Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Nat Med ; 28(9): 1872-1882, 2022 09.
Article in English | MEDLINE | ID: mdl-36038629

ABSTRACT

Axicabtagene ciloleucel (axi-cel) is an anti-CD19 chimeric antigen receptor (CAR) T cell therapy approved for relapsed/refractory large B cell lymphoma (LBCL) and has treatment with similar efficacy across conventional LBCL subtypes. Toward patient stratification, we assessed whether tumor immune contexture influenced clinical outcomes after axi-cel. We evaluated the tumor microenvironment (TME) of 135 pre-treatment and post-treatment tumor biopsies taken from 51 patients in the ZUMA-1 phase 2 trial. We uncovered dynamic patterns that occurred within 2 weeks after axi-cel. The biological associations among Immunoscore (quantification of tumor-infiltrating T cell density), Immunosign 21 (expression of pre-defined immune gene panel) and cell subsets were validated in three independent LBCL datasets. In the ZUMA-1 trial samples, clinical response and overall survival were associated with pre-treatment immune contexture as characterized by Immunoscore and Immunosign 21. Circulating CAR T cell levels were associated with post-treatment TME T cell exhaustion. TME enriched for chemokines (CCL5 and CCL22), γ-chain receptor cytokines (IL-15, IL-7 and IL-21) and interferon-regulated molecules were associated with T cell infiltration and markers of activity. Finally, high density of regulatory T cells in pre-treatment TME associated with reduced axi-cel-related neurologic toxicity. These findings advance the understanding of LBCL TME characteristics associated with clinical responses to anti-CD19 CAR T cell therapy and could foster biomarker development and treatment optimization for patients with LBCL.


Subject(s)
Biological Products , Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Antigens, CD19 , Cell Count , Humans , Immunotherapy, Adoptive/adverse effects , Interferons/therapeutic use , Interleukin-15 , Interleukin-7/therapeutic use , Lymphoma, Large B-Cell, Diffuse/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use , Tumor Microenvironment
3.
Cancer Discov ; 11(3): 599-613, 2021 03.
Article in English | MEDLINE | ID: mdl-33334730

ABSTRACT

T cell-based therapies have induced cancer remissions, though most tumors ultimately progress, reflecting inherent or acquired resistance including antigen escape. Better understanding of how T cells eliminate tumors will help decipher resistance mechanisms. We used a CRISPR/Cas9 screen and identified a necessary role for Fas-FasL in antigen-specific T-cell killing. We also found that Fas-FasL mediated off-target "bystander" killing of antigen-negative tumor cells. This localized bystander cytotoxicity enhanced clearance of antigen-heterogeneous tumors in vivo, a finding that has not been shown previously. Fas-mediated on-target and bystander killing was reproduced in chimeric antigen receptor (CAR-T) and bispecific antibody T-cell models and was augmented by inhibiting regulators of Fas signaling. Tumoral FAS expression alone predicted survival of CAR-T-treated patients in a large clinical trial (NCT02348216). These data suggest strategies to prevent immune escape by targeting both the antigen expression of most tumor cells and the geography of antigen-loss variants. SIGNIFICANCE: This study demonstrates the first report of in vivo Fas-dependent bystander killing of antigen-negative tumors by T cells, a phenomenon that may be contributing to the high response rates of antigen-directed immunotherapies despite tumoral heterogeneity. Small molecules that target the Fas pathway may potentiate this mechanism to prevent cancer relapse.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Cytotoxicity, Immunologic , Immunotherapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , fas Receptor/metabolism , Animals , Antigens, Neoplasm/immunology , Bystander Effect/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CRISPR-Cas Systems , Disease Models, Animal , Gene Editing , Genetic Engineering , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Mice , Mice, Knockout , Neoplasms/etiology , Neoplasms/therapy , Receptors, Chimeric Antigen , T-Cell Antigen Receptor Specificity , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Nat Med ; 26(2): 270-280, 2020 02.
Article in English | MEDLINE | ID: mdl-31959992

ABSTRACT

Anti-CD19 chimeric antigen receptor (CAR)-expressing T cells are an effective treatment for B-cell lymphoma, but often cause neurologic toxicity. We treated 20 patients with B-cell lymphoma on a phase I, first-in-human clinical trial of T cells expressing the new anti-CD19 CAR Hu19-CD828Z (NCT02659943). The primary objective was to assess safety and feasibility of Hu19-CD828Z T-cell therapy. Secondary objectives included assessments of blood levels of CAR T cells, anti-lymphoma activity, second infusions and immunogenicity. All objectives were met. Fifty-five percent of patients who received Hu19-CD828Z T cells obtained complete remission. Hu19-CD828Z T cells had clinical anti-lymphoma activity similar to that of T cells expressing FMC63-28Z, an anti-CD19 CAR tested previously by our group, which contains murine binding domains and is used in axicabtagene ciloleucel. However, severe neurologic toxicity occurred in only 5% of patients who received Hu19-CD828Z T cells, whereas 50% of patients who received FMC63-28Z T cells experienced this degree of toxicity (P = 0.0017). T cells expressing Hu19-CD828Z released lower levels of cytokines than T cells expressing FMC63-28Z. Lower levels of cytokines were detected in blood from patients who received Hu19-CD828Z T cells than in blood from those who received FMC63-28Z T cells, which could explain the lower level of neurologic toxicity associated with Hu19-CD828Z. Levels of cytokines released by CAR-expressing T cells particularly depended on the hinge and transmembrane domains included in the CAR design.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/immunology , Adolescent , Adult , Aged , Cytokines/metabolism , Feasibility Studies , Female , Humans , K562 Cells , Male , Middle Aged , Phenotype , Protein Domains , Remission Induction , Young Adult
6.
Sci Rep ; 9(1): 5101, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911061

ABSTRACT

Tumor-specific glycosylation changes are an attractive target for the development of diagnostic and therapeutic applications. Periostin is a glycoprotein with high expression in many tumors of epithelial origin including ovarian cancer. Strategies to target the peptide portion of periostin as a diagnostic or therapeutic biomarker for cancer are limited due to increased expression of periostin in non-cancerous inflammatory conditions. Here, we have screened for antibody fragments that recognize the tumor-specific glycosylation present on glycoforms of periostin containing bisecting N-glycans in ovarian cancer using a yeast-display library of antibody fragments, while subtracting those that bind to the periostin protein with glycoforms found in non-malignant cell types. We generated a biotinylated form of a fully human scFv antibody (scFvC9) that targets the bisecting N-glycans expressed by cancer cells. Validation studies in vitro and in vivo using scFvC9 indicate this antibody can be useful for the development of diagnostic, imaging, and therapeutic applications for cancers that express the antigen.


Subject(s)
Single-Chain Antibodies/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Female , Glycosylation , Humans , Immunochemistry , Immunoglobulin Fragments/metabolism , Magnetic Resonance Imaging , Mice , Ovarian Neoplasms/metabolism , Peptide Library , Polysaccharides/metabolism
7.
Methods Mol Biol ; 1827: 211-233, 2018.
Article in English | MEDLINE | ID: mdl-30196500

ABSTRACT

The critical need for renewable, high-quality affinity reagents in biological research, as well as for diagnostic and therapeutic applications, has required the development of new platforms of discovery. Yeast display is one of the main methods of in vitro display technology with phage display. Yeast display has been chosen by numerous groups to refine both affinity and specificity of antibodies because it enables fine discrimination between mutant clones of similar affinity. In addition, the construction of display libraries of antibody fragments in yeast permits to sample the immune antibody repertoire more fully than using phage. This chapter gives an updated overview of the available systems of yeast display platforms and libraries, followed up by technical descriptions of selection methods of antibody fragments by yeast display.


Subject(s)
Cell Surface Display Techniques/methods , Immunoglobulin Fragments/isolation & purification , Saccharomyces cerevisiae/metabolism , Antigens/metabolism , Biotinylation , Flow Cytometry , Peptide Library , Recombinant Proteins/metabolism , Single-Chain Antibodies/metabolism
8.
Mol Pharm ; 15(4): 1403-1411, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29462558

ABSTRACT

Mesothelin is an epithelial marker highly expressed at the cell surface of cancer cells from diverse origins, including ovarian and pancreatic adenocarcinomas and mesotheliomas. Previously, we identified and characterized an antimesothelin nanobody (NbG3a) for in vitro diagnostic applications. The main goal of this research was to establish the potential of NbG3a as a molecular imaging agent. Site-specific biotinylated NbG3a (bNbG3a) was bound to streptavidin-conjugated reagents for in vitro and in vivo assays. Initially, we performed microscale thermophoresis to determine the binding affinity between bNbG3a and human ( Kd = 46 ± 8 nM) or mouse ( Kd = 4.8 ± 0.4 nM) mesothelin protein. The human and mouse cross-reactivity was confirmed by in vivo optical imaging using bNbG3a bound to fluorescent streptavidin. We also localized the binding site of nNbG3a on human mesothelin using overlapping peptide scan. NbG3a recognized an epitope within residues 21-65 of the mature membrane bound form of human mesothelin, which is part of the N-terminal region of mesothelin that is important for interactions between mesothelin on peritoneal cells and CA125 on tumor cells. Next, the bNbG3a in vivo half-life after intravenous injection in healthy mice was estimated by ELISA assay to be 5.3 ± 1.3 min. In tumor-bearing animals, fluorescent bNbG3a accumulated in a subcutaneous ovarian xenograft (A1847) and in two syngeneic, orthotopic ovarian tumors (intraovary and intraperitoneal ID8) within an hour of intravenous injection that peaked by 4 h and persisted up to 48 h. MRI analysis of bNbG3a-targeted streptavidin-labeled iron oxides showed that the MRI signal intensity decreased 1 h after injection for a subcutaneous xenograft model of ovarian cancer for bNbG3a-labeled iron oxides compared to unlabeled iron oxides. The signal intensity differences continued up to the final time point at 24 h post injection. Finally, in vivo immunofluorescence 24 or 48 h after bNbG3a intravenous injection showed bNbG3a diffuse distribution of both xenograft and syngeneic ovarian tumors, with local areas of high concentration throughout A1847 human tumor. The data support the use of NbG3a for continued preclinical development and translation to human applications for cancers that overexpress mesothelin.


Subject(s)
Cross Reactions/immunology , GPI-Linked Proteins/metabolism , Ovarian Neoplasms/pathology , Single-Domain Antibodies/immunology , Animals , CA-125 Antigen/metabolism , Cell Line, Tumor , Female , Ferric Compounds/metabolism , Fluorescent Dyes/metabolism , GPI-Linked Proteins/immunology , Heterografts , Humans , Magnetic Resonance Imaging/methods , Mesothelin , Mice , Mice, Inbred C57BL , Molecular Imaging/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Streptavidin/metabolism
9.
Cancer Immunol Immunother ; 67(2): 329-339, 2018 02.
Article in English | MEDLINE | ID: mdl-29313073

ABSTRACT

Tumor endothelial marker 1 (TEM1) has been identified as a novel surface marker upregulated on the blood vessels and stroma in many solid tumors. We previously isolated a novel single-chain variable fragment (scFv) 78 against TEM1 from a yeast display scFv library. Here we evaluated the potential applications of scFv78 as a tool for tumor molecular imaging, immunotoxin-based therapy and nanotherapy. Epitope mapping, three-dimensional (3D) structure docking and affinity measurements indicated that scFv78 could bind to both human and murine TEM1, with equivalent affinity, at a well-conserved conformational epitope. The rapid internalization of scFv78 and scFv78-labeled nanoparticles was triggered after specific TEM1 binding. The scFv78-saporin immunoconjugate also exerted dose-dependent cytotoxicity with high specificity to TEM1-positive cells in vitro. Finally, specific and sensitive tumor localization of scFv78 was confirmed with optical imaging in a mouse tumor model that has highly endogenous mTEM1 expression in the vasculature. Our data indicate that scFv78, the first fully human anti-TEM1 recombinant antibody, recognizes both human and mouse TEM1 and has unique and favorable features that are advantageous for the development of imaging probes or antibody-toxin conjugates for a large spectrum of human TEM1-positive solid tumors.

10.
Nat Commun ; 8(1): 607, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28928360

ABSTRACT

In melanoma, therapies with inhibitors to oncogenic BRAFV600E are highly effective but responses are often short-lived due to the emergence of drug-resistant tumor subpopulations. We describe here a mechanism of acquired drug resistance through the tumor microenvironment, which is mediated by human tumor-associated B cells. Human melanoma cells constitutively produce the growth factor FGF-2, which activates tumor-infiltrating B cells to produce the growth factor IGF-1. B-cell-derived IGF-1 is critical for resistance of melanomas to BRAF and MEK inhibitors due to emergence of heterogeneous subpopulations and activation of FGFR-3. Consistently, resistance of melanomas to BRAF and/or MEK inhibitors is associated with increased CD20 and IGF-1 transcript levels in tumors and IGF-1 expression in tumor-associated B cells. Furthermore, first clinical data from a pilot trial in therapy-resistant metastatic melanoma patients show anti-tumor activity through B-cell depletion by anti-CD20 antibody. Our findings establish a mechanism of acquired therapy resistance through tumor-associated B cells with important clinical implications.Resistance to BRAFV600E inhibitors often occurs in melanoma patients. Here, the authors describe a potential mechanism of acquired drug resistance mediated by tumor-associated B cells-derived IGF-1.


Subject(s)
Antineoplastic Agents/therapeutic use , B-Lymphocytes/metabolism , Drug Resistance, Neoplasm , Insulin-Like Growth Factor I/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Survival , Cisplatin/therapeutic use , Fibroblast Growth Factor 2/metabolism , Humans , In Vitro Techniques , Melanoma/genetics , Paclitaxel/therapeutic use , Pilot Projects , Proto-Oncogene Proteins B-raf/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Skin Neoplasms/genetics , Tumor Microenvironment
11.
Mol Ther ; 24(11): 1987-1999, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27439899

ABSTRACT

B7-H4 protein is frequently overexpressed in ovarian cancer. Here, we engineered T cells with novel B7-H4-specific chimeric antigen receptors (CARs) that recognized both human and murine B7-H4 to test the hypothesis that B7-H4 CAR T cell therapy can be applied safely in preclinical models. B7-H4 CAR T cells specifically secreted IFN-γ and lysed B7-H4(+) targets. In vivo, B7-H4 CAR T cells displayed antitumor reactivity against B7-H4(+) human ovarian tumor xenografts. Unexpectedly, B7-H4 CAR T cell treatment reproducibly showed delayed, lethal toxicity 6-8 weeks after therapy. Comprehensive assessment of murine B7-H4 protein distribution uncovered expression in ductal and mucosal epithelial cells in normal tissues. Postmortem analysis revealed the presence of widespread histologic lesions that correlated with B7-H4(+) expression, and were inconsistent with graft versus host disease. Lastly, expression patterns of B7-H4 protein in normal human tissue were comparable to distribution in mice, advancing our understanding of B7-H4. We conclude that B7-H4 CAR therapy mediates control of cancer outgrowth. However, long-term engraftment of B7-H4 CAR T cells mediates lethal, off-tumor toxicity that is likely due to wide expression of B7-H4 in healthy mouse organs. This model system provides a unique opportunity for preclinical evaluation of safety approaches that limit CAR-mediated toxicity after tumor destruction in vivo.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Ovarian Neoplasms/therapy , Receptors, Antigen/metabolism , T-Lymphocytes/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Ovarian Neoplasms/immunology , T-Lymphocytes/transplantation , Treatment Outcome , Xenograft Model Antitumor Assays
12.
J Biomed Nanotechnol ; 11(7): 1201-12, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26307843

ABSTRACT

Mesothelin, a cancer biomarker overexpressed in tumors of epithelial origin, is a target for nanotechnology-based diagnostic, therapeutic, and prognostic applications. The currently available anti-mesothelin antibodies present limitations, including low penetration due to large size and/or lack of in vivo stability. Single domain antibodies (sdAbs) or nanobodies (Nbs) provide powerful solutions to these specific problems. We generated a phage-display library of Nbs that were amplified from B cells of a llama that was immunized with human recombinant mesothelin. Two nanobodies (Nb A1 and Nb C6) were selected on the basis of affinity (K(D) = 15 and 30 nM, respectively). Nb A1 was further modified by adding either a cysteine to permit maleimide-based bioconjugations or a sequence for the site-specific metabolic addition of a biotin in vivo. Both systems of conjugation (thiol-maleimide and streptavidin/biotin) were used to characterize and validate Nb A1 and to functionalize nanoparticles. We showed that anti-mesothelin Nb A1 could detect native and denatured mesothelin in various diagnostic applications, including flow cytometry, western blotting, immunofluorescence, and optical imaging. In conclusion, anti-mesothelin Nbs are novel, cost-effective, small, and single domain reagents with high affinity and specificity for the tumor-associated antigen mesothelin, which can be simply bioengineered for attachment to nanoparticles or modified surfaces using multiple bioconjugation strategies. These anti-mesothelin Nbs can be useful in both conventional and nanotechnology-based diagnostic, therapeutic and prognostic biomedical applications.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , GPI-Linked Proteins/immunology , Nanoparticles/therapeutic use , Subcellular Fractions/immunology , Antibodies, Monoclonal/genetics , GPI-Linked Proteins/chemistry , GPI-Linked Proteins/genetics , HeLa Cells , Humans , Mesothelin , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Protein Engineering/methods , Subcellular Fractions/pathology
13.
Methods Mol Biol ; 1319: 37-49, 2015.
Article in English | MEDLINE | ID: mdl-26060068

ABSTRACT

B7-H4 (VTCN1, B7x, B7s) is an inhibitory modulator of T-cell response implicated in antigen tolerization. As such, B7-H4 is an immune checkpoint of potential therapeutic interest. To generate anti-B7-H4 targeting reagents, we isolated antibodies by differential cell screening of a yeast-display library of recombinant antibodies (scFvs) derived from ovarian cancer patients and we screened for functional scFvs capable to interfere with B7-H4-mediated inhibition of antitumor responses. We found one antibody binding to B7-H4 that could restore antitumor T cell responses. This chapter gives an overview of the methods we developed to isolate a functional anti-B7-H4 antibody fragment.


Subject(s)
Ovarian Neoplasms/immunology , Saccharomyces cerevisiae/genetics , Single-Chain Antibodies/isolation & purification , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Animals , Antigen Presentation , B-Lymphocytes/immunology , Female , Gene Library , Humans , Mice , Ovarian Neoplasms/blood , Saccharomyces cerevisiae/metabolism , Single-Chain Antibodies/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
14.
Blood ; 124(7): 1070-80, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24986688

ABSTRACT

With the notable exception of B-cell malignancies, the efficacy of chimeric antigen receptor (CAR) T cells has been limited, and CAR T cells have not been shown to expand and persist in patients with nonlymphoid tumors. Here we demonstrate that redirection of primary human T cells with a CAR containing the inducible costimulator (ICOS) intracellular domain generates tumor-specific IL-17-producing effector cells that show enhanced persistence. Compared with CARs containing the CD3ζ chain alone, or in tandem with the CD28 or the 4-1BB intracellular domains, ICOS signaling increased IL-17A, IL-17F, and IL-22 following antigen recognition. In addition, T cells redirected with an ICOS-based CAR maintained a core molecular signature characteristic of TH17 cells and expressed higher levels of RORC, CD161, IL1R-1, and NCS1. Of note, ICOS signaling also induced the expression of IFN-γ and T-bet, consistent with a TH17/TH1 bipolarization. When transferred into mice with established tumors, TH17 cells that were redirected with ICOS-based CARs mediated efficient antitumor responses and showed enhanced persistence compared with CD28- or 4-1BB-based CAR T cells. Thus, redirection of TH17 cells with a CAR encoding the ICOS intracellular domain is a promising approach to augment the function and persistence of CAR T cells in hematologic malignancies.


Subject(s)
Inducible T-Cell Co-Stimulator Protein/immunology , Receptors, Antigen, T-Cell/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Line, Tumor , Cells, Cultured , Flow Cytometry , Humans , Immunotherapy, Adoptive/methods , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukins/immunology , Interleukins/metabolism , K562 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Th1 Cells/metabolism , Th17 Cells/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Xenograft Model Antitumor Assays , Interleukin-22
15.
J Nanosci Nanotechnol ; 14(1): 115-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24730254

ABSTRACT

Nanoparticles are diligently crafted with exacting control over size, shape, and composition. The pristine nanoparticles are rigorously characterized in vitro by numerous physical and materials science techniques. Immediately after being exposed to body fluids, nanoparticles interact with a heterogeneous mixture of proteins and numerous different cell types that modify the nanoparticle surface and affect their bioavailability. Understanding the mechanisms behind the recognition and elimination of these modified nanoparticles is the key to the successful translation of nanomaterials from preclinical to clinical applications. This paper reviews the anatomy of the primary organs (kidney, liver, and spleen) responsible for nanoparticle bioelimination and the components of the innate immune system (complement system and scavenger receptors) that indirectly and directly recognize nanoparticles as foreign. Recent results using PEG as a steric barrier, generating biomimetic nanoparticles, and the effect of nanoparticle material properties to increase the bioavailability of nanoparticles are presented.


Subject(s)
Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Drug Design , Kidney/metabolism , Liver/metabolism , Nanoparticles/chemistry , Spleen/metabolism , Animals , Biological Availability , Humans , Kidney/chemistry , Liver/chemistry , Spleen/chemistry
16.
J Hematol Oncol ; 7: 15, 2014 Feb 24.
Article in English | MEDLINE | ID: mdl-24565018

ABSTRACT

BACKGROUND: Although dendritic cell (DC) vaccines are considered to be promising treatments for advanced cancer, their production and administration is costly and labor-intensive. We developed a novel immunotherapeutic agent that links a single-chain antibody variable fragment (scFv) targeting mesothelin (MSLN), which is overexpressed on ovarian cancer and mesothelioma cells, to Mycobacterium tuberculosis (MTB) heat shock protein 70 (Hsp70), which is a potent immune activator that stimulates monocytes and DCs, enhances DC aggregation and maturation and improves cross-priming of T cells mediated by DCs. METHODS: Binding of this fusion protein with MSLN on the surface of tumor cells was measured by flow cytometry and fluorescence microscopy. The therapeutic efficacy of this fusion protein was evaluated in syngeneic and orthotopic mouse models of papillary ovarian cancer and malignant mesothelioma. Mice received 4 intraperitoneal (i.p.) treatments with experimental or control proteins post i.p. injection of tumor cells. Ascites-free and overall survival time was measured. For the investigation of anti-tumor T-cell responses, a time-matched study was performed. Splenocytes were stimulated with peptides, and IFNγ- or Granzyme B- generating CD3+CD8+ T cells were detected by flow cytometry. To examine the role of CD8+ T cells in the antitumor effect, we performed in vivo CD8+ cell depletion. We further determined if the fusion protein increases DC maturation and improves antigen presentation as well as cross-presentation by DCs. RESULTS: We demonstrated in vitro that the scFvMTBHsp70 fusion protein bound to the tumor cells used in this study through the interaction of scFv with MSLN on the surface of these cells, and induced maturation of bone marrow-derived DCs. Use of this bifunctional fusion protein in both mouse models significantly enhanced survival and slowed tumor growth while augmenting tumor-specific CD8+ T-cell dependent immune responses. We also demonstrated in vitro and in vivo that the fusion protein enhanced antigen presentation and cross-presentation by targeting tumor antigens towards DCs. CONCLUSIONS: This new cancer immunotherapy has the potential to be cost-effective and broadly applicable to tumors that overexpress mesothelin.


Subject(s)
Cancer Vaccines/pharmacology , GPI-Linked Proteins/immunology , HSP70 Heat-Shock Proteins/immunology , Lung Neoplasms/therapy , Mesothelioma/therapy , Ovarian Neoplasms/therapy , Animals , Cancer Vaccines/immunology , Dendritic Cells/immunology , Disease Models, Animal , Disease-Free Survival , Female , GPI-Linked Proteins/pharmacology , HSP70 Heat-Shock Proteins/genetics , Humans , Lung Neoplasms/immunology , Mesothelin , Mesothelioma/immunology , Mesothelioma, Malignant , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Survival Analysis
17.
Oncoimmunology ; 2(8): e25913, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24083083

ABSTRACT

B7-H4 inhibits T-cell activation and is widely expressed by solid neoplasms. We have recently demonstrated that the expression of B7-H4 on the surface of malignant cells in vivo is inducible, and that novel anti-B7-H4 recombinant antibodies can reverse the inhibition of tumor-specific T cells. Thus, antibodies targeting the B7-H4 pathways may extend the survival of cancer patients by restoring T cell-mediated antitumor responses.

18.
J Biomed Nanotechnol ; 9(10): 1686-97, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24015498

ABSTRACT

Nanotechnology is actively being developed for preclinical and clinical oncology applications. Nanoparticle-based immunotargeting against tumor antigens with antibodies or antibody fragments is designed to increase the nanoparticle concentration at the tumor site. However, chemical-based strategies for bioconjugating antibody fragments to nanoparticles typically result in a functionally heterogeneous population of conjugates due to alteration of amino acids within the antigen binding site. The loss of function can be prevented by expressing recombinant antibodies that contain a unique bioconjugation site, which is isolated from the antigen binding site. Biobodies are antibody fragments biosynthetically biotinylated by yeast at a specific biotin acceptor site and secreted into the culture supernatant. The high specificity and affinity between streptavidin-labeled nanoparticles and soluble biobodies allow self-assembly of immunotargeted nanoparticles directly in the yeast culture supernatant. Here, we demonstrate the versatility of biobodies for nanoparticle immunotargeting using streptavidin-labeled superparamagnetic iron oxide nanoparticles as a general, modular scaffold. Biobody-mediated targeting was performed against two antigens (mesothelin and TEM1) that are upregulated in solid tumors. The technology for biosynthetic biotinylation can be extended to proteins other than antibody fragments and adopted by fields outside of oncology for directed modification of any streptavidin-functionalized surface.


Subject(s)
Antigens, Neoplasm/immunology , Biotin/immunology , Immunoglobulin Fragments/immunology , Nanoparticles/chemistry , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Biotinylation/methods , Cell Line, Tumor , Contrast Media/chemical synthesis , Humans , Nanoparticles/ultrastructure
19.
Methods Mol Biol ; 1049: 425-33, 2013.
Article in English | MEDLINE | ID: mdl-23913235

ABSTRACT

Syngeneic and transgenic mouse models are important tools for the study of the biology of cancer. While syngeneic mouse models are generated through the implantation in host animals of tumor cells from genetically and immunologically compatible donors, transgenic mouse models are engineered to express genetic material with oncogenic properties in predetermined location. We have developed a syngeneic mouse model of ovarian cancer permitting in vivo imaging in immunocompetent recipients by implanting ovaries with fluorescently labeled cancer cells that derived from a spontaneous ovarian tumor developing in a transgenic mouse model. Tumor cells were retrovirally transduced with a far-red fluorescent protein. This animal model combines the advantages of syngeneic and transgenic mouse models as it permits to both monitor tumor growth by in vivo imaging and to analyze the tumor microenvironment of an immunocompetent host.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Molecular Imaging/methods , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Female , Green Fluorescent Proteins , Humans , Luminescent Proteins , Mice , Ovarian Neoplasms/genetics , Tumor Microenvironment , Red Fluorescent Protein
20.
Cancer Res ; 73(15): 4820-9, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23722540

ABSTRACT

B7-H4 (VTCN1, B7x, B7s) is a ligand for inhibitory coreceptors on T cells implicated in antigenic tolerization. B7-H4 is expressed by tumor cells and tumor-associated macrophages (TAM), but its potential contributions to tumoral immune escape and therapeutic targeting have been less studied. To interrogate B7-H4 expression on tumor cells, we analyzed fresh primary ovarian cancer cells collected from patient ascites and solid tumors, and established cell lines before and after in vivo passaging. B7-H4 expression was detected on the surface of all fresh primary human tumors and tumor xenotransplants, but not on most established cell lines, and B7-H4 was lost rapidly by tumor xenograft cells after short-term in vitro culture. These results indicated an in vivo requirement for B7-H4 induction and defined conditions for targeting studies. To generate anti-B7-H4-targeting reagents, we isolated antibodies by differential cell screening of a yeast-display single-chain fragments variable (scFv) library derived from patients with ovarian cancer. We identified anti-B7-H4 scFv that reversed in vitro inhibition of CD3-stimulated T cells by B7-H4 protein. Notably, these reagents rescued tumor antigen-specific T-cell activation, which was otherwise inhibited by coculture with antigen-loaded B7-H4+ APCs, B7-H4+ tumor cells, or B7-H4- tumor cells mixed with B7-H4+ TAMs; peritoneal administration of anti-B7-H4 scFv delayed the growth of established tumors. Together, our findings showed that cell surface expression of B7-H4 occurs only in tumors in vivo and that antibody binding of B7-H4 could restore antitumor T-cell responses. We suggest that blocking of B7-H4/B7-H4 ligand interactions may represent a feasible therapeutic strategy for ovarian cancer.


Subject(s)
Ovarian Neoplasms/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/immunology , Animals , Ascites , Cell Line, Tumor , Female , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL