rluc8 and E1-EGFP specifically bound to hEphA2 in human prostate cancer PC3 cells but not in human cervical cancer HeLa cells, which express hEphA2 at high and low levels, respectively. These recombinant proteins maintained >40% activity in mouse serum at 24 h. In vivo optical imaging for 24 h did not detect E1-EGFP signals, whereas E1-Rluc8 showed tumor-specific luminescence signals in PC3 but not in HeLa xenograft mice. E1-Rluc8 signals were detected at 4 h, peaked at 12 h, and were undetectable at 24 h. These results suggest the potential of E1-Rluc8 as an EphA2-specific optical imaging agent.


Subject(s)
Antibodies, Neoplasm/chemistry , Biomarkers, Tumor/analysis , Immunoconjugates/chemistry , Receptor, EphA2/analysis , Recombinant Fusion Proteins/genetics , Animals , Antibodies, Neoplasm/biosynthesis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Heterografts , Humans , Immunoconjugates/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Optical Imaging , Organ Specificity , Protein Engineering , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Recombinant Fusion Proteins/metabolism
5.
Protein Eng Des Sel ; 30(4): 291-301, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28062646

ABSTRACT

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.


Subject(s)
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
6.
Hum Vaccin Immunother ; 13(5): 1112-1114, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28118089

ABSTRACT

In light of the increasing number of approved monoclonal antibodies for the treatment of cancer, it seems peculiar that the development of antibody inducing vaccines gets so little attention. In our view there is a tremendous opportunity in the development of cancer vaccines inducing humoral immune responses, involving a couple of major advantages. Firstly, the effectivity of a polyclonal antibody response is expected to exceed the one of monoclonal antibodies. This is supported by preclinical data that show pronounced anti-tumor responses and early clinical trials in which benefit is observed in patients with advanced cancer. Secondly, vaccination strategies are expected to reduce hospital visits, resulting in enhanced quality of life. And last but not least, vaccination strategies are extremely cost effective, alleviating the socioeconomic problems of prohibitively high drug costs. To reach further clinical success, efforts should focus on target identification, optimization of vaccination strategies and adjuvant development.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immunity, Humoral , Neoplasms/immunology , Neoplasms/therapy , Adjuvants, Immunologic , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Humans , Mice , Quality of Life , Rats , Vaccination/economics
7.
J Proteome Res ; 16(1): 204-216, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27700103

ABSTRACT

There is a demand for novel targets and approaches to diagnose and treat prostate cancer (PCA). In this context, serum and plasma samples from a total of 609 individuals from two independent patient cohorts were screened for IgG reactivity against a sum of 3833 human protein fragments. Starting from planar protein arrays with 3786 protein fragments to screen 80 patients with and without PCA diagnosis, 161 fragments (4%) were chosen for further analysis based on their reactivity profiles. Adding 71 antigens from literature, the selection of antigens was corroborated for their reactivity in a set of 550 samples using suspension bead arrays. The antigens prostein (SLC45A3), TATA-box binding protein (TBP), and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) showed higher reactivity in PCA patients with late disease compared with early disease. Because of its prostate tissue specificity, we focused on prostein and continued with mapping epitopes of the 66-mer protein fragment using patient samples. Using bead-based assays and 15-mer peptides, a minimal peptide epitope was identified and refined by alanine scanning to the KPxAPFP. Further sequence alignment of this motif revealed homology to transmembrane protein 79 (TMEM79) and TGF-beta-induced factor 2 (TGIF2), thus providing a reasoning for cross-reactivity found in females. A comprehensive workflow to discover and validate IgG reactivity against prostein and homologous targets in human serum and plasma was applied. This study provides useful information when searching for novel biomarkers or drug targets that are guided by the reactivity of the immune system against autoantigens.


Subject(s)
Biomarkers, Tumor/genetics , Epitopes/chemistry , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor II/genetics , Membrane Proteins/genetics , Prostatic Neoplasms/genetics , TATA-Box Binding Protein/genetics , Aged , Amino Acid Motifs , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/chemistry , Autoimmunity , Biomarkers, Tumor/immunology , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Case-Control Studies , Cross Reactions , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/chemistry , Insulin-Like Growth Factor II/immunology , Male , Membrane Proteins/immunology , Middle Aged , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Protein Array Analysis , Repressor Proteins/genetics , Repressor Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , TATA-Box Binding Protein/immunology
8.
Cancer Immunol Res ; 4(10): 858-868, 2016 10.
Article in English | MEDLINE | ID: mdl-27549123

ABSTRACT

Immune recognition of tumor targets by specific cytotoxic lymphocytes is essential for the effective rejection of tumors. A phase I clinical trial of ipilimumab (an antibody that blocks CTLA-4 function) in combination with bevacizumab (an antibody that inhibits angiogenesis) in patients with metastatic melanoma found favorable clinical outcomes were associated with increased tumor endothelial activation and lymphocyte infiltration. To better understand the underlying mechanisms, we sought features and factors that changed as a function of treatment in patients. Ipilimumab plus bevacizumab (Ipi-Bev) increased tumor vascular expression of ICAM1 and VCAM1. Treatment also altered concentrations of many circulating cytokines and chemokines, including increases of CXCL10, IL1α, TNFα, CXCL1, IFNα2, and IL8, with decreases in VEGF-A in most patients. IL1α and TNFα induced expression of E-selectin, CXCL1, and VCAM1 on melanoma tumor-associated endothelial cells (TEC) in vitro and promoted adhesion of activated T cells onto TEC. VEGFA inhibited TNFα-induced expression of ICAM1 and VCAM1 and T-cell adhesion, which was blocked by bevacizumab. CXCL10 promoted T-cell migration across TEC in vitro, was frequently expressed by melanoma cells, and was upregulated in a subset of tumors in treated patients. Robust upregulation of CXCL10 in tumors was accompanied by increased T-cell infiltration. Ipi-Bev also augmented humoral immune responses recognizing targets in melanoma, tumor endothelial, and tumor mesenchymal stem cells. Our findings suggest that Ipi-Bev therapy augments immune recognition in the tumor microenvironment through enhancing lymphocyte infiltration and antibody responses. IL1α, TNFα, and CXCL10, together with VEGF neutralization, contribute to Ipi-Bev-induced melanoma immune recognition. Cancer Immunol Res; 4(10); 858-68. ©2016 AACR.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma/drug therapy , Adult , Aged , Aged, 80 and over , Bevacizumab/administration & dosage , CTLA-4 Antigen/antagonists & inhibitors , Cell Adhesion/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokines/blood , Cytokines/blood , Female , Humans , Intercellular Adhesion Molecule-1/metabolism , Ipilimumab/administration & dosage , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/metabolism
9.
Mol Immunol ; 72: 49-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26943931

ABSTRACT

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.


Subject(s)
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
10.
Small ; 12(17): 2321-33, 2016 05.
Article in English | MEDLINE | ID: mdl-26953489

ABSTRACT

Targeted subunit vaccines for cancer immunotherapy do not capture tumor antigenic complexity, and approaches employing tumor lysate are often limited by inefficient antigen uptake and presentation, and low immunogenicity. Here, whole cancer cells are processed to generate antigen-rich, membrane-enclosed subcellular particles, termed "reduced cancer cells", that reflect the diversity and breadth of the parent cancer cell antigen repertoire, and can be loaded with disparate adjuvant payloads. These vesicular particles enhance the uptake of the adjuvant payload, and potentiate the activation of primary dendritic cells in vitro. Similarly, reduced cancer cell-associated antigens are more efficiently presented by primary dendritic cells in vitro than their soluble counterparts or lysate control. In mice, vaccination using adjuvant-loaded reduced cancer cells facilitates the induction of antigen-specific cellular and humoral immune responses. Taken together, these observations demonstrate that adjuvant-loaded reduced cancer cells could be utilized in cancer vaccines as an alternative to lysate.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Drug Carriers , Animals , Antibodies, Neoplasm/biosynthesis , Cancer Vaccines/immunology , Cell Line, Tumor , Immunity, Cellular , Mice
11.
Bioorg Med Chem ; 24(4): 915-20, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26787275

ABSTRACT

Tumor-associated carbohydrate antigens (TACAs), which are aberrantly expressed on the surface of tumor cells, are important targets for anticancer vaccine development. Herein, several N-acyl modified Tn analogues were synthesized and conjugated with carrier protein CRM197. The immunological results of these glycoconjugates indicated that 6-CRM197 elicited higher titers of antibodies which cross-reacted with native Tn antigen than the unmodified 2-CRM197 did. The IFN-γ-producing frequency of lymphocytes in mice treated with 6-CRM197 was obviously increased, compared to that of mice vaccinated with 2-CRM197 (p=0.016), which was typically associated with the Th1 response. Moreover, the elicited antisera against antigen 6-CRM197 reacted strongly with the Tn-positive tumor cells, implying the potential of this glycoconjugate as an anticancer vaccine.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Antigens, Tumor-Associated, Carbohydrate/immunology , Bacterial Proteins/immunology , Cancer Vaccines/immunology , Glycoconjugates/immunology , Interferon-gamma/biosynthesis , Acylation , Animals , Antigens, Tumor-Associated, Carbohydrate/chemistry , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/chemistry , Diphtheria Toxin/immunology , Female , Glycoconjugates/administration & dosage , Glycoconjugates/chemical synthesis , Humans , Immune Sera/chemistry , Immunity, Humoral/drug effects , Immunoglobulin G/biosynthesis , Jurkat Cells , MCF-7 Cells , Mice , Mice, Inbred BALB C , Th1 Cells/drug effects , Th1 Cells/immunology
12.
Hum Antibodies ; 24(3-4): 45-51, 2016.
Article in English | MEDLINE | ID: mdl-28128764

ABSTRACT

Objectives were to: 1) induce a lytic IgG antibody (ab) response (via the so called `third vaccination method') against CD38 antigen (ag) residing on the extra-cellular domain of multiple myeloma (MM) cells in recipient rabbits, by combining the CD38 ag with donor-derived anti-CD38 ag lytic IgG ab into an immune complex (IC); and 2) determine whether abs produced would cause complement-mediated lysis (in vitro) of human MM cells containing CD38 ag. The vaccine was created in a two-step process. First, ab (rabbit anti-CD38 ag IgG ab) was raised in donor rabbits by injections of low molecular weight soluble CD38 ag in Freund's complete adjuvant (FCA) and aqueous solution. Second, transfer of pathogenic lytic IgG ab response into recipient rabbits was achieved by injections of ICs composed of CD38 ag and homologous anti-CD38 ag IgG ab. Consequently, recipient rabbits produced the same ab with the same specificity against the target ag as was present in the inoculum, namely agglutinating, precipitating and lytic (as demonstrated in vitro). In an in vitro study, in the presence of complement, donor and recipient rabbits' immune sera caused lysis of CD38 ag associated human MM cells. The most effective lytic ab response causing sera were those from donor rabbits injected with CD38 ag in FCA and those from rabbits injected with ICs, especially when they were administered in adjuvants. These results provided proof of concept that the third vaccination method has good potential as a stand-alone and efficacious method of controlling cancer.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antigen-Antibody Complex/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Multiple Myeloma/therapy , Vaccination/methods , ADP-ribosyl Cyclase 1/administration & dosage , ADP-ribosyl Cyclase 1/genetics , Agglutination Tests , Animals , Antibodies, Neoplasm/biosynthesis , Antigen-Antibody Complex/genetics , Antigen-Antibody Complex/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cell Line, Tumor , Complement System Proteins/pharmacology , Cytotoxicity, Immunologic , Female , Freund's Adjuvant/administration & dosage , Gene Expression , Humans , Immune Sera/pharmacology , Immunoglobulin G/biosynthesis , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Rabbits
13.
J Control Release ; 216: 37-46, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26151293

ABSTRACT

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.


Subject(s)
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
15.
J Immunol ; 195(1): 166-73, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019274

ABSTRACT

The generation of CTLs is crucial in the immunological fight against cancer and many infectious diseases. To achieve this, vaccine Ags need to be targeted to the cytosol of dendritic cells, which can activate CD8 T cells via MHC class I (MHCI). Therefore, such targeting has become one of the major objectives of vaccine research. In this study, we aimed to bypass the unwanted and default MHC class II Ag presentation and trigger MHCI presentation by using a photosensitizer that, upon light activation, would facilitate cytosolic targeting of codelivered Ag. Poly(lactide-co-glycolide) microparticles ∼1 µm size were loaded with OVA and the photosensitizer tetraphenyl chlorine disulphonate (TPCS2a) and administered intradermally in mice, which were illuminated 1 d later for activation of the photosensitizer. Immunization in the presence of TPCS2a significantly increased activation of CD8 T cells compared with immunization without TPCS2a and as measured by CD8 T cell proliferation, production of proinflammatory IFN-γ, TNF-α, and IL-2, and prevention of tumor growth. Cytotoxicity was demonstrated by granzyme B production in vitro and by in vivo killing of CFSE-labeled targets. CD4-dependent Ab responses were abrogated in mice immunized with TPCS2a-containing particles, suggesting that photosensitization facilitated a shift from default MHC class II toward MHCI Ag presentation. Hence, vaccine particles with Ag and photosensitizers proved an effective vehicle or adjuvant for stimulation of CTLs, and they may find potential application in therapeutic cancer vaccination and in prophylactic and therapeutic vaccination against intracellular infections.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Dendritic Cells/drug effects , Melanoma, Experimental/prevention & control , Porphyrins/administration & dosage , Skin Neoplasms/prevention & control , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/administration & dosage , Cytosol/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Drug Carriers/chemistry , Female , Granzymes/biosynthesis , Immunization , Injections, Intradermal , Interleukin-2/biosynthesis , Lactic Acid/chemistry , Light , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neoplasm Transplantation , Ovalbumin/administration & dosage , Ovalbumin/immunology , Photosensitizing Agents/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Biotechnol ; 208: 13-21, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26015261

ABSTRACT

Monoclonal antibody (mAb) therapy has been successfully used for the treatment of B-cell lymphomas and is currently extended for the treatment of multiple myeloma (MM). New developments in MM therapeutics have achieved significant survival gains in patients but the disease still remains incurable. Elotuzumab (HuLuc63), an anti-CS1 monoclonal IgG1 antibody, is believed to induce anti-tumor activity and MM cytotoxicity through antibody dependent cellular cytotoxicity (ADCC) and inhibition of MM cell adhesion to bone marrow stromal cells (BMSCs). Modulations of the Fc glycan composition at the N297 site by selective mutations or afucosylation have been explored as strategies to develop bio-better therapeutics with enhanced ADCC activity. Afucosylated therapeutic antibodies with enhanced ADCC activity have been reported to possess greater efficacy in tumor growth inhibition at lower doses when compared to fucosylated therapeutic antibodies. The N-linked glycosylation pathway in Pichia pastoris has been engineered to produce human-like N-linked glycosylation with uniform afucosylated complex type glycans. The purpose of this study was to compare afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris with fucosylated anti-CS1 mAb expressed in mammalian HEK293 cells through in vitro ADCC and in vivo tumor inhibition models. Our results indicate that Fc glycosylation is critical for in vivo efficacy and afucosylated anti-CS1 mAb expressed in glycoengineered Pichia pastoris shows a better in vivo efficacy in tumor regression when compared to fucosylated anti-CS1 mAb expressed in HEK293 cells. Glycoengineered Pichia pastoris could provide an alternative platform for generating homogeneous afucosylated recombinant antibodies where Fc mediated immune effector function is important for efficacy.


Subject(s)
Antibodies, Monoclonal , Antibodies, Neoplasm , Cell Engineering , Multiple Myeloma/drug therapy , Neoplasms, Experimental/drug therapy , Pichia , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/chemistry , Antibodies, Neoplasm/genetics , Cell Line, Tumor , Glycosylation , HEK293 Cells , Humans , Mice, SCID , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Xenograft Model Antitumor Assays
17.
Biomed Pharmacother ; 70: 53-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25776479

ABSTRACT

Modified melanoma B16 cells inhibited in their IGF-1 expression (B16MOD), on the contrary to the IGF-1 fully expressed parental wild-type (B16WT) counterpart, were shown to stimulate humoral as well as cellular immune responses. Among humoral components, the neutralizing and complement-fixing antibodies of IgM and essentially IgG2 (a+b) isotypes exhibited in vitro and in vivo effects upon tumour growth, while the IgG1 antibody isotype promoted enhanced tumour proliferation. As for the cellular immunity, it was found that the T CD8(+) lymphocyte subpopulation remained the main potent and long lasting immune active effector regulating tumour growth.


Subject(s)
Antibodies, Neoplasm/biosynthesis , Immunity, Cellular/physiology , Immunity, Humoral/physiology , Insulin-Like Growth Factor I/metabolism , Melanoma/immunology , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor I/genetics , Melanoma/metabolism , Mice
18.
Cancer Immunol Res ; 3(3): 236-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25623164

ABSTRACT

CD40 stimulation on antigen-presenting cells (APC) allows direct activation of CD8(+) cytotoxic T cells, independent of CD4⁺ T-cell help. Agonistic anti-CD40 antibodies have been demonstrated to induce beneficial antitumor T-cell responses in mouse models of cancer and early clinical trials. We report here that anti-CD40 treatment induces programmed death ligand-1 (PD-L1) upregulation on tumor-infiltrating monocytes and macrophages, which was strictly dependent on T cells and IFNγ. PD-L1 expression could be counteracted by coadministration of antibodies blocking the PD-1 (programmed death-1)/PD-L1 axis as shown for T cells from tumor models and human donors. The combined treatment was highly synergistic and induced complete tumor rejection in about 50% of mice bearing MC-38 colon and EMT-6 breast tumors. Mechanistically, this was reflected by a strong increase of IFNγ and granzyme-B production in intratumoral CD8⁺ T cells. Concomitant CTLA-4 blockade further improved rejection of established tumors in mice. This study uncovers a novel mechanism of acquired resistance upon agonistic CD40 stimulation and proposes that the concomitant blockade of the PD-1/PD-L1 axis is a viable therapeutic strategy to optimize clinical outcomes.


Subject(s)
Antibodies, Neoplasm/biosynthesis , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/agonists , CD8-Positive T-Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Animals , Antigen-Presenting Cells/immunology , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Granzymes/immunology , Humans , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology
19.
Methods Mol Biol ; 1256: 111-21, 2015.
Article in English | MEDLINE | ID: mdl-25626535

ABSTRACT

Ovarian cancer is a leading cause of death from gynecologic cancers in the USA, and early diagnosis can potentially increase 5-year survival rate. Detection of biomarkers derived from hyperplasia of epithelial tissue by enzyme-linked immunosorbent assay (ELISA) proves to be a practical way of early diagnosis of ovarian cancer. However, ELISA is commonly performed in a laboratory setting, and it cannot be used in a clinical setting for on-site consultation. We have shown a microchip ELISA that detects HE4, an ovarian cancer biomarker, from urine using a cell phone integrated with a mobile application for imaging and data analysis. In microchip ELISA, HE4 from urine was first absorbed on the surface; the primary and secondary antibodies were subsequently anchored on the surface via immuno-reaction; and addition of substrate led to color development because of enzymatic labeling. The microchip after color development was imaged using a cell phone, and the color intensity was analyzed by an integrated mobile application. By comparing with an ELISA standard curve, the concentration of HE4 was reported on the cell phone screen. The presented microchip ELISA coupled with a cell phone is portable as opposed to traditional ELISA, and this method can facilitate the detection of ovarian cancer at the point-of-care (POC).


Subject(s)
Biomarkers, Tumor/urine , Enzyme-Linked Immunosorbent Assay/methods , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/urine , Protein Array Analysis/instrumentation , Proteins/analysis , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/chemistry , Biomarkers, Tumor/genetics , Cell Phone , Colorimetry/methods , Early Diagnosis , Enzyme-Linked Immunosorbent Assay/instrumentation , Female , Gene Expression , Horseradish Peroxidase/chemistry , Humans , Immobilized Proteins/analysis , Immobilized Proteins/genetics , Limit of Detection , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteins/genetics , Rabbits , Telemedicine , WAP Four-Disulfide Core Domain Protein 2
SELECTION OF CITATIONS
SEARCH DETAIL
...