Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nature ; 545(7655): 495-499, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28514441

ABSTRACT

Programmed cell death protein 1 (PD-1) is an immune checkpoint receptor that is upregulated on activated T cells for the induction of immune tolerance. Tumour cells frequently overexpress the ligand for PD-1, programmed cell death ligand 1 (PD-L1), facilitating their escape from the immune system. Monoclonal antibodies that block the interaction between PD-1 and PD-L1, by binding to either the ligand or receptor, have shown notable clinical efficacy in patients with a variety of cancers, including melanoma, colorectal cancer, non-small-cell lung cancer and Hodgkin's lymphoma. Although it is well established that PD-1-PD-L1 blockade activates T cells, little is known about the role that this pathway may have in tumour-associated macrophages (TAMs). Here we show that both mouse and human TAMs express PD-1. TAM PD-1 expression increases over time in mouse models of cancer and with increasing disease stage in primary human cancers. TAM PD-1 expression correlates negatively with phagocytic potency against tumour cells, and blockade of PD-1-PD-L1 in vivo increases macrophage phagocytosis, reduces tumour growth and lengthens the survival of mice in mouse models of cancer in a macrophage-dependent fashion. This suggests that PD-1-PD-L1 therapies may also function through a direct effect on macrophages, with substantial implications for the treatment of cancer with these agents.


Subject(s)
Colonic Neoplasms/immunology , Macrophages/immunology , Macrophages/metabolism , Phagocytosis , Programmed Cell Death 1 Receptor/metabolism , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Humans , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Neoplasm Staging , Phagocytosis/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Xenograft Model Antitumor Assays
2.
Proc Natl Acad Sci U S A ; 112(47): E6506-14, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26604307

ABSTRACT

Signaling through the immune checkpoint programmed cell death protein-1 (PD-1) enables tumor progression by dampening antitumor immune responses. Therapeutic blockade of the signaling axis between PD-1 and its ligand programmed cell death ligand-1 (PD-L1) with monoclonal antibodies has shown remarkable clinical success in the treatment of cancer. However, antibodies have inherent limitations that can curtail their efficacy in this setting, including poor tissue/tumor penetrance and detrimental Fc-effector functions that deplete immune cells. To determine if PD-1:PD-L1-directed immunotherapy could be improved with smaller, nonantibody therapeutics, we used directed evolution by yeast-surface display to engineer the PD-1 ectodomain as a high-affinity (110 pM) competitive antagonist of PD-L1. In contrast to anti-PD-L1 monoclonal antibodies, high-affinity PD-1 demonstrated superior tumor penetration without inducing depletion of peripheral effector T cells. Consistent with these advantages, in syngeneic CT26 tumor models, high-affinity PD-1 was effective in treating both small (50 mm(3)) and large tumors (150 mm(3)), whereas the activity of anti-PD-L1 antibodies was completely abrogated against large tumors. Furthermore, we found that high-affinity PD-1 could be radiolabeled and applied as a PET imaging tracer to efficiently distinguish between PD-L1-positive and PD-L1-negative tumors in living mice, providing an alternative to invasive biopsy and histological analysis. These results thus highlight the favorable pharmacology of small, nonantibody therapeutics for enhanced cancer immunotherapy and immune diagnostics.


Subject(s)
Immunotherapy , Mutant Proteins/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/therapy , Positron-Emission Tomography , Programmed Cell Death 1 Receptor/therapeutic use , Protein Engineering , Amino Acid Sequence , Animals , Cell Line, Tumor , Directed Molecular Evolution , Disease Models, Animal , Humans , Lymphocyte Depletion , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms/immunology , Programmed Cell Death 1 Receptor/chemistry , Protein Binding , T-Lymphocytes/metabolism
3.
Proc Natl Acad Sci U S A ; 111(3): 1108-13, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24390540

ABSTRACT

The noninvasive detection and quantification of CD8(+) T cells in vivo are important for both the detection and staging of CD8(+) lymphomas and for the monitoring of successful cancer immunotherapies, such as adoptive cell transfer and antibody-based immunotherapeutics. Here, antibody fragments are constructed to target murine CD8 to obtain rapid, high-contrast immuno-positron emission tomography (immuno-PET) images for the detection of CD8 expression in vivo. The variable regions of two anti-murine CD8-depleting antibodies (clones 2.43 and YTS169.4.2.1) were sequenced and reformatted into minibody (Mb) fragments (scFv-CH3). After production and purification, the Mbs retained their antigen specificity and bound primary CD8(+) T cells from the thymus, spleen, lymph nodes, and peripheral blood. Importantly, engineering of the parental antibodies into Mbs abolished the ability to deplete CD8(+) T cells in vivo. The Mbs were subsequently conjugated to S-2-(4-isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid for (64)Cu radiolabeling. The radiotracers were injected i.v. into antigen-positive, antigen-negative, immunodeficient, antigen-blocked, and antigen-depleted mice to evaluate specificity of uptake in lymphoid tissues by immuno-PET imaging and ex vivo biodistribution. Both (64)Cu-radiolabeled Mbs produced high-contrast immuno-PET images 4 h postinjection and showed specific uptake in the spleen and lymph nodes of antigen-positive mice.


Subject(s)
Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/cytology , Immunoglobulin Fragments/immunology , Positron-Emission Tomography , Alleles , Animals , Antibody Specificity/immunology , Antigens/chemistry , Copper Radioisotopes/chemistry , Epitopes/chemistry , Flow Cytometry , Immunotherapy/methods , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Rats , Tissue Distribution
4.
Proc Natl Acad Sci U S A ; 111(28): E2866-74, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982199

ABSTRACT

PET is a powerful technique for quantifying and visualizing biochemical pathways in vivo. Here, we develop and validate a novel PET probe, [(18)F]-2-deoxy-2-fluoroarabinose ([(18)F]DFA), for in vivo imaging of ribose salvage. DFA mimics ribose in vivo and accumulates in cells following phosphorylation by ribokinase and further metabolism by transketolase. We use [(18)F]DFA to show that ribose preferentially accumulates in the liver, suggesting a striking tissue specificity for ribose metabolism. We demonstrate that solute carrier family 2, member 2 (also known as GLUT2), a glucose transporter expressed in the liver, is one ribose transporter, but we do not know if others exist. [(18)F]DFA accumulation is attenuated in several mouse models of metabolic syndrome, suggesting an association between ribose salvage and glucose and lipid metabolism. These results describe a tool for studying ribose salvage and suggest that plasma ribose is preferentially metabolized in the liver.


Subject(s)
Liver , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Ribose/metabolism , Animals , Arabinose/analogs & derivatives , Arabinose/pharmacology , Cell Line , Disease Models, Animal , Fluorine Radioisotopes/pharmacology , Glucose/genetics , Glucose/metabolism , Glucose Transporter Type 2/metabolism , Humans , Lipid Metabolism , Liver/diagnostic imaging , Liver/metabolism , Metabolic Syndrome/diagnostic imaging , Metabolic Syndrome/metabolism , Mice , Organ Specificity , Radiography
5.
Proc Natl Acad Sci U S A ; 110(5): 1857-62, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23319634

ABSTRACT

Positron emission tomography (PET) reporter genes allow noninvasive whole-body imaging of transplanted cells by detection with radiolabeled probes. We used a human deoxycytidine kinase containing three amino acid substitutions within the active site (hdCK3mut) as a reporter gene in combination with the PET probe [(18)F]-L-FMAU (1-(2-deoxy-2-(18)fluoro-ß-L-arabinofuranosyl)-5-methyluracil) to monitor models of mouse and human hematopoietic stem cell (HSC) transplantation. These mutations in hdCK3mut expanded the substrate capacity allowing for reporter-specific detection with a thymidine analog probe. Measurements of long-term engrafted cells (up to 32 wk) demonstrated that hdCK3mut expression is maintained in vivo with no counter selection against reporter-labeled cells. Reporter cells retained equivalent engraftment and differentiation capacity being detected in all major hematopoietic lineages and tissues. This reporter gene and probe should be applicable to noninvasively monitor therapeutic cell transplants in multiple tissues.


Subject(s)
Deoxycytidine Kinase/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/chemistry , Arabinofuranosyluracil/metabolism , Blotting, Western , Cell Line, Tumor , Deoxycytidine Kinase/genetics , Female , Fluorine Radioisotopes/chemistry , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Kaplan-Meier Estimate , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Mice, Knockout , Mice, SCID , Mutation , Thymus Gland/diagnostic imaging , Thymus Gland/metabolism , Time Factors , Transplantation, Heterologous
7.
Blood Adv ; 3(18): 2713-2721, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31519647

ABSTRACT

Peritoneal adhesions are pathological fibroses that ensnare organs after abdominal surgery. This dense connective tissue can cause small bowel obstruction, female infertility, and chronic abdominal pain. The pathogenesis of adhesions is a fibrotic response to tissue damage coordinated between mesothelial cells, fibroblasts, and immune cells. We have previously demonstrated that peritoneal adhesions are a consequence of mechanical injury to the mesothelial layer sustained during surgery. Neutrophils are among the first leukocytes involved in the early response to tissue damage. Here, we show that when subjected to mechanical stress, activated mesothelial cells directly recruit neutrophils and monocytes through upregulation of chemokines such as CXCL1 and monocyte chemoattractant protein 1 (MCP-1). We find that neutrophils within the adhesion sites undergo cell death and form neutrophil extracellular traps (NETosis) that contribute to pathogenesis. Conversely, tissue-resident macrophages were profoundly depleted throughout the disease time course. We show that this is distinct from traditional inflammatory kinetics such as after sham surgery or chemically induced peritonitis, and suggest that adhesions result from a primary difference in inflammatory kinetics. We find that transient depletion of circulating neutrophils significantly decreases adhesion burden, and further recruitment of monocytes with thioglycolate or MCP-1 also improves outcomes. Our findings suggest that the combination of neutrophil depletion and monocyte recruitment is sufficient to prevent adhesion formation, thus providing insight for potential clinical interventions.


Subject(s)
Monocytes/metabolism , Neutrophils/metabolism , Tissue Adhesions/metabolism , Animals , Female , Humans , Mice
8.
Methods Mol Biol ; 1790: 137-151, 2018.
Article in English | MEDLINE | ID: mdl-29858789

ABSTRACT

Positron emission tomography (PET) is a three dimensional imaging modality that detects the accumulation of radiolabeled isotopes in vivo. Ectopic expression of a thymidine kinase reporter gene allows for the specific detection of reporter cells in vivo by imaging with the reporter specific probe. PET reporter imaging is sensitive, quantitative and can be scaled into larger tumors or animals with little to no tissue diffraction. Here, we describe how thymidine kinase PET reporter genes can be used to noninvasively image cancer cells in vivo.


Subject(s)
Genes, Reporter , Molecular Imaging/methods , Positron-Emission Tomography/methods , Prostatic Neoplasms/pathology , Thymidine Kinase/metabolism , Animals , Gene Expression Regulation, Neoplastic , Herpesvirus 1, Human/genetics , Humans , Male , Mice , Mice, Nude , Mice, SCID , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Thymidine Kinase/genetics , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Nucl Med ; 58(4): 538-546, 2017 04.
Article in English | MEDLINE | ID: mdl-27980047

ABSTRACT

Immune checkpoint blockade has emerged as a promising cancer treatment paradigm. Unfortunately, there are still a large number of patients and malignancies that do not respond to therapy. A major barrier to validating biomarkers for the prediction and monitoring of responders to clinical checkpoint blockade has been the lack of imaging tools to accurately assess dynamic immune checkpoint expression. Here, we sought to optimize noninvasive immuno-PET imaging of human programmed death-ligand 1 (PD-L1) expression, in a preclinical model, using a small high-affinity engineered protein scaffold (HAC-PD1). Six HAC-PD1 radiotracer variants were developed and used in preclinical imaging and biodistribution studies to assess their ability to detect human PD-L1 expression in vivo. Radiotracer design modifications included chelate, glycosylation, and radiometal. HACA-PD1 was adopted as the naming convention for aglycosylated tracer variants. NOD scid γ-(NSG) mice were inoculated with subcutaneous tumors engineered to either be constitutively positive (CT26 hPD-L1) or be negative (ΔmPD-L1 CT26) for human PD-L1 expression. When the tumors had grown to an average size of 1 cm in diameter, mice were injected with 0.75-2.25 MBq (∼10 µg) of an engineered radiotracer variant and imaged. At 1 h after injection, organs were harvested for biodistribution. Of the practical immuno-PET tracer modifications considered, glycosylation was the most prominent design factor affecting tracer uptake, specificity, and clearance. In imaging studies, aglycosylated 64Cu-NOTA-HACA-PD1 most accurately visualized human PD-L1 expression in vivo. We reasoned that because of the scaffold's small size (14 kDa), its pharmacokinetics may be suitable for labeling with the short-lived and widely clinically available radiometal 68Ga. At 1 h after injection, 68Ga-NOTA-HACA-PD1 and 68Ga-DOTA-HACA-PD1 exhibited promising target-to-background ratios in ex vivo biodistribution studies (12.3 and 15.2 tumor-to-muscle ratios, respectively). Notably, all HAC-PD1 radiotracer variants enabled much earlier detection of human PD-L1 expression (1 h after injection) than previously reported radiolabeled antibodies (>24 h after injection). This work provides a template for assessing immuno-PET tracer design parameters and supports the translation of small engineered protein radiotracers for imaging human immune checkpoints.


Subject(s)
Drug Design , Immunoconjugates , Positron-Emission Tomography/methods , Animals , B7-H1 Antigen/chemistry , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Cell Line, Tumor , Copper Radioisotopes , Gene Expression Regulation, Neoplastic , Glycosylation , Heterocyclic Compounds, 1-Ring/chemistry , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Mice , Models, Molecular , Protein Conformation , Protein Engineering , Radioactive Tracers , Tissue Distribution
10.
Article in English | MEDLINE | ID: mdl-28416577

ABSTRACT

A stem cell is broadly defined as a cell that retains the capacity to self-renew, a feature that confers the ability to continuously make identical daughter cells or additional cells that will differentiate into downstream progeny. This highly regulated genetic program to retain "stemness" is under active investigation. Research in our laboratory has explored similarities and differences in embryonic, tissue-specific, and neoplastic stem cells and their terminally differentiated counterparts. In this review, we will focus on the contributions of our laboratory, in particular on the studies that identified the mouse hematopoietic stem cell (HSC) and the human leukemic stem cell. These studies have led to significant improvements in both preclinical and clinical research, including improved clinical bone marrow transplantation protocols, isolation of nonleukemic HSCs, a cancer immunotherapy currently in clinical trials, and development of a HSC reporter mouse. These studies and the current follow-up research by us and others will continue to identify the properties, function, and regulation of both normal and neoplastic stem cells.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Transformation, Neoplastic/metabolism , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Transplantation/methods , Humans
11.
Cancer Res ; 76(1): 73-82, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26573799

ABSTRACT

The rapidly advancing field of cancer immunotherapy is currently limited by the scarcity of noninvasive and quantitative technologies capable of monitoring the presence and abundance of CD8(+) T cells and other immune cell subsets. In this study, we describe the generation of (89)Zr-desferrioxamine-labeled anti-CD8 cys-diabody ((89)Zr-malDFO-169 cDb) for noninvasive immuno-PET tracking of endogenous CD8(+) T cells. We demonstrate that anti-CD8 immuno-PET is a sensitive tool for detecting changes in systemic and tumor-infiltrating CD8 expression in preclinical syngeneic tumor immunotherapy models including antigen-specific adoptive T-cell transfer, agonistic antibody therapy (anti-CD137/4-1BB), and checkpoint blockade antibody therapy (anti-PD-L1). The ability of anti-CD8 immuno-PET to provide whole body information regarding therapy-induced alterations of this dynamic T-cell population provides new opportunities to evaluate antitumor immune responses of immunotherapies currently being evaluated in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes/diagnostic imaging , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Positron-Emission Tomography/methods , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Zirconium/administration & dosage , Animals , Antibodies, Bispecific , CD8 Antigens , Colonic Neoplasms/immunology , Deferoxamine/administration & dosage , Deferoxamine/chemistry , Deferoxamine/immunology , Disease Models, Animal , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Immunoconjugates/immunology , Lymphocytes, Tumor-Infiltrating/diagnostic imaging , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radioisotopes/chemistry , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/immunology , Zirconium/chemistry
12.
PLoS One ; 11(4): e0153550, 2016.
Article in English | MEDLINE | ID: mdl-27092773

ABSTRACT

Tumor-associated macrophages (TAMs) represent an important cellular subset within the glioblastoma (WHO grade IV) microenvironment and are a potential therapeutic target. TAMs display a continuum of different polarization states between antitumorigenic M1 and protumorigenic M2 phenotypes, with a lower M1/M2 ratio correlating with worse prognosis. Here, we investigated the effect of macrophage polarization on anti-CD47 antibody-mediated phagocytosis of human glioblastoma cells in vitro, as well as the effect of anti-CD47 on the distribution of M1 versus M2 macrophages within human glioblastoma cells grown in mouse xenografts. Bone marrow-derived mouse macrophages and peripheral blood-derived human macrophages were polarized in vitro toward M1 or M2 phenotypes and verified by flow cytometry. Primary human glioblastoma cell lines were offered as targets to mouse and human M1 or M2 polarized macrophages in vitro. The addition of an anti-CD47 monoclonal antibody led to enhanced tumor-cell phagocytosis by mouse and human M1 and M2 macrophages. In both cases, the anti-CD47-induced phagocytosis by M1 was more prominent than that for M2. Dissected tumors from human glioblastoma xenografted within NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice and treated with anti-CD47 showed a significant increase of M1 macrophages within the tumor. These data show that anti-CD47 treatment leads to enhanced tumor cell phagocytosis by both M1 and M2 macrophage subtypes with a higher phagocytosis rate by M1 macrophages. Furthermore, these data demonstrate that anti-CD47 treatment alone can shift the phenotype of macrophages toward the M1 subtype in vivo.


Subject(s)
Antibodies/pharmacology , CD47 Antigen/metabolism , Glioblastoma/drug therapy , Glioblastoma/pathology , Macrophages/drug effects , Macrophages/pathology , Phagocytosis/drug effects , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Glioblastoma/metabolism , Humans , Macrophages/metabolism , Mice , Mice, Inbred NOD , Phenotype
13.
Clin Cancer Res ; 21(16): 3597-601, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26116271

ABSTRACT

Recent advances with immunotherapy agents for the treatment of cancer have provided remarkable, and in some cases, curative results. Our laboratory has identified CD47 as an important "don't eat me" signal expressed on malignant cells. Blockade of the CD47:SIRP-α axis between tumor cells and innate immune cells (monocytes, macrophages, and dendritic cells) increases tumor cell phagocytosis in both solid tumors (including, but not limited to, bladder, breast, colon, lung, and pancreatic) and hematologic malignancies. These phagocytic innate cells are also professional antigen-presenting cells (APC), providing a link from innate to adaptive antitumor immunity. Preliminary studies have demonstrated that APCs present antigens from phagocytosed tumor cells, causing T-cell activation. Therefore, agents that block the CD47:SIRP-α engagement are attractive therapeutic targets as a monotherapy or in combination with additional immune-modulating agents for activating antitumor T cells in vivo.


Subject(s)
Antigens, Differentiation/genetics , CD47 Antigen/genetics , Immunity, Innate/drug effects , Lymphocyte Activation/immunology , Neoplasms/drug therapy , Receptors, Immunologic/genetics , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Differentiation/immunology , CD47 Antigen/drug effects , CD47 Antigen/immunology , Cytophagocytosis/drug effects , Cytophagocytosis/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Neoplasms/genetics , Neoplasms/pathology , Receptors, Immunologic/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
14.
J Clin Invest ; 125(5): 1815-26, 2015 May.
Article in English | MEDLINE | ID: mdl-25822024

ABSTRACT

Adoptive transfer of tumor-reactive T cells can successfully reduce tumor burden; however, in rare cases, lethal on-target/off-tumor effects have been reported. A noninvasive method to track engineered cells with high sensitivity and resolution would allow observation of correct cell homing and/or identification of dangerous off-target locations in preclinical and clinical applications. Human deoxycytidine kinase triple mutant (hdCK3mut) is a nonimmunogenic PET reporter that was previously shown to be an effective tool to monitor whole-body hematopoiesis. Here, we engineered a construct in which hdCK3mut is coexpressed with the anti-melanoma T cell receptor F5, introduced this construct into human CD34 cells or PBMCs, and evaluated this approach in multiple immunotherapy models. Expression of hdCK3mut allowed engrafted cells to be visualized within recipient bone marrow, while accumulation of [18F]-L-FMAU in hdCK3mut-expressing T cells permitted detection of intratumoral homing. Animals that received T cells coexpressing hdCK3mut and the anti-melanoma T cell receptor had demonstrably higher signals in HLA-matched tumors compared with those in animals that received cells solely expressing hdCK3mut. Engineered T cells caused cytotoxicity in HLA/antigen-matched tumors and induced IFN-γ production and activation. Moreover, hdCK3mut permitted simultaneous monitoring of engraftment and tumor infiltration, without affecting T cell function. Our findings suggest that hdCK3mut reporter imaging can be applied in clinical immunotherapies for whole-body detection of engineered cell locations.


Subject(s)
Deoxycytidine Kinase/analysis , Genes, Reporter , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Positron-Emission Tomography , T-Lymphocytes/immunology , Animals , Bone Marrow/diagnostic imaging , Chemotaxis, Leukocyte , Cytotoxicity Tests, Immunologic , Deoxycytidine Kinase/genetics , Genes, Synthetic , Genetic Vectors/genetics , Graft Survival , HLA-A2 Antigen/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/physiology , Humans , Immunotherapy, Adoptive , Interferon-gamma Release Tests , Lentivirus/genetics , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/physiology , Leukocytes, Mononuclear/transplantation , MART-1 Antigen/immunology , Melanoma, Experimental/immunology , Mice , Mutation , Receptors, Antigen, T-Cell/analysis , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Retroviridae/genetics , T-Lymphocytes/transplantation , Thymus Gland/transplantation
15.
J Nucl Med ; 56(8): 1258-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25952734

ABSTRACT

UNLABELLED: The proliferation and trafficking of T lymphocytes in immune responses are crucial events in determining inflammatory responses. To study whole-body T lymphocyte dynamics noninvasively in vivo, we generated anti-CD4 and -CD8 cys-diabodies (cDbs) derived from the parental antibody hybridomas GK1.5 and 2.43, respectively, for (89)Zr-immuno-PET detection of helper and cytotoxic T cell populations. METHODS: Anti-CD4 and -CD8 cDbs were engineered, produced via mammalian expression, purified using immobilized metal affinity chromatography, and characterized for T cell binding. The cDbs were site-specifically conjugated to maleimide-desferrioxamine for (89)Zr radiolabeling and subsequent small-animal PET/CT acquisition and ex vivo biodistribution in both wild-type mice and a model of hematopoietic stem cell (HSC) transplantation. RESULTS: Immuno-PET and biodistribution studies demonstrate targeting and visualization of CD4 and CD8 T cell populations in vivo in the spleen and lymph nodes of wild-type mice, with specificity confirmed through in vivo blocking and depletion studies. Subsequently, a murine model of HSC transplantation demonstrated successful in vivo detection of T cell repopulation at 2, 4, and 8 wk after HSC transplantation using the (89)Zr-radiolabeled anti-CD4 and -CD8 cDbs. CONCLUSION: These newly developed anti-CD4 and -CD8 immuno-PET reagents represent a powerful resource to monitor T cell expansion, localization, and novel engraftment protocols. Future potential applications of T cell-targeted immuno-PET include monitoring immune cell subsets in response to immunotherapy, autoimmunity, and lymphoproliferative disorders, contributing overall to preclinical immune cell monitoring.


Subject(s)
CD4 Antigens/metabolism , CD8 Antigens/metabolism , Hematopoietic Stem Cell Transplantation , Maleimides/chemistry , Positron-Emission Tomography/methods , Single-Chain Antibodies , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Chromatography, Affinity , Immunotherapy/methods , Mice , Mice, Inbred C57BL , T-Lymphocytes/cytology , Time Factors , Tissue Distribution , Zirconium/chemistry
16.
PLoS One ; 9(8): e104125, 2014.
Article in English | MEDLINE | ID: mdl-25101980

ABSTRACT

Efficient and adequate generation of deoxyribonucleotides is critical to successful DNA repair. We show that ataxia telangiectasia mutated (ATM) integrates the DNA damage response with DNA metabolism by regulating the salvage of deoxyribonucleosides. Specifically, ATM phosphorylates and activates deoxycytidine kinase (dCK) at serine 74 in response to ionizing radiation (IR). Activation of dCK shifts its substrate specificity toward deoxycytidine, increases intracellular dCTP pools post IR, and enhances the rate of DNA repair. Mutation of a single serine 74 residue has profound effects on murine T and B lymphocyte development, suggesting that post-translational regulation of dCK may be important in maintaining genomic stability during hematopoiesis. Using [(18)F]-FAC, a dCK-specific positron emission tomography (PET) probe, we visualized and quantified dCK activation in tumor xenografts after IR, indicating that dCK activation could serve as a biomarker for ATM function and DNA damage response in vivo. In addition, dCK-deficient leukemia cell lines and murine embryonic fibroblasts exhibited increased sensitivity to IR, indicating that pharmacologic inhibition of dCK may be an effective radiosensitization strategy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/physiology , DNA Repair/radiation effects , Deoxycytidine Kinase/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Line, Tumor , DNA Damage , DNA Repair/drug effects , Deoxycytidine/metabolism , Deoxycytidine Kinase/chemistry , Deoxycytidine Kinase/genetics , Deoxyribonucleosides/metabolism , Genomic Instability , Hematopoiesis/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutagenesis, Site-Directed , Phosphorylation , Protein Processing, Post-Translational , Substrate Specificity , T-Lymphocytes/cytology , T-Lymphocytes/physiology
17.
Cancer Res ; 74(18): 5173-83, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25038231

ABSTRACT

Engineering immunity against cancer by the adoptive transfer of hematopoietic stem cells (HSC) modified to express antigen-specific T-cell receptors (TCR) or chimeric antigen receptors generates a continual supply of effector T cells, potentially providing superior anticancer efficacy compared with the infusion of terminally differentiated T cells. Here, we demonstrate the in vivo generation of functional effector T cells from CD34-enriched human peripheral blood stem cells modified with a lentiviral vector designed for clinical use encoding a TCR recognizing the cancer/testes antigen NY-ESO-1, coexpressing the PET/suicide gene sr39TK. Ex vivo analysis of T cells showed antigen- and HLA-restricted effector function against melanoma. Robust engraftment of gene-modified human cells was demonstrated with PET reporter imaging in hematopoietic niches such as femurs, humeri, vertebrae, and the thymus. Safety was demonstrated by the in vivo ablation of PET signal, NY-ESO-1-TCR-bearing cells, and integrated lentiviral vector genomes upon treatment with ganciclovir, but not with vehicle control. Our study provides support for the efficacy and safety of gene-modified HSCs as a therapeutic modality for engineered cancer immunotherapy. Cancer Res; 74(18); 5173-83. ©2014 AACR.


Subject(s)
Genes, Transgenic, Suicide , Hematopoietic Stem Cells/physiology , Herpesvirus 1, Human/genetics , Immunotherapy/methods , Positron-Emission Tomography/methods , T-Lymphocytes/immunology , Animals , Antigens, CD34/blood , Antigens, CD34/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Disease Models, Animal , Genetic Therapy , Genetic Vectors/genetics , Hematopoietic Stem Cells/diagnostic imaging , Hematopoietic Stem Cells/immunology , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL