Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cancer Immunol Immunother ; 71(1): 97-109, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34028568

ABSTRACT

Cyclophosphamide plus fludarabine (C/F) are currently used to improve the expansion and effectiveness of adoptive cell therapy (ACT). However, these chemotherapeutics cause pan-leukopenia and adverse events, suggesting that safer and more effective conditioning treatments are needed to improve ACT outcomes. Previously, we reported that varlilumab, a CD27-targeting antibody, mediates Treg -preferential T cell depletion, CD8-T cell dominant costimulation, and systemic immune activation in hCD27 transgenic mice and cancer patients. We reasoned that the activities induced by varlilumab may provide an effective conditioning regimen for ACT. Varlilumab pretreatment of hCD27 +/+mCD27 - /- mice resulted in prominent proliferation of transferred T cells isolated from wild-type mice. These studies uncovered a critical role for CD27 signaling for the expansion of transferred T cells, as transfer of T cells from CD27 deficient mice or treatment with a CD70 blocking antibody greatly reduced their proliferation. In this model, varlilumab depletes endogenous hCD27+/+ T cells and blocks their subsequent access to CD70, allowing for more CD70 costimulation available to the mCD27 +/+ transferred T cells. CD27-targeted depletion led to a greater expansion of transferred T cells compared to C/F conditioning and resulted in longer median survival and more cures than C/F conditioning in the E.G7 tumor model receiving OT-I cell therapy. We propose that translation of this work could be achieved through engineering of T cells for ACT to abrogate varlilumab binding but preserve CD70 ligation. Thus, varlilumab could be an option to chemotherapy as a conditioning regimen for ACT.


Subject(s)
Adoptive Transfer , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/chemistry , Neoplasms/therapy , T-Lymphocytes/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , Animals , CD27 Ligand/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Proliferation , Immune System , Immunotherapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/metabolism , Signal Transduction , Transplantation Conditioning , Treatment Outcome
2.
Cancer Immunol Immunother ; 69(10): 2125-2137, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32451681

ABSTRACT

CD27 is a costimulatory molecule that provides a complementary target to the PD-1/PD-L1 checkpoint axis on T cells. Combining a CD27 agonist antibody with PD-1/PD-L1 blockade has shown synergistic antitumor activity in preclinical models, which led to clinical studies of the combination in cancer patients. We theorized that coupling CD27 costimulation with PD-1/PD-L1 blockade in a bispecific antibody (BsAb) may provide greater immune activating properties than combining the individual mAbs due to enhanced CD27 activation by cross-linking through PD-L1 and Fc receptors. To test this approach, we developed CDX-527, a tetravalent PD-L1xCD27 IgG1-scFv BsAb. CDX-527 potently inhibits PD-1 signaling and induces CD27-mediated T cell costimulation through PD-L1 cross-linking. In mixed lymphocyte reaction assays, CDX-527 is more potent than the combination of the parental antibodies, suggesting that cross-linking through both Fc receptors and PD-L1 results in enhanced CD27 agonist activity. CDX-527 was shown to mediate effector function against tumor cells overexpressing either CD27 or PD-L1. In human CD27 transgenic mice, we observed that antigen-specific T cell responses to a vaccine are greatly enhanced with a surrogate PD-L1xCD27 BsAb. Furthermore, the BsAb exhibits greater antitumor activity than the combination of the parental antibodies in a syngeneic lymphoma model. A pilot study of CDX-527 in cynomolgus macaques confirmed a mAb-like pharmacokinetic profile without noted toxicities. These studies demonstrate that CDX-527 effectively combines PD-1 blockade and CD27 costimulation into one molecule that is more potent than combination of the parental antibodies providing the rationale to advance this BsAb toward clinical studies in cancer patients.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibody Formation , Immunotherapy/methods , Lymphoma, B-Cell/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors , Animals , Antibodies, Bispecific/chemistry , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Macaca fascicularis , Male , Mice , Mice, Transgenic
3.
Cancer Immunol Immunother ; 68(2): 233-245, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30382327

ABSTRACT

Limitations of immunotherapy include poorly functioning events early in the immune response cycle, such as efficient antigen presentation and T cell priming. CD40 signaling in dendritic cells leads to upregulation of cell surface costimulatory and MHC molecules and the generation of cytokines, which promotes effective priming of CD8+ effector T cells while minimizing T cell anergy and the generation of regulatory T cells. This naturally occurs through interaction with CD40 ligand (CD40L) expressed on CD4+ T-helper cells. CD40 signaling can also be achieved using specific antibodies, leading to several agonist CD40 antibodies entering clinical development. Our approach to select a CD40 agonist antibody was to define a balanced profile between sufficiently strong immune stimulation and the untoward effects of systemic immune activation. CDX-1140 is a human IgG2 antibody that activates DCs and B cells and drives NFkB stimulation in a CD40-expressing reporter cell line. These activities are Fc-independent and are maintained using an F(ab')2 fragment of the antibody. CDX-1140 binds outside of the CD40L binding site, and addition of recombinant CD40L greatly enhances DC and B activation by CDX-1140, suggesting that CDX-1140 may act synergistically with naturally expressed CD40L. CDX-1140 also has both direct and immune-mediated anti-tumor activity in xenograft models. CDX-1140 does not promote cytokine production in whole blood assays and has good pharmacodynamic and safety profiles in cynomolgus macaques. These data support the potential of CDX-1140 as part of a cancer therapy regimen, and a phase 1 trial has recently commenced.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/agonists , Immunotherapy/methods , Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , CD40 Antigens/immunology , CD40 Antigens/metabolism , CD40 Ligand/immunology , CD40 Ligand/metabolism , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HEK293 Cells , Humans , Macaca fascicularis , Mice, SCID , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
4.
J Immunol ; 199(12): 4110-4123, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29109120

ABSTRACT

CD27, a member of the TNFR superfamily, is constitutively expressed in most T cells and plays crucial roles in T cell effector functions. The costimulation and antitumor activity of CD27 agonistic Abs have been well documented in mouse models. Clinical testing of a human IgG1 anti-CD27 Ab, varlilumab (clone 1F5), is ongoing in cancer patients. In this study, we set out to further understand CD27 as an immunomodulatory target and to address the mechanism of antitumor efficacy using different IgG isotypes of 1F5 in human CD27-transgenic mice. 1F5mIgG1, the only isotype engaging inhibitory FcγRIIB expressed in B cells, elicited the most potent and broad immune response, but terminal differentiation, exhaustion, and apoptosis in the activated effector T cells were inevitable. Accordingly, this isotype was the most effective in eradicating BCL1 lymphoma but had limited efficacy in s.c. tumors. Conversely, 1F5mIgG2a, which interacts with cells expressing activating FcγRs, led to moderate immune activation, as well as to prominent reduction in the number and suppressive activity of regulatory T cells. These combined mechanisms imparted potent antitumor activity to 1F5mIgG2a, particularly against the s.c. tumors. 1F5hIgG1, varlilumab, showed balanced agonistic activity that was prominent at lower doses and depleting activity that was greater at higher doses. 1F5hIgG1 had good antitumor activity in all tumor models tested. Thus, both agonist and depleting properties contribute to the antitumor efficacy of CD27-targeted immunotherapy, and modulation of these activities in patients may be achieved by varying the dose and regimen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Lymphocyte Depletion , Neoplasms, Experimental/drug therapy , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological/immunology , Apoptosis , CD27 Ligand/immunology , Drug Screening Assays, Antitumor , Female , Humans , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/therapeutic use , Immunologic Memory , Immunotherapy , Lymphoma, B-Cell/drug therapy , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mutation, Missense , Receptors, IgG/immunology , Receptors, IgG/metabolism , Specific Pathogen-Free Organisms , Tumor Microenvironment , Tumor Necrosis Factor Receptor Superfamily, Member 7/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 7/antagonists & inhibitors
5.
J Immunol ; 191(8): 4174-83, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24026078

ABSTRACT

The CD70/CD27 pathway plays a significant role in the control of immunity and tolerance, and previous studies demonstrated that targeting murine CD27 (mCD27) with agonist mAbs can mediate antitumor efficacy. We sought to exploit the potential of this pathway for immunotherapy by developing 1F5, a fully human IgG1 mAb to human CD27 (hCD27) with agonist activity. We developed transgenic mice expressing hCD27 under control of its native promoter for in vivo testing of the Ab. The expression and regulation of hCD27 in hCD27-transgenic (hCD27-Tg) mice were consistent with the understood biology of CD27 in humans. In vitro, 1F5 effectively induced proliferation and cytokine production from hCD27-Tg-derived T cells when combined with TCR stimulation. Administration of 1F5 to hCD27-Tg mice enhanced Ag-specific CD8(+) T cell responses to protein vaccination comparably to an agonist anti-mCD27 mAb. In syngeneic mouse tumor models, 1F5 showed potent antitumor efficacy and induction of protective immunity, which was dependent on CD4(+) and CD8(+) T cells. The requirement of FcR engagement for the agonistic and antitumor activities of 1F5 was demonstrated using an aglycosylated version of the 1F5 mAb. These data with regard to the targeting of hCD27 are consistent with previous reports on targeting mCD27 and provide a rationale for the clinical development of the 1F5 mAb, for which studies in advanced cancer patients have been initiated under the name CDX-1127.


Subject(s)
Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/therapy , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Cell Proliferation , Humans , Immunoglobulin G/immunology , Immunotherapy , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/agonists , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics
6.
J Immunol ; 186(2): 1218-27, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21149605

ABSTRACT

Immunization of cancer patients with vaccines containing full-length tumor Ags aims to elicit specific Abs and both CD4(+) and CD8(+) T cells. Vaccination with protein Ags, however, often elicits only CD4(+) T cell responses without inducing Ag-specific CD8(+) T cells, as exogenous protein is primarily presented to CD4(+) T cells. Recent data revealed that Ab-mediated targeting of protein Ags to cell surface receptors on dendritic cells could enhance the induction of both CD4(+) and CD8(+) T cells. We investigated in this study if these observations were applicable to NY-ESO-1, a cancer-testis Ag widely used in clinical cancer vaccine trials. We generated two novel targeting proteins consisting of the full-length NY-ESO-1 fused to the C terminus of two human mAbs against the human mannose receptor and DEC-205, both internalizing molecules expressed on APC. These targeting proteins were evaluated for their ability to activate NY-ESO-1-specific human CD4(+) and CD8(+) T cells in vitro. Both targeted NY-ESO-1 proteins rapidly bound to their respective targets on APC. Whereas nontargeted and Ab-targeted NY-ESO-1 proteins similarly activated CD4(+) T cells, cross-presentation to CD8(+) T cells was only efficiently induced by targeted NY-ESO-1. In addition, both mannose receptor and DEC-205 targeting elicited specific CD4(+) and CD8(+) T cells from PBLs of cancer patients. Receptor-specific delivery of NY-ESO-1 to APC appears to be a promising vaccination strategy to efficiently generate integrated and broad Ag-specific immune responses against NY-ESO-1 in cancer patients.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Membrane Proteins/immunology , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Clone Cells , Cross-Priming/genetics , Cross-Priming/immunology , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Minor Histocompatibility Antigens , Molecular Sequence Data , Protein Binding/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism
7.
Breast Cancer Res ; 14(2): R39, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22397502

ABSTRACT

INTRODUCTION: Given their relative simplicity of manufacture and ability to be injected repeatedly, vaccines in a protein format are attractive for breast and other cancers. However, soluble human epidermal growth factor receptor (HER2)/neu protein as a vaccine has not been immunogenic. When protein is directly targeted to antigen uptake receptors, such as DEC205 (DEC), efficient processing and presentation of antigen take place. The aim of this study was to determine the immunogenicity of a HER2 protein vaccine that directly targets to DEC+ dendritic cells (DCs) in a mouse breast cancer model. METHODS: We genetically engineered the HER2 extracellular domain into a monoclonal antibody specific for DEC (DEC-HER2). Mice of various genetic backgrounds were immunized with DEC-HER2 in combination with DC maturation stimuli (poly IC ± CD40 Ab). Vaccine-induced T cell immunity was determined by analyzing the ability of CD4+/CD8+ T cell to produce interferon (IFN)-gamma and proliferate upon antigen rechallenge. Sera were assessed for the presence of antigen specific antibody (Ab). For vaccine efficacy, FVB/N mice were immunized with DEC-HER2 in combination with poly IC and protection against neu-expressing mammary tumors was assessed. Protection mechanisms and tumor-specific T cell responses were also evaluated. RESULTS: We demonstrate that DEC-HER2 fusion mAb, but not Ctrl Ig-HER2, elicits strong, broad and multifunctional CD4+ T cell immunity, CD8+ T cell responses, and humoral immunity specific for HER2 antigen. Cross-reactivity to rat neu protein was also observed. Importantly, mice xeno-primed with DEC-HER2 were protected from a neu-expressing mammary tumor challenge. Both CD4+ and CD8+ T cells mediated the tumor protection. Robust anti-tumor T cell immunity was detected in tumor protected mice. CONCLUSIONS: Immunization of mice with HER2 protein vaccine targeting DEC+ DCs in vivo induced high levels of T- and B-cell immunity. Non-targeted HER2 protein was poorly immunogenic for CD4+ and CD8+ T cells. This vaccination approach provided long-term survival benefit for mice challenged with neu-expressing tumor following as little as 2.7 µg of HER2 protein incorporated in the vaccine. Vaccine-induced CD4+ and CD8+ T cells were both essential for tumor protection. This immunization strategy demonstrates great potential towards the development of vaccines for breast cancer patients.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/genetics , Cell Line, Tumor , Female , Humans , Immunity, Humoral , Interferon-gamma/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Poly I-C/immunology , Poly I-C/pharmacology , Protein Structure, Tertiary/genetics , Receptor, ErbB-2/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes/immunology
8.
Blood ; 116(19): 3828-38, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20668230

ABSTRACT

Protein vaccines for T-cell immunity are not being prioritized because of poor immunogenicity. To overcome this hurdle, proteins are being targeted to maturing dendritic cells (DCs) within monoclonal antibodies (mAbs) to DC receptors. To extend the concept to humans, we immunized human immunoglobulin-expressing mice with human DEC205 (hDEC205) extracellular domain. 3D6 and 3G9 mAbs were selected for high-affinity binding to hDEC205. In addition, CD11c promoter hDEC205 transgenic mice were generated, and 3G9 was selectively targeted to DCs in these animals. When mAb heavy chain was engineered to express HIV Gag p24, the fusion mAb induced interferon-γ- and interleukin-2-producing CD4(+) T cells in hDEC205 transgenic mice, if polynocinic polycytidylic acid was coadministered as an adjuvant. The T-cell response was broad, recognizing at least 3 Gag peptides, and high titers of anti-human immunoglobulin G antibody were made. Anti-hDEC205 also improved the cross-presentation of Gag to primed CD8(+) T cells from HIV-infected individuals. In all tests, 3D6 and 3G9 targeting greatly enhanced immunization relative to nonbinding control mAb. These results provide preclinical evidence that in vivo hDEC205 targeting increases the efficiency with which proteins elicit specific immunity, setting the stage for proof-of-concept studies of these new protein vaccines in human subjects.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , HIV Core Protein p24/immunology , HIV-1/immunology , Lectins, C-Type/immunology , Receptors, Cell Surface/immunology , AIDS Vaccines/genetics , AIDS Vaccines/immunology , Animals , Antibodies, Monoclonal , Antigens, CD/genetics , Base Sequence , Cross-Priming , DNA Primers/genetics , HIV Core Protein p24/genetics , HIV-1/genetics , Humans , Immunity, Cellular , Immunity, Humoral , Lectins, C-Type/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens , Receptors, Cell Surface/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
9.
Elife ; 112022 08 15.
Article in English | MEDLINE | ID: mdl-35969037

ABSTRACT

Knockout (KO) mouse models play critical roles in elucidating biological processes behind disease-associated or disease-resistant traits. As a presumed consequence of gene KO, mice display certain phenotypes. Based on insight into the molecular role of said gene in a biological process, it is inferred that the particular biological process causally underlies the trait. This approach has been crucial towards understanding the basis of pathological and/or advantageous traits associated with Mertk KO mice. Mertk KO mice suffer from severe, early-onset retinal degeneration. MERTK, expressed in retinal pigment epithelia, is a receptor tyrosine kinase with a critical role in phagocytosis of apoptotic cells or cellular debris. Therefore, early-onset, severe retinal degeneration was described to be a direct consequence of failed MERTK-mediated phagocytosis of photoreceptor outer segments by retinal pigment epithelia. Here, we report that the loss of Mertk alone is not sufficient for retinal degeneration. The widely used Mertk KO mouse carries multiple coincidental changes in its genome that affect the expression of a number of genes, including the Mertk paralog Tyro3. Retinal degeneration manifests only when the function of Tyro3 is concomitantly lost. Furthermore, Mertk KO mice display improved anti-tumor immunity. MERTK is expressed in macrophages. Therefore, enhanced anti-tumor immunity was inferred to result from the failure of macrophages to dispose of cancer cell corpses, resulting in a pro-inflammatory tumor microenvironment. The resistance against two syngeneic mouse tumor models observed in Mertk KO mice is not, however, phenocopied by the loss of Mertk alone. Neither Tyro3 nor macrophage phagocytosis by alternate genetic redundancy accounts for the absence of anti-tumor immunity. Collectively, our results indicate that context-dependent epistasis of independent modifier alleles determines Mertk KO traits.


Subject(s)
Retinal Degeneration , Alleles , Animals , Disease Models, Animal , Mice , Mice, Knockout , Phagocytosis/genetics , Phenotype , Proto-Oncogene Proteins/genetics , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Pigments , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism
10.
Front Immunol ; 12: 741268, 2021.
Article in English | MEDLINE | ID: mdl-34745113

ABSTRACT

Objective: We aimed to evaluate the diagnostic value of soluble interleukin-2 receptor (sIL-2R), tumor necrosis factor-α (TNF-α), procalcitonin (PCT), and combined detection for sepsis infection in patients with closed abdominal injury complicated with severe multiple abdominal injuries. Patients and Methods: One hundred forty patients with closed abdominal injury complicated with severe multiple abdominal injuries who were diagnosed and treated from 2015 to 2020 were divided into a sepsis group (n = 70) and an infection group (n = 70). Results: The levels of sIL-2R, TNF-α, and PCT in the sepsis group were higher than those in the infection group (p < 0.05). The receiver operating characteristic (ROC) curve showed that the areas under the ROC curve (AUCs) of sIL-2R, TNF-α, PCT and sIL-2R+TNF-a+PCT were 0.827, 0.781, 0.821, and 0.846, respectively, which were higher than those of white blood cells (WBC, 0.712), C-reactive protein (CRP, 0.766), serum amyloid A (SAA, 0.666), and IL-6 (0.735). The AUC of the three combined tests was higher than that of TNF-α, and the difference was statistically significant (p < 0.05). There was no significant difference in the AUCs of sIL-2R and TNF-α, sIL-2R and PCT, TNF-α and PCT, the three combined tests and sIL-2R, and the three combined tests and PCT (p > 0.05). When the median was used as the cut point, the corrected sIL-2R, TNF-α, and PCT of the high-level group were not better than those of the low-level group (p > 0.05). When the four groups were classified by using quantile as the cut point, the OR risk values of high levels of TNF-α and PCT (Q4) and the low level of PCT (Q1) after correction were 7.991 and 21.76, respectively, with statistical significance (p < 0.05). Conclusions: The detection of sIL-2R, TNF-α, and PCT has good value in the diagnosis of sepsis infection in patients with closed abdominal injury complicated with severe multiple abdominal injuries. The high concentrations of PCT and TNF-α can be used as predictors of the risk of septic infection.


Subject(s)
Abdominal Injuries/diagnosis , Multiple Trauma/diagnosis , Procalcitonin/blood , Receptors, Interleukin-2/blood , Sepsis/diagnosis , Tumor Necrosis Factor-alpha/blood , Abdominal Injuries/complications , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Multiple Trauma/complications , Predictive Value of Tests , Prognosis , Risk , Sepsis/etiology
11.
Commun Biol ; 3(1): 375, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665635

ABSTRACT

Regulatory T cells (Tregs) are critical mediators of immune homeostasis. The co-stimulatory molecule CD27 is a marker of highly suppressive Tregs, although the role of the CD27-CD70 receptor-ligand interaction in Tregs is not clear. Here we show that after prolonged in vitro stimulation, a significant proportion of human Tregs gain stable CD70 expression while losing CD27. The expression of CD70 in expanded Tregs is associated with a profound loss of regulatory function and an unusual ability to provide CD70-directed co-stimulation to TCR-activated conventional T cells. Genetic deletion of CD70 or its blockade prevents Tregs from delivering this co-stimulatory signal, thus maintaining their regulatory activity. High resolution targeted single-cell RNA sequencing of human peripheral blood confirms the presence of CD27-CD70+ Treg cells. These findings have important implications for Treg-based clinical studies where cells are expanded over extended periods in order to achieve sufficient treatment doses.


Subject(s)
CD27 Ligand/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD27 Ligand/genetics , CRISPR-Associated Protein 9 , CRISPR-Cas Systems , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Gene Editing , Gene Knockout Techniques , Humans , Mice, Inbred BALB C , Sequence Analysis, RNA , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Transcriptome , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
12.
Immunohorizons ; 3(6): 236-253, 2019 06 26.
Article in English | MEDLINE | ID: mdl-31356169

ABSTRACT

Dendritic cells (DCs) are crucial for the production of adaptive immune responses to disease-causing microbes. However, in the steady state (i.e., in the absence of an infection or when Ags are experimentally delivered without a DC-activating adjuvant), DCs present Ags to T cells in a tolerogenic manner and are important for the establishment of peripheral tolerance. Delivery of islet Ags to DCs using Ag-linked Abs to the DC endocytic receptor CD205 has shown promise in the NOD mouse model of type 1 diabetes (T1D). It is important to note, however, that all myeloid DCs express CD205 in humans, whereas in mice, only one of the classical DC subsets does (classical DC1; CD8α+ in spleen). Thus, the evaluation of CD205-targeted treatments in mice will likely not accurately predict the results observed in humans. To overcome this challenge, we have developed and characterized a novel NOD mouse model in which all myeloid DCs transgenically express human CD205 (hCD205). This NOD.hCD205 strain displays a similar T1D incidence profile to standard NOD mice. The presence of the transgene does not alter DC development, phenotype, or function. Importantly, the DCs are able to process and present Ags delivered via hCD205. Because Ags taken up via hCD205 can be presented on both class I and class II MHC, both CD4+ and CD8+ T cells can be modulated. As both T cell subsets are important for T1D pathogenesis, NOD.hCD205 mice represent a unique, patient-relevant tool for the development and optimization of DC-directed T1D therapies.


Subject(s)
Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Type 1/immunology , Immunotherapy/methods , Lectins, C-Type/metabolism , Minor Histocompatibility Antigens/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigen Presentation , Antigens, CD/genetics , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Immune Tolerance , Lectins, C-Type/genetics , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Receptors, Cell Surface/genetics
13.
Clin Cancer Res ; 24(10): 2383-2394, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29514845

ABSTRACT

Purpose: PD-1 checkpoint blockade has revolutionized the field of cancer immunotherapy, yet the frequency of responding patients is limited by inadequate T-cell priming secondary to a paucity of activatory dendritic cells (DC). DC signals can be bypassed by CD27 agonists, and we therefore investigated if the effectiveness of anti-PD-1/L1 could be improved by combining with agonist anti-CD27 monoclonal antibodies (mAb).Experimental Design: The efficacy of PD-1/L1 blockade or agonist anti-CD27 mAb was compared with a dual-therapy approach in multiple tumor models. Global transcriptional profiling and flow cytometry analysis were used to delineate mechanisms underpinning the observed synergy.Results: PD-1/PD-L1 blockade and agonist anti-CD27 mAb synergize for increased CD8+ T-cell expansion and effector function, exemplified by enhanced IFNγ, TNFα, granzyme B, and T-bet. Transcriptome analysis of CD8+ T cells revealed that combination therapy triggered a convergent program largely driven by IL2 and Myc. However, division of labor was also apparent such that anti-PD-1/L1 activates a cytotoxicity-gene expression program whereas anti-CD27 preferentially augments proliferation. In tumor models, either dependent on endogenous CD8+ T cells or adoptive transfer of transgenic T cells, anti-CD27 mAb synergized with PD-1/L1 blockade for antitumor immunity. Finally, we show that a clinically relevant anti-human CD27 mAb, varlilumab, similarly synergizes with PD-L1 blockade for protection against lymphoma in human-CD27 transgenic mice.Conclusions: Our findings suggest that suboptimal T-cell invigoration in cancer patients undergoing treatment with PD-1 checkpoint blockers will be improved by dual PD-1 blockade and CD27 agonism and provide mechanistic insight into how these approaches cooperate for CD8+ T-cell activation. Clin Cancer Res; 24(10); 2383-94. ©2018 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/genetics , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Transcriptome , Tumor Necrosis Factor Receptor Superfamily, Member 7/agonists , Adoptive Transfer , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Cytotoxicity, Immunologic , Disease Models, Animal , Gene Expression Profiling , Humans , Immunomodulation/drug effects , Immunomodulation/genetics , Immunomodulation/immunology , Lymphocyte Activation/immunology , Melanoma, Experimental , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/metabolism
14.
Oncoimmunology ; 7(12): e1502904, 2018.
Article in English | MEDLINE | ID: mdl-30524899

ABSTRACT

Despite their promise, tumor-specific peptide vaccines have limited efficacy. CD27 is a costimulatory molecule expressed on CD4+ and CD8+ T cells that is important in immune activation. Here we determine if a novel CD27 agonist antibody (αhCD27) can enhance the antitumor T cell response and efficacy of peptide vaccines. We evaluated the effects of αhCD27 on the immunogenicity and antitumor efficacy of whole protein, class I-restricted, and class II-restricted peptide vaccines using a transgenic mouse expressing human CD27. We found that αhCD27 preferentially enhances the CD8+ T cell response in the setting of vaccines comprised of linked class I and II ovalbumin epitopes (SIINFEKL and TEWTSSNVMEERKIKV, respectively) compared to a peptide vaccine comprised solely of SIINFEKL, resulting in the antitumor efficacy of adjuvant αhCD27 against intracranial B16.OVA tumors when combined with vaccines containing linked class I/II ovalbumin epitopes. Indeed, we demonstrate that this efficacy is both CD8- and CD4-dependent and αhCD27 activity on ovalbumin-specific CD4+ T cells is necessary for its adjuvant effect. Importantly for clinical translation, a linked universal CD4+ helper epitope (tetanus P30) was sufficient to instill the efficacy of SIINFEKL peptide combined with αhCD27, eliminating the need for a tumor-specific class II-restricted peptide. This approach unveiled the efficacy of a class I-restricted peptide vaccine derived from the tumor-associated Trp2 antigen in mice bearing intracranial B16 tumors. CD27 agonist antibodies combined with peptide vaccines containing linked tumor-specific CD8+ epitopes and tumor-specific or universal CD4+ epitopes enhance the efficacy of active cancer immunotherapy.

15.
Zhonghua Zhong Liu Za Zhi ; 29(4): 245-8, 2007 Apr.
Article in Zh | MEDLINE | ID: mdl-17760247

ABSTRACT

OBJECTIVE: To investigate the biological effects of PTEN gene on the malignant glioma cell line SHG-44. Firstly, A recombinant eukaryotic expression plasmid containing PTEN gene fused with EGFP (enhanced green fluorescence protein) gene was constructed. Secondly, the expression of the recombinant vector in human glioma cells was detected. METHODS: (1) The human PTEN gene was amplified by RT-PCR and inserted into pEGFP-N1 that was selected by T-A subclone, and the recombinant expression vector was obtained. After the recombinant plasmids were transfected into glioma SHG-44 cells by cation polymex, expression of fusion protein was tested. (2) The stable transfected cells were selected by G418 and amplified. Light microscopy and growth curve were used to measure the effects of PTEN expression on cell morphology and proliferation. Expression of GFAP (glial fibillary acidic protein) was detected immunohistochemically. RESULTS: (1) The positive recombinant was sequenced and demonstrated to have the same sequence as that of PTEN gene in GenBank. It was proved that the eukaryotic expression vector pEGFP-PTEN have been constructed successfully and expressed in SHG-44 cells. (2) The SHG-44 cell growth was changed obviously. The expression of GFAP was increased. CONCLUSION: The construction of PTEN eukaryotic expression vector containing green fluorescence protein gene will lay the basis for carrying out further studies on the function of PTEN gene.


Subject(s)
Cell Proliferation , Glial Fibrillary Acidic Protein/metabolism , PTEN Phosphohydrolase/physiology , Recombinant Fusion Proteins/physiology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Flow Cytometry , Gene Expression , Genetic Vectors , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
Medicine (Baltimore) ; 96(33): e7807, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28816976

ABSTRACT

The aim of this study was to evaluate prognostic values of ambulatory blood pressure (ABP) and clinic blood pressure (CBP) in diabetic patients with hypertension. A total of 450 diabetic hypertensive patients without established cardiovascular diseases were enrolled and 416 patients who had finished 12months' follow-up were included in final analysis. Baseline data were collected and Cox proportional hazards regression analysis was used to evaluate prognostic value of ABP and CBP. Compared to those without study endpoints (n = 370), those experienced study endpoints (n = 46) were more elderly and more likely to be male, and had longer hypertension duration (7.0 ±â€Š3.0 years vs. 6.4 ±â€Š2.1 years, P < .05). No significant between-group differences in CBP indices were observed. However, those with study endpoints had significantly higher 24-hour systolic BP (SBP) (134 ±â€Š10 mmHg vs. 128 ±â€Š7 mmHg), nighttime SBP (130 ±â€Š7 mmHg vs. 120 ±â€Š5 mmHg), night/day SBP ratio (0.97 ±â€Š0.09 vs. 0.94 ±â€Š0.08), higher proportion of non-dipping BP pattern (39.1% vs. 31.4%) and higher 24-hour SBP variability. After extensively adjusted for traditional risk factors, nondipping BP pattern and 24-hourSBP, only 24-hour SBP and nighttime SBP remained independently related with cardiovascular outcomes, with hazard ratios and associated 95% confidence interval as 1.53 (1.28-2.03) and 1.50 (1.26-1.89), respectively. Although no independent relationship between BP pattern and cardiovascular outcomes was observed. In summary, in diabetic hypertensive patients without established cardiovascular diseases, baseline 24-hour SBP and nighttime SBP are useful markers for predicting short-term cardiovascular outcomes.


Subject(s)
Blood Pressure/physiology , Diabetes Mellitus/epidemiology , Diabetes Mellitus/physiopathology , Hypertension/epidemiology , Hypertension/physiopathology , Age Factors , Aged , Ambulatory Care Facilities , Blood Pressure Monitoring, Ambulatory/methods , Blood Pressure Monitoring, Ambulatory/standards , Female , Humans , Male , Middle Aged , Proportional Hazards Models , Regression Analysis , Sex Factors
17.
Cell Rep ; 21(1): 126-140, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978467

ABSTRACT

Despite its prominent role as a C-type lectin (CTL) pattern recognition receptor, mannose receptor (MR, CD206)-specific signaling molecules and pathways are unknown. The MR is highly expressed on human macrophages, regulating endocytosis, phagocytosis, and immune responses and mediating Mycobacterium tuberculosis (M.tb) phagocytosis by human macrophages, thereby limiting phagosome-lysosome (P-L) fusion. We identified human MR-associated proteins using phosphorylated and non-phosphorylated MR cytoplasmic tail peptides. We found that MR binds FcRγ-chain, which is required for MR plasma membrane localization and M.tb cell association. Additionally, we discovered that MR-mediated M.tb association triggers immediate MR tyrosine residue phosphorylation and Grb2 recruitment, activating the Rac/Pak/Cdc-42 signaling cascade important for M.tb uptake. MR activation subsequently recruits SHP-1 to the M.tb-containing phagosome, where its activity limits PI(3)P generation at the phagosome and M.tb P-L fusion and promotes M.tb growth. In sum, we identify human MR signaling pathways that temporally regulate phagocytosis and P-L fusion during M.tb infection.


Subject(s)
GRB2 Adaptor Protein/genetics , Host-Pathogen Interactions , Lectins, C-Type/genetics , Macrophages/metabolism , Mannose-Binding Lectins/genetics , Mycobacterium tuberculosis/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Receptors, Cell Surface/genetics , Receptors, IgG/genetics , GRB2 Adaptor Protein/metabolism , Gene Expression Regulation , Humans , Lectins, C-Type/metabolism , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/microbiology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Membrane Fusion , Mycobacterium tuberculosis/growth & development , Phagocytosis/genetics , Phagosomes/metabolism , Phagosomes/microbiology , Phosphorylation , Primary Cell Culture , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Cell Surface/metabolism , Receptors, IgG/metabolism , Signal Transduction , Transport Vesicles/metabolism , Transport Vesicles/microbiology , Tyrosine/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
18.
Cancer Cell ; 32(6): 777-791.e6, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29198913

ABSTRACT

Monoclonal antibodies (mAbs) can destroy tumors by recruiting effectors such as myeloid cells, or targeting immunomodulatory receptors to promote cytotoxic T cell responses. Here, we examined the therapeutic potential of combining a direct tumor-targeting mAb, anti-CD20, with an extended panel of immunomodulatory mAbs. Only the anti-CD27/CD20 combination provided cures. This was apparent in multiple lymphoma models, including huCD27 transgenic mice using the anti-huCD27, varlilumab. Detailed mechanistic analysis using single-cell RNA sequencing demonstrated that anti-CD27 stimulated CD8+ T and natural killer cells to release myeloid chemo-attractants and interferon gamma, to elicit myeloid infiltration and macrophage activation. This study demonstrates the therapeutic advantage of using an immunomodulatory mAb to regulate lymphoid cells, which then recruit and activate myeloid cells for enhanced killing of mAb-opsonized tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Lymphoma/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/agonists , Animals , Antibodies, Monoclonal, Humanized , Humans , Immunotherapy/methods , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Mice , Mice, Transgenic
19.
Clin Cancer Res ; 10(6): 1920-7, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15041707

ABSTRACT

PURPOSE: The oncofetal antigen, human chorionic gonadotropin beta subunit (hCGbeta), is expressed by a number of carcinomas and is a prognostic indicator in renal, colorectal, bladder, and pancreatic cancers. We describe the development of a novel antibody-based dendritic cell (DC)-targeted cancer vaccine capable of eliciting cellular immune responses directed against hCGbeta. EXPERIMENTAL DESIGN: The tumor-associated antigen hCGbeta was coupled genetically to a human anti-DC antibody (B11). The resulting fusion protein (B11-hCGbeta) was evaluated for its ability to promote tumor antigen-specific cellular immune responses in a human in vitro model. Monocyte-derived human DCs from normal donors were exposed to purified B11-hCGbeta, activated with CD40 ligand, mixed with autologous lymphocytes, and tested for their ability to promote hCGbeta-specific proliferative and cytotoxic T-lymphocyte responses. RESULTS: B11-hCGbeta was found to be a soluble, well-defined, and readily purified product that specifically recognized the human mannose receptor via the B11 antibody portion of the fusion protein. B11-hCGbeta functionally promoted the uptake and processing of tumor antigen by DCs, which led to the generation of tumor-specific HLA class I and class II-restricted T-cell responses, including CTLs capable of killing human cancer cell lines expressing hCGbeta. CONCLUSIONS: Although other hCG vaccines have been shown to be capable of eliciting antibody responses to hCGbeta, this is the first time that cellular immune responses to hCGbeta have been induced by a vaccine in a human system. This DC-targeted hCGbeta vaccine holds promise for the management of a number of cancers and merits additional clinical development.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/pharmacology , Chorionic Gonadotropin, beta Subunit, Human/immunology , Dendritic Cells/immunology , Animals , Antigens, Neoplasm/isolation & purification , Cancer Vaccines/immunology , Cancer Vaccines/isolation & purification , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Dendritic Cells/drug effects , Humans , Immunity, Cellular/drug effects , Immunoglobulin G , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Nude , Monocytes/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
20.
Cell Mol Immunol ; 12(6): 719-28, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25345808

ABSTRACT

Previous studies have documented that selective delivery of protein antigens to cells expressing mannose receptor (MR) can lead to enhanced immune responses. We postulated that agents that influenced the MR expression level, and the activation and migration status of MR-expressing antigen presenting cells, would modulate immune responses to MR-targeted vaccines. To address this question, we investigated the effect of clinically used adjuvants in human MR transgenic (hMR-Tg) mice immunized with an MR-targeting cancer vaccine composed of the human anti-MR monoclonal antibody B11 fused with the oncofetal protein, human chorionic gonadotropin beta chain (hCGß), and referred to as B11-hCGß. We found that humoral responses to low doses of B11-hCGß could be enhanced by prior administration of GM-CSF, which upregulated MR expression in vivo. However, co-administration of the Toll-like receptor (TLR) agonists, poly-ICLC and/or CpG with B11-hCGß was required to elicit Th1 immunity, as measured by antigen-specific T-cell production of IFN-γ. The TLR agonists were shown to increase the number of vaccine-containing cells in the draining lymph nodes of immunized hMR-Tg mice. In particular, with B11-hCGß and poly-ICLC, a dramatic increase in vaccine-positive cells was observed in the T-cell areas of the lymph nodes, compared to the vaccine alone or combined with GM-CSF. Importantly, the absence of the TLR agonists during the priming immunization led to antigen-specific tolerance. Therefore, this study provides insight into the mechanisms by which adjuvants can augment immune responses to B11-hCGß and have implications for the rationale design of clinical studies combining MR-targeted vaccination with TLR agonists.


Subject(s)
Antigen-Presenting Cells/drug effects , Cancer Vaccines/genetics , Carboxymethylcellulose Sodium/analogs & derivatives , Lectins, C-Type/genetics , Mannose-Binding Lectins/genetics , Oligodeoxyribonucleotides/pharmacology , Poly I-C/pharmacology , Polylysine/analogs & derivatives , Receptors, Cell Surface/genetics , T-Lymphocytes/drug effects , Toll-Like Receptors/agonists , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/pharmacology , Chorionic Gonadotropin, beta Subunit, Human/administration & dosage , Chorionic Gonadotropin, beta Subunit, Human/genetics , Chorionic Gonadotropin, beta Subunit, Human/immunology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunity, Cellular/drug effects , Interferon-gamma/genetics , Interferon-gamma/immunology , Lectins, C-Type/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymphocyte Count , Mannose Receptor , Mannose-Binding Lectins/immunology , Mice , Mice, Transgenic , Polylysine/pharmacology , Receptors, Cell Surface/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Th1 Cells/cytology , Th1 Cells/drug effects , Th1 Cells/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL