Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Cell Rep Med ; 5(3): 101469, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508137

ABSTRACT

Fibrolamellar carcinoma (FLC) is a liver tumor with a high mortality burden and few treatment options. A promising therapeutic vulnerability in FLC is its driver mutation, a conserved DNAJB1-PRKACA gene fusion that could be an ideal target neoantigen for immunotherapy. In this study, we aim to define endogenous CD8 T cell responses to this fusion in FLC patients and evaluate fusion-specific T cell receptors (TCRs) for use in cellular immunotherapies. We observe that fusion-specific CD8 T cells are rare and that FLC patient TCR repertoires lack large clusters of related TCR sequences characteristic of potent antigen-specific responses, potentially explaining why endogenous immune responses are insufficient to clear FLC tumors. Nevertheless, we define two functional fusion-specific TCRs, one of which has strong anti-tumor activity in vivo. Together, our results provide insights into the fragmented nature of neoantigen-specific repertoires in humans and indicate routes for clinical development of successful immunotherapies for FLC.


Subject(s)
Carcinoma, Hepatocellular , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/pathology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/pathology , Cell- and Tissue-Based Therapy , HSP40 Heat-Shock Proteins/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics
2.
Front Immunol ; 14: 1199747, 2023.
Article in English | MEDLINE | ID: mdl-37638040

ABSTRACT

Multiple Sclerosis (MS) is a chronic neurodegenerative disease with limited therapeutic options. Recombinant Fc multimers (rFc), designed to mirror many of the anti-inflammatory activities of Intravenous Immunoglobulin (IVIG), have been shown to effectively treat numerous immune-mediated diseases in rodents. In this study we used the experimental autoimmune encephalomyelitis (EAE) murine model of MS to test the efficacy of a rFc, M019, that consists of multimers of the Fc portion of IgG2, in inhibiting disease severity. We show that M019 effectively reduced clinical symptoms when given either pre- or post-symptom onset compared to vehicle treated EAE induced mice. M019 was effective in reducing symptoms in both SJL model of relapsing remitting MS as well as the B6 model of chronic disease. M019 binds to FcγR bearing-monocytes both in vivo and in vitro and prevented immune cell infiltration into the CNS of treated mice. The lack of T cell infiltration into the spinal cord was not due to a decrease in T cell priming; there was an equivalent frequency of Th17 cells in the spleens of M019 and vehicle treated EAE induced mice. Surprisingly, there was an increase in chemokines in the sera but not in the CNS of M019 treated mice compared to vehicle treated animals. We postulate that M019 interacts with a FcγR rich monocyte intermediary to prevent T cell migration into the CNS and demyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Neurodegenerative Diseases , Animals , Mice , Multiple Sclerosis/drug therapy , Disease Models, Animal , Receptors, IgG
3.
Immunogenetics ; 75(4): 369-383, 2023 08.
Article in English | MEDLINE | ID: mdl-37322230

ABSTRACT

Though binding sites for the complement factor C1q and the canonical fragment crystallizable (Fc) gamma receptors (Fc[Formula: see text]Rs) on immunoglobulin G (IgG) molecules overlap, how C1q decoration of immune complexes (ICs) influences their ability to engage Fc[Formula: see text]Rs remains unknown. In this report, we use recombinant human Fc multimers as stable IC mimics to show that C1q engagement of ICs directly and transiently inhibits their interactions with Fc[Formula: see text]RIII (CD16) on human natural killer (NK) cells. This inhibition occurs by C1q engagement alone as well as in concert with other serum factors. Furthermore, the inhibition of Fc[Formula: see text]RIII engagement mediated by avid binding of C1q to ICs is directly associated with IC size and dependent on the concentrations of both C1q and Fc multimers present. Functionally, C1q-mediated Fc blockade limits the ability of NK cells to induce the upregulation of the cosignaling molecule, 4-1BB (CD137), and to mediate antibody-dependent cell-mediated cytotoxicity (ADCC). Although C1q is traditionally viewed as a soluble effector molecule, we demonstrate that C1q may also take on the role of an "immunologic rheostat," buffering Fc[Formula: see text]R-mediated activation of immune cells by circulating ICs. These data define a novel role for C1q as a regulator of immune homeostasis and add to our growing understanding that complement factors mediate pleiotropic effects.


Subject(s)
Complement C1q , Receptors, IgG , Humans , Complement C1q/metabolism , Immunoglobulin G , Antibody-Dependent Cell Cytotoxicity , Killer Cells, Natural
4.
Head Neck ; 41(8): 2514-2524, 2019 08.
Article in English | MEDLINE | ID: mdl-30869813

ABSTRACT

BACKGROUND: We sought to evaluate intratumor heterogeneity in squamous cell carcinoma of the oral cavity (OCC) and specifically determine the effect of physical separation and histologic differentiation within the same tumor. METHODS: We performed whole exome sequencing on five biopsy sites-two from well-differentiated, two from poorly differentiated regions, and one from normal parenchyma-from five primary OCC specimens. RESULTS: We found high levels of intratumor heterogeneity and, in four primary tumors, identified only 0 to 2 identical mutations in all subsites. We found that the heterogeneity inversely correlated with physical separation and that pairs of well-differentiated samples were more similar to each other than analogous poorly differentiated specimens. Only TP53 mutations, but not other purported "driver mutations" in head and neck squamous cell carcinoma, were found in multiple biopsy sites. CONCLUSION: These data highlight the challenges to characterization of the mutational landscape of OCC with single site biopsy and have implications for personalized medicine.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genetic Heterogeneity , Gingival Neoplasms/genetics , Tongue Neoplasms/genetics , Aged , Aged, 80 and over , Biopsy , Carcinoma, Squamous Cell/pathology , Gene Frequency , Gingival Neoplasms/pathology , Humans , Middle Aged , Polymorphism, Single Nucleotide , Tongue Neoplasms/pathology , Exome Sequencing
5.
Laryngoscope Investig Otolaryngol ; 4(1): 62-69, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30828620

ABSTRACT

Monoclonal antibodies (mAbs) that target immune co-signaling pathways have the potential to enable immune mediated tumor eradication. While early adoption of these agents for the treatment of advanced squamous cell carcinoma of the head and neck (SCCHN) has produced some astounding clinical successes, the majority of patients fail to respond to therapy. The purpose of this review is to first provide a broad overview of the immuno-oncology (I-O) landscape and to then focus on the current status of mAb-based I-O (mAb:I-O) for the treatment of SCCHN, with particular attention to the development of strategies for improving treatment responses.

6.
Immunobiology ; 224(3): 362-370, 2019 05.
Article in English | MEDLINE | ID: mdl-30876792

ABSTRACT

Given the heightened interest in manipulation of co-signaling cascades for cancer immunotherapy, we sought to determine how/whether tumors decorated with therapeutic monoclonal antibodies (mAbs) impact the expression of co-signaling molecules on human NK cells. Stimulation of NK cells with aggregated IgG1 resulted in the upregulation of HAVCR2 - the gene encoding T-cell immunoglobulin and mucin-containing domain (Tim)-3 - known to be involved in the induction of peripheral T cell tolerance. This upregulation of HAVCR2 was recapitulated at the protein level, following NK cell stimulation by either mAb opsonized tumors, recombinant human IgG1 Fc multimer, and/or non-Fc stimuli e.g. IL-12/IL-18. The patterns of Tim-3 expression were temporally distinct from the FcR mediated induction of the co-signaling molecule, 4-1BB (CD137), with Tim-3 increases observed twenty minutes following exposure to Fc multimers and remaining at high levels for at least six hours, while increases in CD137 expression were first observed at the four-hour time point. Importantly, these Tim-3+ NK cells were functionally diverse, as evidenced by the fact that their ability to produce IFN-γ in response to an NK cell responsive tumor was strictly dependent upon the stimuli employed for Tim-3 induction. These data suggest that Tim-3 upregulation is the common end-result of NK cell activation by a variety of unique and overlapping stimuli and is not an independent marker of NK cell exhaustion. Furthermore, our observations potentially explain the diverse functionality attributed to Tim-3+ NK cells and should be considered prior to use of anti-Tim-3 inhibitory mAbs for cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hepatitis A Virus Cellular Receptor 2/metabolism , Immunoglobulin G/metabolism , Immunotherapy/methods , Killer Cells, Natural/immunology , Neoplasms/therapy , Cells, Cultured , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Humans , Immune Tolerance , Interferon-gamma/metabolism , K562 Cells , Lymphocyte Activation , Neoplasms/immunology , Protein Multimerization , Receptor Aggregation , Receptors, Fc/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Up-Regulation
7.
JCI Insight ; 4(2)2019 Jan 24.
Article in English | MEDLINE | ID: mdl-30674715

ABSTRACT

The antiinflammatory effects of i.v. Ig (IVIG) in the treatment of autoimmune disease are due, in part, to the Fc fragments of Ig aggregates. In order to capitalize on the known antiinflammatory and tolerogenic properties of Ig Fc aggregates, we created a recombinant human IgG1 Fc multimer, GL-2045. In vitro, GL-2045 demonstrated high-avidity binding to Fc receptors, blocked the binding of circulating immune complexes from patients with rheumatoid arthritis to human Fcγ receptors (FcγRs), and inhibited antibody-mediated phagocytosis at log order-lower concentrations than IVIG. In vivo, administration of GL-2045 conferred partial protection against antibody-mediated platelet loss in a murine immune thrombocytopenic purpura (ITP) model. GL-2045 also suppressed disease activity in a therapeutic model of murine collagen-induced arthritis (CIA), which was associated with reduced circulating levels of IL-6. Furthermore, GL-2045 administration to nonhuman primates (NHPs) transiently increased systemic levels of the antiinflammatory cytokines IL-10 and IL-1RA, reduced the proinflammatory cytokine IL-8, and decreased surface expression of CD14 and HLA-DR on monocytes. These findings demonstrate the immunomodulatory properties of GL-2045 and suggest that it has potential as a treatment for autoimmune and inflammatory diseases, as a recombinant alternative to IVIG.

9.
J Autoimmun ; 84: 97-108, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28830653

ABSTRACT

There is a lack of effective targeted therapies for the treatment of complement dependent diseases. We developed two recombinant Fc multimers, G207 and G211, with limited ability to interact with low/moderate affinity FcγRs, but with high avidity for C1q. These drugs effectively inhibited complement dependent cytotoxicity (CDC) in vitro, and prevented the deposition of C1q, C3b and MAC, on the surface of Ab-opsonized cells. Importantly, these inhibitory effects were both C1q dependent and independent. In order to determine the biologic relevance of our findings, we evaluated the clinical efficacy of these drugs in three different animal models, acute RBC hemolysis, anti-Thy-1 nephritis and passive Heymann's nephropathy (PHN), in which disease pathophysiology relies preferentially on complement activation. While G207 was protective in the anti-Thy-1 nephritis and PHN models, G211 was protective in all of the models tested and could effectively treat PHN. In the anti-Thy-1 nephritis model, G211 prevented the characteristic histologic changes associated with the disease and limited glomerular deposition of C3. Collectively, these data suggest that "complement preferential" Fc multimers offer a novel approach to the treatment of complement mediated diseases.


Subject(s)
Complement C1q/immunology , Complement System Proteins/metabolism , Erythrocytes/physiology , Immune System Diseases/therapy , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Animals , Cells, Cultured , Complement C3/metabolism , Complement System Proteins/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Glomerulonephritis, Membranous , Hemolysis , Humans , Immune System Diseases/immunology , Molecular Targeted Therapy , Protein Binding , Protein Multimerization , Receptors, Fc/metabolism , Thy-1 Antigens/immunology , Transgenes/genetics
11.
Blood Adv ; 1(8): 504-515, 2017 Mar 14.
Article in English | MEDLINE | ID: mdl-29296968

ABSTRACT

GL-2045 is a recombinant human immunoglobulin G1 (IgG1)-based Fc multimer designed to recapitulate the anti-inflammatory activities of intravenous immunoglobulin (IVIG) on the innate and adaptive immune responses. We used functional in vitro studies to determine if GL-2045 could mimic the modulatory activity of IVIG on complement activation. GL-2045, at log-order lower concentrations than heat-aggregated IgG (HAGG) and IVIG, protected antibody-opsonized cells from complement-dependent cytotoxicity. These protective effects were completely mediated by the higher order multimer fractions of GL-2045 and were partially dependent upon sequestration of C1q. Exposure of serum to GL-2045 and, to a lesser extent, IVIG, resulted in high levels of C4a, limited levels of C3a, and no C5a. In contrast, HAGG induced high levels of C4a, C3a, and C5a. The means by which GL-2045 governed complement activation was dependent on its ability to augment the function of factor H, alone and in combination with factor I, to indirectly limit the alternative form of C3 convertase, with resultant increases in the anti-inflammatory molecule, the "inactive" form of C3b, called iC3b. Although IVIG, like GL-2045, potentiated factor H function, it also directly inhibited the alternative form of C3 convertase. Our findings help elucidate how IVIG, GL-2045, and HAGG regulate complement function. Furthermore, the capacity of GL-2045 to sequester C1q and augment factor H activity, in combination with its ability to generate activation-induced immunomodulatory complement split products, such as iC3b, make it a viable drug candidate for the treatment of diverse complement-mediated diseases.

12.
Cancer ; 123(7): 1259-1271, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27906454

ABSTRACT

Recent advances have permitted successful therapeutic targeting of the immune system in head and neck squamous cell carcinoma (HNSCC). These new immunotherapeutic targets and agents are being rapidly adopted by the oncologic community and hold considerable promise. The National Cancer Institute sponsored a Clinical Trials Planning Meeting to address the issue of how to further investigate the use of immunotherapy in patients with HNSCC. The goals of the meeting were to consider phase 2 or 3 trial designs primarily in 3 different patient populations: those with previously untreated, human papillomavirus-initiated oropharyngeal cancers; those with previously untreated, human papillomavirus-negative HNSCC; and those with recurrent/metastatic HNSCC. In addition, a separate committee was formed to develop integrative biomarkers for the clinical trials. The meeting started with an overview of key immune components and principles related to HNSCC, including immunosurveillance and immune escape. Four clinical trial concepts were developed at the meeting integrating different immunotherapies with existing standards of care. These designs were presented for implementation by the head and neck committees of the National Cancer Institute-funded National Clinical Trials Network. This article summarizes the proceedings of this Clinical Trials Planning Meeting, the purpose of which was to facilitate the rigorous development and design of randomized phase 2 and 3 immunotherapeutic trials in patients with HNSCC. Although reviews usually are published immediately after the meeting is held, this report is unique because there are now tangible clinical trial designs that have been funded and put into practice and the studies are being activated to accrual. Cancer 2017;123:1259-1271. © 2016 American Cancer Society.


Subject(s)
Head and Neck Neoplasms/therapy , Immunotherapy , Antibodies, Monoclonal/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/etiology , Head and Neck Neoplasms/immunology , Humans , Immunologic Factors/therapeutic use , Immunotherapy/adverse effects , Immunotherapy/methods , Molecular Targeted Therapy , National Cancer Institute (U.S.) , Neoplasm Staging , Patient Selection , Treatment Outcome , United States
13.
Oral Oncol ; 61: 152-8, 2016 10.
Article in English | MEDLINE | ID: mdl-27503244

ABSTRACT

We review the current clinical knowledge surrounding one of the most promising immune checkpoint pathways currently investigated in head and neck squamous cell carcinoma patients, programmed cell death-1 (PD-1) and its ligands (PD-L1 and PD-L2). We review ongoing clinical trials and associated clinical responses observed with targeting the receptor, PD-1, and its ligand, PD-L1. A recent phase III clinical trial (Checkmate 141) demonstrated an improved overall survival in head and neck cancer patients treated with anti-PD-1 monotherapy as compared to standard of care for recurrent and/or metastatic disease, which raises questions on how best to incorporate immunotherapy in the context of standard of care. We discuss biomarkers of response to this class of novel drugs, which is an area of active investigation. Lastly, we project future directions in the field wherein understanding how the Fc portions of the various monoclonal antibodies may impact their clinical efficacy as well as discuss areas where our next advances may take place, such as combination strategies.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Head and Neck Neoplasms/therapy , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Head and Neck Neoplasms/immunology , Humans , Tumor Microenvironment
14.
J Immunol ; 196(3): 1165-76, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26695368

ABSTRACT

We developed a fully recombinant anti-CD20 protein derived from cDNA encoding one Fab domain, two IgG1 Fc regions, the IgG2 hinge, and an isoleucine zipper. This protein, called GB4542, contained both the homodimer and higher-order multimers. Binding studies revealed that GB4542 preferentially bound CD20(+) cells yet also recognized CD20(-)FcγR(+) PBMC. In contrast, a control mAb containing the identical Fab region, GB4500, failed to bind CD20(-)FcγR(+) PBMC. Consistent with these findings, interactions between GB4542 and the canonical FcγRs had substantially lower KD values than correlate interfaces between GB4500 and these receptors. At low concentrations, GB4542 showed enhanced Ab-dependent cellular cytotoxicity, Ab-dependent cellular phagocytosis, and complement-dependent cytotoxicity compared with GB4500. However, at higher concentrations, an Fc analog of GB4542 inhibited anti-CD20 mAb-mediated B cell clearance through direct blocking of both Fc-FcγR interactions and C1q deposition on target cells. Furthermore, the higher-order multimer fraction of GB4542 demonstrated greater binding avidity with the canonical FcγRs and was associated with inhibitory effects observed in Ab-dependent cellular phagocytosis and complement-dependent cytotoxicity assays. These data suggest that GB4542 might have utility in the treatment of autoimmune diseases by combining both mAb-mediated B cell depletion and multimerized Fc-mediated tolerogenic effects.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD20/immunology , B-Lymphocytes/immunology , Immunoglobulin Fc Fragments/immunology , Antibodies, Monoclonal/pharmacology , Antibody Affinity , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/immunology , Autoimmune Diseases/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Receptors, IgG/immunology , Recombinant Proteins/immunology , Surface Plasmon Resonance
15.
Head Neck ; 38(4): 564-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25488341

ABSTRACT

BACKGROUND: Racial outcome disparities have been observed in head and neck squamous cell carcinoma (HNSCC) with diminished survival for black patients compared with white patients. METHODS: We retrospectively analyzed 1318 patients with primary HNSCC treated at the University of Maryland Greenebaum Cancer Center (UMGCC) from 2000 to 2010. RESULTS: Of all the patients, 65.9% were white, 30.7% were black, and 3.3% were of other races. Black patients were less likely to present with oral cavity cancer, and more likely to present with laryngeal or hypopharyngeal cancers. White patients were more likely to have early stage disease, especially in the oral cavity. Black race was independently associated with worse overall survival (OS) in the entire cohort. Black patients had a significantly worse OS among oral cavity and oropharyngeal cancers, with the largest disparity in oropharyngeal cancer. However, in multivariate analysis, race was only still significant in oropharyngeal cancer. CONCLUSION: We observed differences by race in distribution of disease site, stage, and OS. Survival disparity in the entire cohort was driven mostly by differences among oropharyngeal cancer.


Subject(s)
Carcinoma, Squamous Cell/mortality , Head and Neck Neoplasms/mortality , Oropharyngeal Neoplasms/mortality , Adult , Black or African American , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/ethnology , Carcinoma, Squamous Cell/pathology , Female , Head and Neck Neoplasms/ethnology , Head and Neck Neoplasms/pathology , Health Status Disparities , Humans , Male , Maryland , Middle Aged , Oropharyngeal Neoplasms/ethnology , Oropharyngeal Neoplasms/pathology , Racial Groups , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck , Survival Rate , White People , Young Adult
16.
Nat Commun ; 6: 8549, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26469318

ABSTRACT

Sensorineural hearing loss is a common and currently irreversible disorder, because mammalian hair cells (HCs) do not regenerate and current stem cell and gene delivery protocols result only in immature HC-like cells. Importantly, although the transcriptional regulators of embryonic HC development have been described, little is known about the postnatal regulators of maturating HCs. Here we apply a cell type-specific functional genomic analysis to the transcriptomes of auditory and vestibular sensory epithelia from early postnatal mice. We identify RFX transcription factors as essential and evolutionarily conserved regulators of the HC-specific transcriptomes, and detect Rfx1,2,3,5 and 7 in the developing HCs. To understand the role of RFX in hearing, we generate Rfx1/3 conditional knockout mice. We show that these mice are deaf secondary to rapid loss of initially well-formed outer HCs. These data identify an essential role for RFX in hearing and survival of the terminally differentiating outer HCs.


Subject(s)
DNA-Binding Proteins/metabolism , Hair Cells, Auditory/metabolism , Hearing/physiology , Transcription Factors/metabolism , Animals , Animals, Newborn , Biological Evolution , Chromatin Immunoprecipitation , Female , Gene Expression Regulation , Hair Cells, Auditory/ultrastructure , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Multigene Family , Regulatory Factor X Transcription Factors , Regulatory Factor X1 , Sequence Analysis, DNA , Transcriptome , Zebrafish
17.
J Immunol Methods ; 426: 56-61, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26253940

ABSTRACT

The methods of cell detachment influence phenotype and function of human macrophages cultured in vitro. However, comparative studies defining the influence of cell detachment techniques on secondary characterization of M1 or M2 polarized macrophages are largely absent from the literature. In this study we evaluated the impact of trypsin, accutase, EDTA, PBS, and cell scraping on: A. cell recovery, B. phenotype and C. function of in vitro polarized macrophages. Our data demonstrate that while exposure to trypsin or accutase yields highly efficient recovery of viable cells, such chemical cleavage results in loss of select M2 cell surface markers with correlative changes in cell function. In contrast, phenotype and function are maintained following detachment by EDTA on ice. Our data suggest that seemingly "trivial" changes in methodologies for macrophage detachment induce both variable and profound changes on cell phenotype and function which can dramatically impact the results of polarization experiments.


Subject(s)
Cell Separation/methods , Collagenases/metabolism , Edetic Acid/metabolism , Macrophages/immunology , Peptide Hydrolases/metabolism , Trypsin/metabolism , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Biomarkers/metabolism , Cell Culture Techniques , Cells, Cultured , Humans , Lectins, C-Type/biosynthesis , Leukocytes, Mononuclear/immunology , Lipopolysaccharide Receptors/biosynthesis , Mannose Receptor , Mannose-Binding Lectins/biosynthesis , Phenotype , Receptors, Cell Surface/biosynthesis
18.
Cytokine ; 72(1): 48-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25569376

ABSTRACT

Invariant natural killer T (iNKT) cells constitute an important subset of T cells that can both directly and indirectly mediate anti-tumor immunity. However, cancer patients have a reduction in both iNKT cell number and function, and these deficits limit the potential clinical application of iNKT cells for cancer therapy. To overcome the problem of limited iNKT cell numbers, we investigated whether iNKT cells can be generated in vitro from bone marrow-derived adult hematopoietic stem-progenitor cells (HSPC). Our data demonstrate that co-culture of HSPC with OP9-DL1 stromal cells, results in a functional CD3(+) T cell population. These T cells can be further differentiated into iNKT cells by secondary culture with CD1d-Ig-based artificial antigen-presenting cells (aAPC). Importantly, these in vitro-generated iNKT cells are functional, as demonstrated by their ability to proliferate and secrete IFN-γ and GM-CSF following stimulation.


Subject(s)
Hematopoietic Stem Cells/physiology , Lymphocyte Activation , Natural Killer T-Cells/immunology , Adult , Antigen-Presenting Cells/immunology , Antigens, CD1d/immunology , Cell Differentiation , Coculture Techniques , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/cytology , Humans , Interferon-gamma/metabolism , Natural Killer T-Cells/cytology , Stromal Cells/immunology
19.
Trends Biotechnol ; 33(1): 27-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25488117

ABSTRACT

The platform technology of fragment crystallizable (Fc) fusion, in which the Fc region of an antibody is genetically linked to an active protein drug, is among the most successful of a new generation of bioengineering strategies. Immunogenicity is a critical safety concern in the development of any protein therapeutic. While the therapeutic goal of generating Fc-fusion proteins has been to extend half-life, there is a critical mass of literature from immunology indicating that appropriate design of the Fc component has the potential to engage the immune system for product-specific outcomes. In the context of Fc-fusion therapeutics, a review of progress in understanding Fc biology suggests the prospect of engineering products that have an extended half-life and are able to modulate the immune system.


Subject(s)
Antibodies, Monoclonal/chemistry , Protein Engineering/methods , Autoimmune Diseases/drug therapy , Drug Delivery Systems , Humans , Immunoglobulin Fc Fragments/therapeutic use , Immunomodulation/drug effects , Models, Immunological , Recombinant Fusion Proteins/therapeutic use
20.
Cancer Immunol Immunother ; 64(3): 367-79, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25537079

ABSTRACT

BACKGROUND: We conducted a phase I dose escalation study to evaluate the safety and immunologic response to peptide immunomodulatory vaccines GL-0810 (HPV16) and GL-0817 (MAGE-A3) in HPV16 and MAGE-A3-positive RM-SCCHN patients, respectively. METHODS: Three dose levels (500, 1,000, and 1,500 µg) of GL-0810 or GL-0817 with adjuvants Montanide (1.2 ml) and GM-CSF (100 µg/m2) were administered subcutaneously q2 weeks for a total of four vaccinations in HPV16 and MAGE-A3-positive RM-SCCHN patients, respectively. RESULTS: Nine and seven patients were enrolled in the HPV16 and MAGE-A3 cohorts, respectively. No dose-limiting toxicities were observed, and toxicity was predominantly local and grade 1 (erythema, pain, and itching at the injection site). In those patients who received all four vaccinations, 80 % (4/5) of the HPV16 cohort and 67 % (4/6) of the MAGE-A3 cohort developed antigen-specific T cell and antibody responses to the vaccine. Significant concordance between T cell and antibody responses was observed for both groups. No clear dose-response correlation was seen. All patients progressed by RECIST at first repeat imaging, except for one patient in the MAGE-A3 500 µg cohort who had stable disease for 10.5 months. The median PFS and OS for the MAGE-A3 cohorts were 79 and 183 days, respectively, and for the HPV16 cohort 80 and 196 days, respectively. CONCLUSIONS: GL-0810 and GL-0817 were well tolerated in patients with RM-SCCHN with T cell and antibody responses observed in the majority of patients who received all four vaccinations.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Carcinoma, Squamous Cell/therapy , Head and Neck Neoplasms/therapy , Human papillomavirus 16/immunology , Immunologic Factors/administration & dosage , Neoplasm Proteins/immunology , Vaccines, Subunit/administration & dosage , Adult , Aged , Cancer Vaccines/immunology , Carcinoma, Squamous Cell/immunology , Cohort Studies , Disease Progression , Dose-Response Relationship, Immunologic , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Head and Neck Neoplasms/immunology , Humans , Immunologic Factors/immunology , Male , Middle Aged , Squamous Cell Carcinoma of Head and Neck , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...