Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
JCI Insight ; 7(19)2022 10 10.
Article En | MEDLINE | ID: mdl-35998045

New strategies that augment T cell responses are required to broaden the therapeutic arsenal against cancer. CD96, TIGIT, and CD226 are receptors that bind to a communal ligand, CD155, and transduce either inhibitory or activating signals. The function of TIGIT and CD226 is established, whereas the role of CD96 remains ambiguous. Using a panel of engineered antibodies, we discovered that the T cell stimulatory activity of anti-CD96 antibodies requires antibody cross-linking and is potentiated by Fcγ receptors. Thus, soluble "Fc silent" anti-CD96 antibodies failed to stimulate human T cells, whereas the same antibodies were stimulatory after coating onto plastic surfaces. Remarkably, the activity of soluble anti-CD96 antibodies was reinstated by engineering the Fc domain to a human IgG1 isotype, and it was dependent on antibody trans-cross-linking by FcγRI. In contrast, neither human IgG2 nor variants with increased Fcγ receptor IIB binding possessed stimulatory activity. Anti-CD96 antibodies acted directly on T cells and augmented gene expression networks associated with T cell activation, leading to proliferation, cytokine secretion, and resistance to Treg suppression. Furthermore, CD96 expression correlated with survival in HPV+ head and neck squamous cell carcinoma, and its cross-linking activated tumor-infiltrating T cells, thus highlighting the potential of anti-CD96 antibodies in cancer immunotherapy.


Killer Cells, Natural , Neoplasms , Antigens, CD/metabolism , Cytokines/metabolism , Humans , Immunoglobulin G/metabolism , Ligands , Plastics/metabolism , Receptors, IgG/metabolism , Receptors, Immunologic/metabolism
2.
Mol Clin Oncol ; 10(2): 309-317, 2019 Feb.
Article En | MEDLINE | ID: mdl-30680213

The treatment options for advanced (stage IV) non-small cell lung cancer (NSCLC) at diagnosis remain disappointing. The development of immunotherapeutic drugs may represent a promising alternative approach to the treatment of late-stage cancer at diagnosis. These current paradigms in cancer treatment highlight the need for new biomarkers related to the immune status of the patients and/or the tumor microenvironment, for immune as well as chemotherapeutic treatment options. The aim of the present study was to analyze soluble immune factors in patients with lung cancer treated with chemotherapy to identify prognostic biomarkers. For this purpose, the data obtained from two cohorts of patients from different clinical trials were analyzed: A Chinese patient cohort to identify potential prognostic biomarkers, and a validation cohort comprising patients with a similar clinical stage from a clinical trial in Europe. Analyses of soluble markers for inflammation and immune status were performed by standard assays and multiplex Luminex assays. Differences in overall survival (OS) and progression-free survival (PFS) were evaluated with the log-rank test and robustness was evaluated with the resampling approach. In the Chinese cohort, four prognostic biomarkers of poor response to chemotherapy were identified, which had a significant impact on OS and PFS. It was confirmed in the Caucasian validation cohort that an increased value of the interleukin (IL)-6/IL-1Ra cytokine ratio at inclusion was correlated with significantly shorter OS and PFS, whereas no other biomarkers were found to be significant. The IL-6/IL-1Ra ratio reflects the imbalance between pro- and anti-inflammatory status in the plasma of patients and may be associated with tumor inflammatory status and the therapeutic outcome. The present study highlights the identification of the IL-6/IL-1Ra ratio as a biomarker of poor prognosis in terms of response to chemotherapy in two independent clinical studies.

3.
Hum Vaccin Immunother ; 14(1): 140-145, 2018 01 02.
Article En | MEDLINE | ID: mdl-28925793

TG4010, a Modified Vaccinia virus Ankara (MVA) expressing human mucin1 (MUC1) has demonstrated clinical benefit for patients suffering from advanced non-small cell lung cancer (NSCLC) in combination with chemotherapy. To support its development, preclinical experiments were performed with either TG4010 or ß-galactosidase-encoding MVA vector (MVA-ßgal) in mice presenting tumors in the lung. Tumor growth was obtained after intravenous injection of CT26 murine colon cancer cells, engineered to express either MUC1 or ßgal. Mice showed increased survival rates after repeated intravenous injections of TG4010 or MVA-ßgal, compared to an empty MVA control vector. Treatment with MVA vectors led to the accumulation of CD3dimCD8dim T cells, with two subpopulations characterized as KLRG1+CD127- short-lived effector cells (SLECs), and KLRG1-CD127- early effector cells (EECs) comprising cells releasing IFNγ, Granzyme B and CD107a upon antigen-specific peptide stimulation. EECs were characterized by an up-regulation of PD-1. Tumor growth in the diseased lung correlated with the appearance of PD1+ Treg cells that partially disappeared after TG4010 treatment. At late stage of tumor development in the lung, PD-L1 was detected on CD45- tumor cells, on CD4+ cells, including Treg cells, on CD3+CD8+ and CD3dimCD8dim T lymphocytes, on NK cells, on MDSCs and on alveolar macrophages. We demonstrated that targeting the PD-1/PD-L1 pathway with blocking monoclonal antibodies several days after TG4010 treatment, at late stage of tumor development, enhanced the therapeutic protection induced by the vaccine, supporting the ongoing clinical evaluation of TG4010 immunotherapy in combination with Nivolumab.


Antineoplastic Agents, Immunological/administration & dosage , Cancer Vaccines/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy/methods , Membrane Glycoproteins/administration & dosage , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Vaccinia virus/immunology , Animals , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Combined Modality Therapy/methods , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mucin-1/immunology , Nivolumab/administration & dosage , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
4.
J Immunother Cancer ; 5(1): 70, 2017 09 19.
Article En | MEDLINE | ID: mdl-28923084

BACKGROUND: Advanced non-small cell lung cancer patients receiving TG4010, a therapeutic viral vaccine encoding human Mucin 1 and interleukin-2 in addition to standard chemotherapy, displayed longer overall survival in comparison to that of patients treated with standard chemotherapy alone. Our study intended to establish the association between overall survival and vaccine-induced T cell responses against tumor associated antigens (TAA) targeted by the vaccine. METHOD: The TIME trial was a placebo-controlled, randomized phase II study aimed at assessing efficacy of TG4010 with chemotherapy in NSCLC. 78 patients from the TIME study carrying the HLA-A02*01 haplotype were analyzed using combinatorial encoding of MHC multimers to detect low frequencies of cellular immune responses to TG4010 and other unrelated TAA. RESULTS: We report that improvement of survival under TG4010 treatment correlated with development of T cell responses against MUC1. Interestingly, responses against MUC1 were associated with broadening of CD8 responses against non-targeted TAA, thus demonstrating induction of epitope spreading. CONCLUSION: Our results support the causality of specific T-cell response in improved survival in NSCLC. Additionally, vaccine induced epitope spreading to other TAA participates to the enrichment of the diversity of the anti-tumor response. Hence, TG4010 appears as a useful therapeutic option to maximize response rate and clinical benefit in association with other targeted immuno-modulators. TRIAL REGISTRATION: Registered on ClinicalTrials.gov under identifier NCT01383148 on June 23rd, 2011.


Antigens, Neoplasm/metabolism , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Membrane Glycoproteins/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Double-Blind Method , HLA-A2 Antigen/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mucin-1/metabolism , Survival Analysis , T-Lymphocytes/immunology , Treatment Outcome
5.
Cancer Res ; 77(15): 4146-4157, 2017 08 01.
Article En | MEDLINE | ID: mdl-28536278

Athough the clinical efficacy of oncolytic viruses has been demonstrated for local treatment, the ability to induce immune-mediated regression of distant metastases is still poorly documented. We report here that the engineered oncolytic vaccinia virus VVWR-TK-RR--Fcu1 can induce immunogenic cell death and generate a systemic immune response. Effects on tumor growth and survival was largely driven by CD8+ T cells, and immune cell infiltrate in the tumor could be reprogrammed toward a higher ratio of effector T cells to regulatory CD4+ T cells. The key role of type 1 IFN pathway in oncolytic virotherapy was also highlighted, as we observed a strong abscopal response in Ifnar-/- tumors. In this model, single administration of virus directly into the tumors on one flank led to regression in the contralateral flank. Moreover, these effects were further enhanced when oncolytic treatment was combined with immunogenic chemotherapy or with immune checkpoint blockade. Taken together, our results suggest how to safely improve the efficacy of local oncolytic virotherapy in patients whose tumors are characterized by dysregulated IFNα signaling. Cancer Res; 77(15); 4146-57. ©2017 AACR.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Interferon-alpha/antagonists & inhibitors , Neoplasms, Experimental/therapy , Oncolytic Virotherapy/methods , Animals , B7-H1 Antigen/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oncolytic Viruses , Organoplatinum Compounds/pharmacology , Oxaliplatin , Receptor, Interferon alpha-beta/metabolism , Vaccinia virus
6.
Mol Clin Oncol ; 6(2): 279-285, 2017 Feb.
Article En | MEDLINE | ID: mdl-28357111

Non-small-cell lung cancer (NSCLC) may establish an immunosuppressive tumor microenvironment that is conducive to tumor growth. Natural killer (NK) cells play a pivotal role in immunological surveillance. Activation of NK cells partially depends on the interactions between killer-cell immunoglobulin-like receptors (KIRs) and human leukocyte antigen (HLA) class I ligands. We herein investigated the association of KIRs and HLA ligands with survival in metastatic NSCLC (mNSCLC) patients treated with chemotherapy in a Chinese Han population. Polymerase chain reaction with sequence-specific primers was used to type 15 KIRs at the DNA and mRNA level and 6 HLA ligands in 70 mNSCLC patients. Survival curves were estimated using the Kaplan-Meier method and compared with the log-rank test. Cox proportional hazard regression model was applied for multivariate survival analysis, with the stepwise selection, to determine independent predictors of survival. It was observed that patients with KIR2DS4del gene expression at the mRNA level or HLA-Bw4T80 exhibited poor overall survival (OS). The multivariate analysis revealed that HLA-Bw4T80 and KIR2DS4del expression were independent predictors of OS. This observation indicated that the KIR/HLA ligand is a promising predictor of survival in mNSCLC and may also provide a strategy for treatment stratification and patient management.

7.
Vaccine ; 35(4): 577-585, 2017 01 23.
Article En | MEDLINE | ID: mdl-28012777

TG4010 is an immunotherapeutic vaccine based on Modified Vaccinia virus Ankara (MVA) encoding the human tumor-associated antigen MUC1 and human IL-2. In combination with first-line standard of care chemotherapy in advanced metastatic non-small-cell lung cancer (NSCLC), repeated subcutaneous injection of TG4010 improved progression-free survival in phase 2b clinical trials. In preclinical tumor models, MVATG9931, the research version of TG4010, conferred antigen-specific responses against the weak antigen human MUC1. The combination of a suboptimal dose of MVATG9931 and the type B TLR9 ligand Litenimod (Li28) markedly increased survival in a subcutaneous RMA-MUC1 tumor model compared to the treatment with MVATG9931 or Li28 alone. The requirements for this protection were (i) de novo synthesis of MUC1, (ii) Li28 delivered several hours after MVATG9931 at the same site, (iii) at least two vaccination cycles, and (iv) implantation of MUC1-positive tumor cells in the vicinity to the vaccination site. Subcutaneously injected MVATG9931 allowed transient local gene expression and induced the local accumulation of MCP-1, RANTES, M-CSF, IL-15/IL-15R and IP-10. After repeated injection, CD4+ and CD8+ T lymphocytes, B lymphocytes, NK cells, pDCs, neutrophils, and macrophages accumulated around the injection site, local RANTES levels remained high. Delayed injection of Li28 into this environment, led to further accumulation of macrophages, the secretion of IL-18 and IL-1 beta, and an increase of the percentage of activated CD69+ NK cell. Combination treatment augmented the number of activated CD86+ DCs in the draining lymph nodes and increased the percentage of KLRG1+ CD127-CD8+ T cells at the injection site. In vivo depletion of macrophages around the injection site by Clodronate liposomes reduced local IL-18 levels and diminished survival rates significantly. Thus, sequential administration of MVATG9931 and Li28 improves local innate and adaptive immune defense against tumors, arguing for intratumoral delivery of this peculiar sequential combination therapy.


Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Membrane Glycoproteins/immunology , Mucin-1/immunology , Neoplasms/therapy , Toll-Like Receptor 9/agonists , Animals , Cancer Vaccines/administration & dosage , Disease Models, Animal , Drug Carriers/administration & dosage , Injections, Subcutaneous , Membrane Glycoproteins/administration & dosage , Mice, Inbred C57BL , Treatment Outcome , Vaccinia virus/genetics
8.
Oncoimmunology ; 5(10): e1220467, 2016.
Article En | MEDLINE | ID: mdl-27853644

We report here the successful vectorization of a hamster monoclonal IgG (namely J43) recognizing the murine Programmed cell death-1 (mPD-1) in Western Reserve (WR) oncolytic vaccinia virus. Three forms of mPD-1 binders have been inserted into the virus: whole antibody (mAb), Fragment antigen-binding (Fab) or single-chain variable fragment (scFv). MAb, Fab and scFv were produced and assembled with the expected patterns in supernatants of cells infected by the recombinant viruses. The three purified mPD-1 binders were able to block the binding of mPD-1 ligand to mPD-1 in vitro. Moreover, mAb was detected in tumor and in serum of C57BL/6 mice when the recombinant WR-mAb was injected intratumorally (IT) in B16F10 and MCA 205 tumors. The concentration of circulating mAb detected after IT injection was up to 1,900-fold higher than the level obtained after a subcutaneous (SC) injection (i.e., without tumor) confirming the virus tropism for tumoral cells and/or microenvironment. Moreover, the overall tumoral accumulation of the mAb was higher and lasted longer after IT injection of WR-mAb1, than after IT administration of 10 µg of J43. The IT injection of viruses induced a massive infiltration of immune cells including activated lymphocytes (CD8+ and CD4+). Interestingly, in the MCA 205 tumor model, WR-mAb1 and WR-scFv induced a therapeutic control of tumor growth similar to unarmed WR combined to systemically administered J43 and superior to that obtained with an unarmed WR. These results pave the way for next generation of oncolytic vaccinia armed with immunomodulatory therapeutic proteins such as mAbs.

9.
Oncoimmunology ; 5(2): e1080414, 2016 Feb.
Article En | MEDLINE | ID: mdl-27057460

Oncolytic virotherapy is an emergent promising therapeutic approach for the treatment of cancer. We have constructed a vaccinia virus (WR strain) deleted for thymidine kinase (TK) and ribonucleotide reductase (RR) genes that expressed the fusion suicide gene FCU1 derived from the yeast cytosine deaminase and uracil phosphoribosyltransferase genes. We evaluated this construct (VV-FCU1) in the orthotopic model of renal carcinoma (RenCa). Systemic administration of VV-FCU1 resulted in orthotopic tumor growth inhibition, despite temporary expression of viral proteins. VV-FCU1 treatment was associated with an infiltration of tumors by CD8+ T lymphocytes and a decrease in the proportion of infiltrating Tregs, thus modifying the ratio of CD8+/CD4+ Treg in favor of CD8+cytotoxic T cells. We demonstrated that VV-FCU1 treatment prolonged survival of animals implanted with RenCa cells in kidney. Depletion of CD8+ T cells abolished the therapeutic effect of VV-FCU1 while depletion of CD4+ T cells enhanced its protective activity. Administration of the prodrug 5-fluorocytosine (5-FC) resulted in a sustained control of tumor growth but did not extend survival. This study shows the importance of CD4+ and CD8+ T cells in vaccinia virus-mediated oncolytic virotherapy and suggests that this approach may be evaluated for the treatment of human renal cell carcinoma.

10.
Oncoimmunology ; 5(2): e1117740, 2016 Feb.
Article En | MEDLINE | ID: mdl-27057469

Oncolytic virotherapy relies on the administration of non-pathogenic viral strains that selectively infect and kill malignant cells while favoring the elicitation of a therapeutically relevant tumor-targeting immune response. During the past few years, great efforts have been dedicated to the development of oncolytic viruses with improved specificity and potency. Such an intense wave of investigation has culminated this year in the regulatory approval by the US Food and Drug Administration (FDA) of a genetically engineered oncolytic viral strain for use in melanoma patients. Here, we summarize recent preclinical and clinical advances in oncolytic virotherapy.

11.
Oncoimmunology ; 4(5): e1003013, 2015 May.
Article En | MEDLINE | ID: mdl-26155396

Our preclinical data demonstrate that an intravenous injection of Modified Vaccinia virus Ankara induces CD8+ lymphocytes to infiltrate organs to control the growth of orthotopic renal carcinoma upon combination with a toll-like receptor 9 agonist. Such shaping of the tumor microenvironment could constitute the basis of more effective clinical protocols of tumor immunotherapy.

12.
Nat Med ; 20(11): 1301-9, 2014 Nov.
Article En | MEDLINE | ID: mdl-25344738

Some of the anti-neoplastic effects of anthracyclines in mice originate from the induction of innate and T cell-mediated anticancer immune responses. Here we demonstrate that anthracyclines stimulate the rapid production of type I interferons (IFNs) by malignant cells after activation of the endosomal pattern recognition receptor Toll-like receptor 3 (TLR3). By binding to IFN-α and IFN-ß receptors (IFNARs) on neoplastic cells, type I IFNs trigger autocrine and paracrine circuitries that result in the release of chemokine (C-X-C motif) ligand 10 (CXCL10). Tumors lacking Tlr3 or Ifnar failed to respond to chemotherapy unless type I IFN or Cxcl10, respectively, was artificially supplied. Moreover, a type I IFN-related signature predicted clinical responses to anthracycline-based chemotherapy in several independent cohorts of patients with breast carcinoma characterized by poor prognosis. Our data suggest that anthracycline-mediated immune responses mimic those induced by viral pathogens. We surmise that such 'viral mimicry' constitutes a hallmark of successful chemotherapy.


Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Doxorubicin/therapeutic use , Interferon Type I/metabolism , Signal Transduction , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Chemokine CXCL10/metabolism , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunocompetence/drug effects , Interferon Type I/biosynthesis , Mice, Inbred C57BL , Myxovirus Resistance Proteins/metabolism , Neoadjuvant Therapy , Neoplasm Metastasis , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism , Treatment Outcome
13.
Oncoimmunology ; 3: e28694, 2014.
Article En | MEDLINE | ID: mdl-25097804

Oncolytic viruses are natural or genetically modified viral species that selectively infect and kill neoplastic cells. Such an innate or exogenously conferred specificity has generated considerable interest around the possibility to employ oncolytic viruses as highly targeted agents that would mediate cancer cell-autonomous anticancer effects. Accumulating evidence, however, suggests that the therapeutic potential of oncolytic virotherapy is not a simple consequence of the cytopathic effect, but strongly relies on the induction of an endogenous immune response against transformed cells. In line with this notion, superior anticancer effects are being observed when oncolytic viruses are engineered to express (or co-administered with) immunostimulatory molecules. Although multiple studies have shown that oncolytic viruses are well tolerated by cancer patients, the full-blown therapeutic potential of oncolytic virotherapy, especially when implemented in the absence of immunostimulatory interventions, remains unclear. Here, we cover the latest advances in this active area of translational investigation, summarizing high-impact studies that have been published during the last 12 months and discussing clinical trials that have been initiated in the same period to assess the therapeutic potential of oncolytic virotherapy in oncological indications.

14.
Cancer Immunol Res ; 2(12): 1163-74, 2014 Dec.
Article En | MEDLINE | ID: mdl-25168392

Effector T-cell access to tumor tissue is a limiting step for clinical efficacy of antigen-specific T cell-based immunotherapies. Ectopic mouse tumor models, in which a subcutaneously (s.c.) implanted tumor is treated with s.c. or intramuscular therapeutic immunization, may not be optimal for targeting effector T cells to an organ-borne tumor. We used an orthotopic renal carcinoma model to evaluate the impact of injection routes on therapeutic efficacy of a Modified Vaccinia virus Ankara viral vector expressing the human mucin 1 tumor-associated xeno-antigen (MVA-MUC1). We show that intravenous (i.v.) administration of MVA-MUC1 displayed enhanced efficacy when compared with s.c. injection. Therapeutic efficacy of MVA-MUC1 was further enhanced by i.v. injection of a TLR9 agonist. In all cases, infiltration of tumor-bearing kidney by CD8(+) lymphocytes was associated with control of tumor growth. Biodistribution experiments indicate that, following i.v. injection, MVA-encoded antigens are quickly expressed in visceral organs and, in particular, in splenic antigen-presenting cells, compared with those following s.c. injection. This appears to result in a faster generation of MUC1-specific CD8(+) T cells. Lymphocytes infiltrating tumor-bearing kidneys are characterized by an effector memory phenotype and express PD-1 and Tim3 immune checkpoint molecules. Therapeutic efficacy was associated with a modification of the tumor microenvironment toward a Th1-type immune response and recruitment of activated lymphocytes. This study supports the clinical evaluation of MVA-based immunotherapies via the i.v. route.


CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Genetic Vectors/genetics , Neoplasms/immunology , Neoplasms/therapy , Toll-Like Receptor 9/agonists , Vaccinia virus/genetics , Animals , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Genetic Vectors/administration & dosage , Humans , Immunophenotyping , Immunotherapy , Injections, Intravenous , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mucin-1/genetics , Mucin-1/immunology , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacology , Phenotype , Tissue Distribution , Tumor Burden/drug effects , Tumor Burden/immunology
15.
Arthritis Rheumatol ; 66(11): 2989-3000, 2014 Nov.
Article En | MEDLINE | ID: mdl-24623505

OBJECTIVE: Colony-stimulating factor 1 receptor (CSF-1R) essentially modulates monocyte proliferation, migration, and activation, which are considered important for the pathogenesis of rheumatoid arthritis (RA). We undertook this study to determine CSF-1R expression in human RA as well as the efficacy of a specific anti-CSF-1R monoclonal antibody (AFS98) in 2 different animal models of RA. METHODS: CSF-1R expression was examined in blood, synovium, and bone samples from RA patients, osteoarthritis (OA) patients, and healthy subjects. The efficacy of AFS98 was examined by clinical assessment, histology, and bone histomorphometry in collagen-induced arthritis (CIA) and serum-transfer arthritis. RESULTS: CSF-1R expression was increased in the synovium of RA patients compared to OA patients and healthy controls in fibroblast-like synoviocytes, follicular dendritic cells, macrophages, and osteoclasts. Circulating RA monocytes and neutrophils but not lymphocytes were CSF-1R+. In mice, blockade of CSF-1R abrogated cartilage damage, bone erosion, and systemic bone loss, and this was associated with the depletion of osteoclasts in both models. While blockade of CSF-1R did not affect inflammation in passive serum-transfer arthritis, it significantly reduced inflammation in CIA, and this was associated with the absence of synovial macrophages and reduced splenic CD11b+Gr-1- monocytes. CONCLUSION: CSF-1R was broadly expressed in human RA. Blockade of CSF-1R protected against bone and cartilage destruction in both mouse models and also showed significant antiinflammatory effects in the CIA model. These data provide evidence for CSF-1R as a therapeutic target in RA.


Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Bone and Bones/pathology , Cartilage/pathology , Osteoarthritis/pathology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Cartilage/drug effects , Cartilage/metabolism , Case-Control Studies , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Models, Animal , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred DBA , Middle Aged , Monocytes/metabolism , Monocytes/pathology , Osteoarthritis/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Receptor, Macrophage Colony-Stimulating Factor/drug effects , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology
16.
J Virol ; 88(10): 5242-55, 2014 May.
Article En | MEDLINE | ID: mdl-24574403

UNLABELLED: To identify novel stimulators of the innate immune system, we constructed a panel of eight HEK293 cell lines double positive for human Toll-like receptors (TLRs) and an NF-κB-inducible reporter gene. Screening of a large variety of compounds and cellular extracts detected a TLR3-activating compound in a microsomal yeast extract. Fractionation of this extract identified an RNA molecule of 4.6 kb, named nucleic acid band 2 (NAB2), that was sufficient to confer the activation of TLR3. Digests with single- and double-strand-specific RNases showed the double-strand nature of this RNA, and its sequence was found to be identical to that of the genome of the double-stranded RNA (dsRNA) L-BC virus of Saccharomyces cerevisiae. A large-scale process of production and purification of this RNA was established on the basis of chemical cell lysis and dsRNA-specific chromatography. NAB2 complexed with the cationic lipid Lipofectin but neither NAB2 nor Lipofectin alone induced the secretion of interleukin-12(p70) [IL-12(p70)], alpha interferon, gamma interferon-induced protein 10, macrophage inflammatory protein 1ß, or IL-6 in human monocyte-derived dendritic cells. While NAB2 activated TLR3, Lipofectin-stabilized NAB2 also signaled via the cytoplasmic sensor for RNA recognition MDA-5. A significant increase of RMA-MUC1 tumor rejection and survival was observed in C57BL/6 mice after prophylactic vaccination with MUC1-encoding modified vaccinia virus Ankara (MVA) and NAB2-Lipofectin. This combination of immunotherapies strongly increased at the injection sites the percentage of infiltrating natural killer (NK) cells and plasmacytoid dendritic cells (pDCs), cell types which can modulate innate and adaptive immune responses. IMPORTANCE: Virus-based cancer vaccines offer a good alternative to the treatment of cancer but could be improved. Starting from a screening approach, we have identified and characterized an unexplored biological molecule with immunomodulatory characteristics which augments the efficacy of an MVA-based immunotherapeutic agent. The immune modulator consists of the purified dsRNA genome isolated from a commercially used yeast strain, NAB2, mixed with a cationic lipid, Lipofectin. NAB2-Lipofectin stimulates the immune system via TLR3 and MDA-5. When it was injected at the MVA vaccination site, the immune modulator increased survival in a preclinical tumor model. We could demonstrate that NAB2-Lipofectin augments the MVA-induced infiltration of natural killer and plasmacytoid dendritic cells. We suggest indirect mechanisms of activation of these cell types by the influence of NAB2-Lipofectin on innate and adaptive immunity. Detailed analysis of cell migration at the vaccine injection site and the appropriate choice of an immune modulator should be considered to achieve the rational improvement of virus vector-based vaccination by immune modulators.


Dendritic Cells/immunology , Immunologic Factors/immunology , Neoplasms/therapy , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Saccharomyces cerevisiae/virology , Toll-Like Receptor 3/immunology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Immunologic Factors/isolation & purification , Immunologic Factors/therapeutic use , Immunotherapy/methods , Mice , Mice, Inbred C57BL , RNA, Double-Stranded/isolation & purification , RNA, Double-Stranded/therapeutic use , RNA, Viral/isolation & purification , RNA, Viral/therapeutic use , Survival Analysis
17.
Clin Vaccine Immunol ; 21(2): 147-55, 2014 Feb.
Article En | MEDLINE | ID: mdl-24307238

Women showing normal cytology but diagnosed with a persistent high-risk human papillomavirus (HR-HPV) infection have a higher risk of developing high-grade cervical intraepithelial neoplasia and cervical cancer than noninfected women. As no therapeutic management other than surveillance is offered to these women, there is a major challenge to develop novel targeted therapies dedicated to the treatment of these patients. As such, E1 and E2 antigens, expressed early in the HPV life cycle, represent very interesting candidates. Both proteins are necessary for maintaining coordinated viral replication and gene synthesis during the differentiation process of the epithelium and are essential for the virus to complete its normal and propagative replication cycle. In the present study, we evaluated a new active targeted immunotherapeutic, a modified vaccinia virus Ankara (MVA) vector containing the E1 sequence of HPV16, aimed at inducing cellular immune responses with the potential to help and clear persistent HPV16-related infection. We carried out an extensive comparative time course analysis of the cellular immune responses induced by different schedules of immunization in C57BL/6 mice. We showed that multiple injections of MVA-E1 allowed sustained HPV16 E1-specific cellular immune responses in vaccinated mice and had no impact on the exhaustion phenotype of the generated HPV16 E1-specific CD8⁺ T cells, but they led to the differentiation of multifunctional effector T cells with high cytotoxic capacity. This study provides proof of concept that an MVA expressing HPV16 E1 can induce robust and long-lasting E1-specific responses and warrants further development of this candidate.


CD8-Positive T-Lymphocytes/immunology , Drug Carriers , Immunity, Cellular , Oncogene Proteins, Viral/immunology , Papillomavirus Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccinia virus/genetics , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Papillomavirus Vaccines/administration & dosage , Papillomavirus Vaccines/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
18.
PLoS One ; 8(9): e73310, 2013.
Article En | MEDLINE | ID: mdl-24019914

Tumor progression is promoted by Tumor-Associated Macrophages (TAMs) and metastasis-induced bone destruction by osteoclasts. Both myeloid cell types depend on the CD115-CSF-1 pathway for their differentiation and function. We used 3 different mouse cancer models to study the effects of targeting cancer host myeloid cells with a monoclonal antibody (mAb) capable of blocking CSF-1 binding to murine CD115. In mice bearing sub-cutaneous EL4 tumors, which are CD115-negative, the anti-CD115 mAb depleted F4/80(+) CD163(+) M2-type TAMs and reduced tumor growth, resulting in prolonged survival. In the MMTV-PyMT mouse model, the spontaneous appearance of palpable mammary tumors was delayed when the anti-CD115 mAb was administered before malignant transition and tumors became palpable only after termination of the immunotherapy. When administered to mice already bearing established PyMT tumors, anti-CD115 treatment prolonged their survival and potentiated the effect of chemotherapy with Paclitaxel. As shown by immunohistochemistry, this therapeutic effect correlated with the depletion of F4/80(+)CD163(+) M2-polarized TAMs. In a breast cancer model of bone metastasis, the anti-CD115 mAb potently blocked the differentiation of osteoclasts and their bone destruction activity. This resulted in the inhibition of cancer-induced weight loss. CD115 thus represents a promising target for cancer immunotherapy, since a specific blocking antibody may not only inhibit the growth of a primary tumor through TAM depletion, but also metastasis-induced bone destruction through osteoclast inhibition.


Antibodies, Monoclonal/therapeutic use , Macrophages/immunology , Neoplasms, Experimental/therapy , Osteoclasts/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/immunology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Survival Analysis
19.
MAbs ; 5(5): 736-47, 2013.
Article En | MEDLINE | ID: mdl-23924795

Cancer progression has been associated with the presence of tumor-associated M2-macrophages (M2-TAMs) able to inhibit anti-tumor immune responses. It is also often associated with metastasis-induced bone destruction mediated by osteoclasts. Both cell types are controlled by the CD115 (CSF-1R)/colony-stimulating factor-1 (CSF-1, M-CSF) pathway, making CD115 a promising target for cancer therapy. Anti-human CD115 monoclonal antibodies (mAbs) that inhibit the receptor function have been generated in a number of laboratories. These mAbs compete with CSF-1 binding to CD115, dramatically affecting monocyte survival and preventing osteoclast and macrophage differentiation, but they also block CD115/CSF-1 internalization and degradation, which could lead to potent rebound CSF-1 effects in patients after mAb treatment has ended. We thus generated and selected a non-ligand competitive anti-CD115 mAb that exerts only partial inhibitory effects on CD115 signaling without blocking the internalization or the degradation of the CD115/CSF-1 complex. This mAb, H27K15, affects monocyte survival only minimally, but downregulates osteoclast differentiation and activity. Importantly, it inhibits monocyte differentiation to CD163(+)CD64(+) M2-polarized suppressor macrophages, skewing their differentiation toward CD14(-)CD1a(+) dendritic cells (DCs). In line with this observation, H27K15 also drastically inhibits monocyte chemotactic protein-1 secretion and reduces interleukin-6 production; these two molecules are known to be involved in M2-macrophage recruitment. Thus, the non-depleting mAb H27K15 is a promising anti-tumor candidate, able to inhibit osteoclast differentiation, likely decreasing metastasis-induced osteolysis, and able to prevent M2 polarization of TAMs while inducing DCs, hence contributing to the creation of more efficient anti-tumor immune responses.


Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Macrophages/drug effects , Monocytes/drug effects , Osteolysis/prevention & control , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , HL-60 Cells , Humans , Interleukin-6/immunology , Interleukin-6/metabolism , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Monocytes/immunology , Monocytes/metabolism , NIH 3T3 Cells , Osteoclasts/drug effects , Osteoclasts/immunology , Osteoclasts/metabolism , Osteolysis/immunology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
20.
Oncoimmunology ; 1(8): 1271-1280, 2012 Nov 01.
Article En | MEDLINE | ID: mdl-23243590

Cancer immunotherapy is hampered by the immunosuppression maintained by regulatory T cells (Tregs) in tumor-bearing hosts. Stimulation of the Toll-like receptor 2 (TLR2) by Pam3Cys is known to affect Treg-mediated suppression. We found that Pam3Cys increases the proliferation of both CD4(+) effector T cells (Teffs) and Tregs co-cultured in vitro, but did not induce the proliferation of Tregs alone upon CD3 and CD28 stimulation. In a mouse model of RMA-MUC1 tumors, Pam3Cys was administered either alone or in combination with a modified vaccinia ankara (MVA)-based mucin 1 (MUC1) therapeutic vaccine. The combination of Pam3Cys with MVA-MUC1 (1) diminished splenic Treg/CD4(+) T-cell ratios to those found in tumor-free mice, (2) stimulated a specific anti-MUC1 interferon γ (IFNγ) response and (3) had a significant therapeutic effect on tumor growth and mouse survival. When CD4(+) Teffs and Tregs were isolated from Pam3Cys-treated mice, Teffs had become resistant to Treg-mediated suppression while upregulating the expression of BclL-x(L). Tregs from Pam3Cys-treated mice were fully suppressive for Teffs from naïve mice. Bcl-x(L) was induced by Pam3Cys with different kinetics in Tregs and Teffs. Teff from Pam3Cys-treated mice produced increased levels of Th1 and Th2-type cytokines and an interleukin (IL)-6-dependent secretion of IL-17 was observed in Teff:Treg co-cultures, suggesting that TLR2 stimulation had skewed the immune response toward a Th17 profile. Our results show for the first time that in a tumor-bearing host, TLR2 stimulation with Pam3Cys affects both Tregs and Teffs, protects Teff from Treg-mediated suppression and has strong therapeutic effects when combined with an MVA-based antitumor vaccine.

...