Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Oncoimmunology ; 8(5): e1568162, 2019.
Article in English | MEDLINE | ID: mdl-31069131

ABSTRACT

CD40 triggering may result in antitumor effects of potentially high clinical relevance. To gain insights important for patient selection and to identify adequate targeting techniques, we investigated CD40 expression in human cancer tissues and generated a replication-incompetent recombinant vaccinia virus expressing CD40 ligand (rVV40L). Its effects were explored in vitro and in vivo upon direct CD40 targeting on malignant cells or macrophage activation. CD40 expression was analyzed by immunohistochemistry in tumor and stromal cells in a multi-tumor array including 836 specimens from 27 different tumor types. Established tumor cell lines were used to explore the capacity of rVV40L to induce malignant cell apoptosis and modulate functional profiles of polarized macrophages. CD40 expression was detectable in significantly higher numbers of stromal as compared to malignant cells in lung and breast cancers. CD40 ligation following rVV40L infection induced apoptosis in CD40(+) cancer cells, but only in the presence of intact specific signal transduction chain. Importantly, rVV40L infection promoted the induction of TNF-α-dependent antitumor activity of M1-like macrophages directed against CD40(-) targets. CD40-activated M1-like macrophages also displayed enhanced ability to CXCL10-dependently recruit CD8+ T cells and to efficiently present cancer cell intracellular antigens through cross-priming. Moreover, rVV-driven CD40L expression partially "re-educated" M2-like macrophages, as suggested by detectable CXCL10 and IL-12 production. Most importantly, we observed that intra-tumoral injection of rVV40L-infected human macrophages inhibits progression of human CD40(-) tumors in vivo. First evidences of anticancer activity of rVV40L strongly encourage further evaluations.

2.
Gut ; 67(11): 1984-1994, 2018 11.
Article in English | MEDLINE | ID: mdl-29437871

ABSTRACT

OBJECTIVE: Tumour-infiltrating lymphocytes (TILs) favour survival in human colorectal cancer (CRC). Chemotactic factors underlying their recruitment remain undefined. We investigated chemokines attracting T cells into human CRCs, their cellular sources and microenvironmental triggers. DESIGN: Expression of genes encoding immune cell markers, chemokines and bacterial 16S ribosomal RNA (16SrRNA) was assessed by quantitative reverse transcription-PCR in fresh CRC samples and corresponding tumour-free tissues. Chemokine receptor expression on TILs was evaluated by flow cytometry on cell suspensions from digested tissues. Chemokine production by CRC cells was evaluated in vitro and in vivo, on generation of intraperitoneal or intracecal tumour xenografts in immune-deficient mice. T cell trafficking was assessed on adoptive transfer of human TILs into tumour-bearing mice. Gut flora composition was analysed by 16SrRNA sequencing. RESULTS: CRC infiltration by distinct T cell subsets was associated with defined chemokine gene signatures, including CCL5, CXCL9 and CXCL10 for cytotoxic T lymphocytes and T-helper (Th)1 cells; CCL17, CCL22 and CXCL12 for Th1 and regulatory T cells; CXCL13 for follicular Th cells; and CCL20 and CCL17 for interleukin (IL)-17-producing Th cells. These chemokines were expressed by tumour cells on exposure to gut bacteria in vitro and in vivo. Their expression was significantly higher in intracecal than in intraperitoneal xenografts and was dramatically reduced by antibiotic treatment of tumour-bearing mice. In clinical samples, abundance of defined bacteria correlated with high chemokine expression, enhanced T cell infiltration and improved survival. CONCLUSIONS: Gut microbiota stimulate chemokine production by CRC cells, thus favouring recruitment of beneficial T cells into tumour tissues.


Subject(s)
Chemokines/metabolism , Colorectal Neoplasms/immunology , Gastrointestinal Microbiome/immunology , Lymphocytes, Tumor-Infiltrating/microbiology , Adult , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , In Situ Hybridization , Male , Mice , Middle Aged , RNA, Ribosomal, 16S/metabolism , Real-Time Polymerase Chain Reaction
3.
Front Med (Lausanne) ; 4: 18, 2017.
Article in English | MEDLINE | ID: mdl-28337438

ABSTRACT

MAGE-A antigens are expressed in a variety of cancers of diverse histological origin and germinal cells. Due to their relatively high tumor specificity, they represent attractive targets for active specific and adoptive cancer immunotherapies. Here, we (i) review past and ongoing clinical studies targeting these antigens, (ii) analyze advantages and disadvantages of different therapeutic approaches, and (iii) discuss possible improvements in MAGE-A-specific immunotherapies.

4.
Oncotarget ; 8(1): 1725-1736, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27965457

ABSTRACT

Culture of cancerous cells in standard monolayer conditions poorly mirrors growth in three-dimensional architectures typically observed in a wide majority of cancers of different histological origin. Multicellular tumor spheroid (MCTS) culture models were developed to mimic these features. However, in vivo tumor growth is also characterized by the presence of ischemic and necrotic areas generated by oxygenation gradients and differential access to nutrients. Hypoxia and necrosis play key roles in tumor progression and resistance to treatment. To provide in vitro models recapitulating these events in highly controlled and standardized conditions, we have generated colorectal cancer (CRC) cell spheroids of different sizes and analyzed their gene expression profiles and sensitivity to treatment with 5FU, currently used in therapeutic protocols. Here we identify three MCTS stages, corresponding to defined spheroid sizes, characterized by normoxia, hypoxia, and hypoxia plus necrosis, respectively. Importantly, we show that MCTS including both hypoxic and necrotic areas most closely mimic gene expression profiles of in vivo-developing tumors and display the highest resistance to 5FU. Taken together, our data indicate that MCTS may mimic in vitro generation of ischemic and necrotic areas in highly standardized and controlled conditions, thereby qualifying as relevant models for drug screening purposes.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Hypoxia/physiology , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/physiology , Fluorouracil/pharmacology , Necrosis/pathology , Spheroids, Cellular/physiology , Animals , Colorectal Neoplasms/drug therapy , Gene Expression Profiling , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oxygen/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Eur J Immunol ; 46(2): 420-31, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26561341

ABSTRACT

Central memory CD8(+) T cells (TCM ) play key roles in the protective immunity against infectious agents, cancer immunotherapy, and adoptive treatments of malignant and viral diseases. CD8(+) TCM cells are characterized by specific phenotypes, homing, and proliferative capacities. However, CD8(+) TCM -cell generation is challenging, and usually requires CD4(+) CD40L(+) T-cell "help" during the priming of naïve CD8(+) T cells. We have generated a replication incompetent CD40 ligand-expressing recombinant vaccinia virus (rVV40L) to promote the differentiation of human naïve CD8(+) T cells into TCM specific for viral and tumor-associated antigens. Soluble CD40 ligand recombinant protein (sCD40L), and vaccinia virus wild-type (VV WT), alone or in combination, were used as controls. Here, we show that, in the absence of CD4(+) T cells, a single "in vitro" stimulation of naïve CD8(+) T cells by rVV40L-infected nonprofessional CD14(+) antigen presenting cells promotes the rapid generation of viral or tumor associated antigen-specific CD8(+) T cells displaying TCM phenotypic and functional properties. These observations demonstrate the high ability of rVV40L to fine tune CD8(+) mediated immune responses, and strongly support the use of similar reagents for clinical immunization and adoptive immunotherapy purposes.


Subject(s)
CD40 Ligand/metabolism , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Immunotherapy, Adoptive/methods , Neoplasms/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccinia virus/immunology , Antigens, Neoplasm/immunology , Antigens, Viral/immunology , CD40 Ligand/genetics , Cell Differentiation , Cells, Cultured , Combined Modality Therapy , Humans , Immunologic Memory , Neoplasms/therapy , Vaccines, Synthetic/administration & dosage
6.
Biomaterials ; 62: 138-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26051518

ABSTRACT

Anticancer compound screening on 2D cell cultures poorly predicts "in vivo" performance, while conventional 3D culture systems are usually characterized by limited cell proliferation, failing to produce tissue-like-structures (TLS) suitable for drug testing. We addressed engineering of TLS by culturing cancer cells in porous scaffolds under perfusion flow. Colorectal cancer (CRC) HT-29 cells were cultured in 2D, on collagen sponges in static conditions or in perfused bioreactors, or injected subcutaneously in immunodeficient mice. Perfused 3D (p3D) cultures resulted in significantly higher (p < 0.0001) cell proliferation than static 3D (s3D) cultures and yielded more homogeneous TLS, with morphology and phenotypes similar to xenografts. Transcriptome analysis revealed a high correlation between xenografts and p3D cultures, particularly for gene clusters regulating apoptotic processes and response to hypoxia. Treatment with 5-Fluorouracil (5-FU), a frequently used but often clinically ineffective chemotherapy drug, induced apoptosis, down-regulation of anti-apoptotic genes (BCL-2, TRAF1, and c-FLIP) and decreased cell numbers in 2D, but only "nucleolar stress" in p3D and xenografts. Conversely, BCL-2 inhibitor ABT-199 induced cytotoxic effects in p3D but not in 2D cultures. Our findings advocate the importance of perfusion flow in 3D cultures of tumor cells to efficiently mimic functional features observed "in vivo" and to test anticancer compounds.


Subject(s)
Bioreactors , Drug Resistance, Neoplasm , Fluorouracil/therapeutic use , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Tissue Engineering/instrumentation , Antimetabolites, Antineoplastic/therapeutic use , Batch Cell Culture Techniques/instrumentation , Biomimetics/methods , Cell Proliferation/drug effects , Equipment Design , Equipment Failure Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Neoplasm Proteins/metabolism , Neoplasms, Experimental/drug therapy , Phenotype
7.
Ann Rheum Dis ; 74(1): 260-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24249810

ABSTRACT

OBJECTIVE: Human bone marrow mesenchymal stromal cells (hBM-MSC) are being applied in tissue regeneration and treatment of autoimmune diseases (AD). Their cellular and immunophenotype depend on isolation and culture conditions which may influence their therapeutic application and reflect their in vivo biological functions. We have further characterised the phenotype induced by fibroblast growth factor 2 (FGF2) on healthy donor hBM-MSC focusing on the osteoimmunological markers osteoprotegerin (OPG), receptor activator of nuclear factor kB (RANK), RANK ligand (RANKL) and HLA-DR and their regulation of expression by the inflammatory cytokines IL1ß and IFNγ. METHODS: RANK, RANKL, OPG and HLA-DR expression in hBM-MSC expanded under specific culture conditions, were measured by RT-PCR and flow cytometry. MAPKs induction by FGF2, IL1ß and IFNγ in hBM-MSC was analysed by immunoblotting and RT-PCR. RESULTS: In hBM-MSC, OPG expression is constitutive and FGF2 independent. RANKL expression depends on FGF2 and ERK1/2 activation. IL1ß and IFNγ activate ERK1/2 but fail to induce RANKL. Only IL1ß induces P38MAPK. The previously described HLA-DR induced by FGF2 through ERK1/2 on hBM-MSC, is suppressed by IL1ß through inhibition of CIITA transcription. HLA-DR induced by IFNγ is not affected by IL1ß in hBM-MSC, but is suppressed in articular chondrocytes and lung fibroblasts. CONCLUSIONS: RANKL expression and IL1ß regulated MHC-class II, both induced via activation of the ERK1/2 signalling pathway, are specific for progenitor hBM-MSC expanded in the presence of FGF2. HLA-DR regulated by IL1ß and ERK1/2 is observed on hBM-MSC during early expansion without FGF2 suggesting previous in vivo acquisition. Stromal progenitor cells with this phenotype could have an osteoimmunological role during bone regeneration.


Subject(s)
Bone Marrow Cells/metabolism , Fibroblast Growth Factor 2/immunology , HLA-DR Antigens/genetics , Interferon-gamma/immunology , Interleukin-1beta/immunology , Mesenchymal Stem Cells/metabolism , Osteoprotegerin/genetics , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Fibroblast Growth Factor 2/pharmacology , Gene Expression/drug effects , Gene Expression Profiling , HLA-DR Antigens/drug effects , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Osteoprotegerin/drug effects , Osteoprotegerin/metabolism , RANK Ligand/drug effects , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B/drug effects , Reverse Transcriptase Polymerase Chain Reaction
8.
Clin Cancer Res ; 20(12): 3094-106, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24737547

ABSTRACT

PURPOSE: Colorectal cancer infiltration by CD16(+) myeloid cells correlates with improved prognosis. We addressed mechanistic clues and gene and protein expression of cytokines potentially associated with macrophage polarization. EXPERIMENTAL DESIGN: GM-CSF or M-CSF-stimulated peripheral blood CD14(+) cells from healthy donors were cocultured with colorectal cancer cells. Tumor cell proliferation was assessed by (3)H-thymidine incorporation. Expression of cytokine genes in colorectal cancer and autologous healthy mucosa was tested by quantitative, real-time PCR. A tumor microarray (TMA) including >1,200 colorectal cancer specimens was stained with GM-CSF- and M-CSF-specific antibodies. Clinicopathological features and overall survival were analyzed. RESULTS: GM-CSF induced CD16 expression in 66% ± 8% of monocytes, as compared with 28% ± 1% in cells stimulated by M-CSF (P = 0.011). GM-CSF but not M-CSF-stimulated macrophages significantly (P < 0.02) inhibited colorectal cancer cell proliferation. GM-CSF gene was expressed to significantly (n = 45, P < 0.0001) higher extents in colorectal cancer than in healthy mucosa, whereas M-CSF gene expression was similar in healthy mucosa and colorectal cancer. Accordingly, IL1ß and IL23 genes, typically expressed by M1 macrophages, were expressed to significantly (P < 0.001) higher extents in colorectal cancer than in healthy mucosa. TMA staining revealed that GM-CSF production by tumor cells is associated with lower T stage (P = 0.02), "pushing" growth pattern (P = 0.004) and significantly (P = 0.0002) longer survival in mismatch-repair proficient colorectal cancer. Favorable prognostic effect of GM-CSF production by colorectal cancer cells was confirmed by multivariate analysis and was independent from CD16(+) and CD8(+) cell colorectal cancer infiltration. M-CSF expression had no significant prognostic relevance. CONCLUSIONS: GM-CSF production by tumor cells is an independent favorable prognostic factor in colorectal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/mortality , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophages/pathology , Monocytes/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Proliferation , Chemokines/genetics , Chemokines/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Immunocompetence , Immunoenzyme Techniques , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Male , Middle Aged , Monocytes/metabolism , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Real-Time Polymerase Chain Reaction , Survival Rate , Tissue Array Analysis
9.
Int J Cancer ; 131(5): E659-69, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22116674

ABSTRACT

Herpes simplex virus protein ICP47, encoded by US12 gene, strongly downregulates major histocompatibility complex (MHC) class-I antigen restricted presentation by blocking transporter associated with antigen processing (TAP) protein. To decrease viral vector antigenic immunodominance and MHC class-I driven clearance, we engineered recombinant vaccinia viruses (rVV) expressing ICP47 alone (rVV-US12) or together with endoplasmic reticulum (ER)-targeted Melan-A/MART-1(27-35) model tumor epitope (rVV-MUS12). In this study, we show that antigen presenting cells (APC), infected with rVV-US12, display a decreased ability to present TAP dependent MHC class-I restricted viral antigens to CD8+ T-cells. While HLA class-I cell surface expression is strongly downregulated, other important immune related molecules such as CD80, CD44 and, most importantly, MHC class-II are unaffected. Characterization of rVV-MUS12 infected cells demonstrates that over-expression of a TAP-independent peptide, partially compensates for ICP47 induced surface MHC class-I downregulation (30% vs. 70% respectively). Most importantly, in conditions where clearance of infected APC by virus-specific CTL represents a limiting factor, a significant enhancement of CTL responses to the tumor epitope can be detected in cultures stimulated with rVV-MUS12, as compared to those stimulated by rVV-MART alone. Such reagents could become of high relevance in multiple boost protocols required for cancer immunotherapy, to limit vector-specific responsiveness.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigen-Presenting Cells/immunology , Cancer Vaccines/immunology , Epitopes/immunology , HLA-A2 Antigen/immunology , Immediate-Early Proteins/immunology , Immediate-Early Proteins/metabolism , Vaccinia virus/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , Antigen Presentation , Antigens, Viral/immunology , B7-1 Antigen , Cell Proliferation , Cells, Cultured , Cytokines , Endoplasmic Reticulum , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Genetic Vectors , HLA-DR Antigens , Humans , Hyaluronan Receptors , Immediate-Early Proteins/genetics , MART-1 Antigen/immunology , MART-1 Antigen/metabolism , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , T-Lymphocytes, Cytotoxic/immunology
10.
J Cell Physiol ; 227(1): 88-97, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22025108

ABSTRACT

In this study, we aimed at investigating the interactions between primary chondrocytes and mesenchymal stem/stromal cells (MSC) accounting for improved chondrogenesis in coculture systems. Expanded MSC from human bone marrow (BM-MSC) or adipose tissue (AT-MSC) were cultured in pellets alone (monoculture) or with primary human chondrocytes from articular (AC) or nasal (NC) cartilage (coculture). In order to determine the reached cell number and phenotype, selected pellets were generated by combining: (i) human BM-MSC with bovine AC, (ii) BM-MSC from HLA-A2+ with AC from HLA-A2- donors, or (iii) human green fluorescent protein transduced BM-MSC with AC. Human BM-MSC and AC were also cultured separately in transwells. Resulting tissues and/or isolated cells were assessed immunohistologically, biochemically, cytofluorimetrically, and by RT-PCR. Coculture of NC or AC (25%) with BM-MSC or AT-MSC (75%) in pellets resulted in up to 1.6-fold higher glycosaminoglycan content than what would be expected based on the relative percentages of the different cell types. This effect was not observed in the transwell model. BM-MSC decreased in number (about fivefold) over time and, if cocultured with chondrocytes, increased type II collagen and decreased type X collagen expression. Instead, AC increased in number (4.2-fold) if cocultured with BM-MSC and maintained a differentiated phenotype. Chondro-induction in MSC-chondrocyte coculture is a robust process mediated by two concomitant effects: MSC-induced chondrocyte proliferation and chondrocyte-enhanced MSC chondrogenesis. The identified interactions between progenitor and mature cell populations may lead to the efficient use of freshly harvested chondrocytes for ex vivo cartilage engineering or in situ cartilage repair.


Subject(s)
Cell Communication/physiology , Chondrocytes/cytology , Chondrogenesis/physiology , Mesenchymal Stem Cells/cytology , Adolescent , Adult , Animals , Cattle , Cell Proliferation , Cells, Cultured , Chondrocytes/metabolism , Coculture Techniques/methods , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
11.
Int J Cancer ; 129(5): 1137-48, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21710496

ABSTRACT

MAGE-A10 is a highly immunogenic member of the MAGE-A family of cancer/testis tumor-associated antigens (C/T TAAs). Studies performed with broadly reactive antibodies have helped to initially characterize this TAA. However, no specific reagents have been developed so far, thus preventing a thorough analysis of its expression in healthy and tumoral tissues. We have produced MAGE-A10 gene product in soluble recombinant form, and we have used it to generate specific monoclonal antibodies (mAbs). One of these reagents, recognizing an epitope located at the COOH terminus of the MAGE-A10 gene product, was used to stain a multitumor tissue microarray comprising more than 2,500 paraffin-embedded specimens including healthy tissues, benign tumors and malignancies of different histological origin. MAGE-A10 protein was identified as an intranuclear protein of an apparent molecular weight of 70 kDa, expressed in normal spermatogonia and spermatocytes but in no other healthy tissue. Most importantly, this C/T TAA appears to be expressed in high (>50%) percentages of cancer cells from a number of malignancies, including lung, skin and urothelial tumors. Unexpectedly, high expression of MAGE-A10 TAA at the protein level was also detectable in gynecological malignancies and stomach and gall bladder cancers. The characterization of MAGE-A10-specific reagents might set the stage for the development of targeted active immunotherapy by clarifying potential indications and by allowing the selection of patients eligible for treatment and the monitoring of its effectiveness.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Nucleus/metabolism , Lung Neoplasms/metabolism , Neoplasm Proteins/metabolism , Skin Neoplasms/metabolism , Urologic Neoplasms/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Nucleus/genetics , Female , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tissue Array Analysis , Tumor Cells, Cultured , Urologic Neoplasms/genetics , Urologic Neoplasms/immunology
12.
Arthritis Rheum ; 62(12): 3815-25, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20824797

ABSTRACT

OBJECTIVE: To document the specificity and the mechanism of induction of a novel class II major histocompatibility complex (MHC) antigen by mitogenic growth factors in human mesenchymal stem cells (MSCs) expanded in vitro for translational applications. METHODS: Expression of class II MHC molecules was measured in human MSCs and differentiated cells expanded in the presence of fibroblast growth factor 2 (FGF-2), platelet-derived growth factor BB (PDGF-BB), human platelet lysate, or interferon-γ (IFNγ). The roles of cell proliferation and growth factor-induced signaling pathways were investigated as well as the class II MHC assembly machinery and functional capacity. RESULTS: FGF-2 and, to a lesser extent, PDGF-BB induced in adult human MSCs the expression of HLA-DR (normally induced by inflammatory cytokines), which was able to stimulate CD4+ T cells via superantigen binding. In contrast to IFNγ, FGF induced HLA-DR expression only in human MSCs proliferating under its mitogenic effect and not in mouse MSCs or in differentiated human cells. Although it induced cell proliferation, human platelet lysate did not cause HLA-DR expression in human MSCs. HLA-DR expression occurred following FGF-specific binding to its receptor(s), mainly FGF receptor 1, without inducing IFNγ or tumor necrosis factor α expression. Both MAPK/ERK-1/2 and phosphatidylinositol 3-kinase/Akt controlled cell proliferation and HLA-DR expression, but only MAPK/ERK-1/2 controlled the induction of the class II MHC transcription activator protein CIITA, the major determinant of HLA-DR transcription. CONCLUSION: The induction of functional HLA-DR in proliferating progenitor MSCs is a property of human MSCs that have been expanded with mitogenic growth factors. This has potential biologic significance in the regulation and/or protection of progenitor cell subpopulations under sustained mitogenic proliferation and needs to be taken into account when expanding MSCs for use in in vivo applications.


Subject(s)
Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Histocompatibility Antigens Class II/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Platelet-Derived Growth Factor/pharmacology , Cells, Cultured , Chromones/pharmacology , Flavonoids/pharmacology , HLA-DR Antigens/metabolism , Humans , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Morpholines/pharmacology , Nuclear Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quinoxalines/pharmacology , Thiazolidinediones/pharmacology , Trans-Activators/metabolism
13.
Curr Opin Drug Discov Devel ; 13(2): 184-92, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20205052

ABSTRACT

The possibility of using the immune system of patients to control tumor outgrowth in a therapeutic setting has always been highly appealing to both clinicians and researchers. However, although cancer cells express tumor-associated antigens that can be targeted by T-cells, clinical trials suggest that the induction of specific immune responses per se may be insufficient to achieve clinical goals. Based on these trial data, in addition to experimental data revealing the complexity of mechanisms controlling immune responsiveness, a reassessment of immunotherapy procedures is underway. As a result, a second generation of antitumor treatments that includes reagents of potential pharmaceutical relevance is being developed. In this review, the most recent literature addressing issues related to immunotherapy for solid tumors is discussed.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Animals , Cancer Vaccines/therapeutic use , Combined Modality Therapy/methods , Humans
14.
Mol Ther ; 18(3): 651-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19935776

ABSTRACT

Recombinant vaccinia virus (rVV) encoding tumor-associated antigens (TAAs) and adhesion or costimulatory molecules may represent important immunogenic reagents for cancer immunotherapy. Recently, intranodal (IN) antigen administration was suggested to be more immunogenic than intradermal (ID) vaccination. However, IN rVV administration has not been attempted so far. We used a rVV encoding gp100(280-288), Melan-A/MART-1(27-35) and tyrosinase(1-9) HLA-A0201 restricted epitopes and CD80 and CD86 costimulatory molecules in stage III and IV melanoma patients in a phase 1/2 trial. Of 15 patients initiating treatment, including two cycles of IN immunization, each comprising one rVV administration and three recall injections of the corresponding peptides, accompanied by subcutaneous granulocyte macrophage-colony stimulating factor supplementation, five withdrew due to progressing disease. Of 10 remaining patients seven showed evidence of induction of cytotoxic T lymphocytes (CTLs) directed against at least one epitope under investigation, as detectable by limiting dilution analysis (LDA) of specific precursors and multimer staining. Adverse reactions were mild (National Cancer Institute (NCI) grade 1-2) and mainly represented by fever, skin rashes, and pruritus. These data indicate that IN administration of rVV encoding melanoma-associated epitopes and costimulatory molecules is safe and immunogenic.


Subject(s)
Antigens, Neoplasm/metabolism , Immunization/methods , Melanoma/pathology , Melanoma/therapy , Vaccinia virus/metabolism , Adult , Aged , Aged, 80 and over , Cancer Vaccines/therapeutic use , Disease Progression , Epitopes/chemistry , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymph Nodes/pathology , Male , Middle Aged , Neoplasm Metastasis , T-Lymphocytes, Cytotoxic/metabolism
15.
J Immunother ; 32(3): 252-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19242375

ABSTRACT

Common receptor gamma chain (c-gamma) cytokines (CKs) support proliferation of CD8+ T cells in presence or absence of antigen triggering and help maintaining the immunologic memory. We addressed the effects of low (< or = 5 ng/mL)-dose interleukin (IL)-2, IL-7, or IL-15 on human naive and memory antigen-specific CD8+ T cells. Peripheral blood CD8+ lymphocytes proliferated with decreasing efficiency in response to IL-15, IL-7, and IL-2. Of note, IL-15 preferentially promoted expansion of CD45RA/CD8+ T-cell memory subset. Accordingly, cytotoxic T lymphocytes specific for cytomegalovirus-derived antigens from seropositive donors proliferated in response to IL-15 and, to lesser extent to IL-7, but poorly to IL-2. CD8+ T cells were then pretreated with CK before antigen stimulation using, as read out, specific cytotoxic activity. After the pretreatment with IL-15, but not IL-2, previously experienced viral antigens induced vigorous cytotoxic responses. Minor effects of IL-7 were also detectable. In contrast, IL-2 best supported the cytotoxic T lymphocyte generation from prevailingly naive CD8 T cells from HLA-A*0201 healthy donors, specific for L27Melan-A/MART-126-35 melanoma-associated antigen. Cells from melanoma patients were tested before and after Melan-A/MART-1-targeted antigen-specific immunotherapy. Untreated patients showed heterogeneous patterns of responsiveness to c-gamma CK. However, when naive patients whose CD8+ T cells best responded to IL-2 were vaccinated, a modified responsiveness pattern was detectable. After immunization, cells displayed a significantly higher response to IL-15 than to IL-2 pretreatment. Thus, responsiveness to c-gamma CK is critically influenced by naive or memory status of peripheral blood CD8+ T cells.


Subject(s)
Interleukin Receptor Common gamma Subunit/immunology , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Interleukin-7/pharmacology , Melanoma/immunology , T-Lymphocytes, Cytotoxic/drug effects , Antigens, Viral/immunology , Antigens, Viral/metabolism , Humans , Immunologic Memory , Immunotherapy , Peptides/immunology , Peptides/metabolism , T-Lymphocytes, Cytotoxic/immunology
16.
Methods Mol Biol ; 514: 151-62, 2009.
Article in English | MEDLINE | ID: mdl-19048219

ABSTRACT

The following method describes the generation of a recombinant vaccinia virus expressing a costimulatory molecule (human CD80 or B7.1).The procedure first requires the cloning, by classical methods not described here, of the gene of interest, e.g. CD80, into a vaccinia shuttle plasmid under the control of a virus-specific promoter enabling a transcription during the early phase of infection. Flanking the insert, the plasmid contains viral sequences and a selection maker needed for the insertion into the viral genome. The successive plaque isolation of recombinant virus on cell monolayer described here is based on the transient "gpt" selection system which enables other insertions in different loci of the same virus. Finally, after verification amplification and titration of the recombinant vector, replication will be impaired by a psoralen-UV treatment in order to produce a non-replicating virus. Expression and function of inserts, following infection of cells, are verified by specific phenotypic and functional assays.


Subject(s)
B7-1 Antigen/genetics , B7-1 Antigen/metabolism , Genetic Engineering , Immunologic Techniques , Lymphocyte Activation , T-Lymphocytes/immunology , Vaccinia virus/genetics , Cloning, Molecular , DNA, Recombinant/genetics , Electrophoresis, Agar Gel , Gene Expression Regulation , Humans , Plasmids/genetics , T-Lymphocytes/cytology , Titrimetry , Transfection , Vaccinia virus/physiology , Virus Activation , Virus Replication
17.
J Transl Med ; 6: 58, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18925935

ABSTRACT

BACKGROUND: Monitoring of cellular immune responses is indispensable in a number of clinical research areas, including microbiology, virology, oncology and autoimmunity. Purification and culture of peripheral blood mononuclear cells and rapid access to specialized equipment are usually required. We developed a whole blood (WB) technique monitoring antigen specific cellular immune response in vaccinated or naturally sensitized individuals. METHODS: WB (300 microl) was incubated at 37 degrees C with specific antigens, in the form of peptides or commercial vaccines for 5-16 hours. Following RNAlater addition to stabilize RNA, the mixture could be stored over one week at room temperature or at 4 degrees C. Total RNA was then extracted, reverse transcribed and amplified in quantitative real-time PCR (qRT-PCR) assays with primers and probes specific for cytokine and/or chemokine genes. RESULTS: Spiking experiments demonstrated that this technique could detect antigen specific cytokine gene expression from 50 cytotoxic T lymphocytes (CTL) diluted in 300 microl WB. Furthermore, the high sensitivity of this method could be confirmed ex-vivo by the successful detection of CD8+ T cell responses against HCMV, EBV and influenza virus derived HLA-A0201 restricted epitopes, which was significantly correlated with specific multimer staining. Importantly, a highly significant (p = 0.000009) correlation between hepatitis B surface antigen (HBsAg) stimulated IL-2 gene expression, as detectable in WB, and specific antibody titers was observed in donors vaccinated against hepatitis B virus (HBV) between six months and twenty years before the tests. To identify additional markers of potential clinical relevance, expression of chemokine genes was also evaluated. Indeed, HBsAg stimulated expression of MIP-1beta (CCL4) gene was highly significantly (p = 0.0006) correlated with specific antibody titers. Moreover, a longitudinal study on response to influenza vaccine demonstrated a significant increase of antigen specific IFN-gamma gene expression two weeks after immunization, declining thereafter, whereas increased IL-2 gene expression was still detectable four months after vaccination. CONCLUSION: This method, easily amenable to automation, might qualify as technology of choice for high throughput screening of immune responses to large panels of antigens from cohorts of donors. Although analysis of cytokine gene expression requires adequate laboratory infrastructure, initial antigen stimulation and storage of test probes can be performed with minimal equipment and time requirements. This might prove important in "field" studies with difficult access to laboratory facilities.


Subject(s)
Antigens, Viral/immunology , Reverse Transcriptase Polymerase Chain Reaction/methods , T-Lymphocytes, Cytotoxic/immunology , Cell Separation , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Epitopes/immunology , Gene Expression Regulation , Health , Histocompatibility Antigens Class I/immunology , Humans , Time Factors , Viral Vaccines/immunology
18.
J Immunother ; 31(5): 466-74, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18463538

ABSTRACT

Dendritic cells (DC) can be activated by proinflammatory cytokines or upon toll-like receptor (TLR) triggering. These stimuli induce specific patterns of phenotypic modulation and gene expression profiles. We investigated whether TLR triggering represents an indispensable requirement for the induction of T cell responses by human DC generated upon culture of monocytes in the presence of granulocyte macrophage colony-stimulating factor and interferon-alpha (IFN-DC). As model stimulator we chose imidazoquinolone (3M-001), a synthetic TLR7 agonist used in the treatment of skin infections and tumors and as experimental adjuvant. At difference with DC generated upon culture of monocytes in the presence of granulocyte macrophage colony-stimulating factor and interleukin (IL-4) (IL-4-DC), IFN-DC display a semimature phenotype. Furthermore, IFN-DC, but not IL-4-DC are able to induce CD4+ and CD8+ T cell responses, in steady state, for example, in the absence of TLR triggering. 3M-001 treatment induces up-regulation of the surface expression of costimulatory molecules and "de novo" production of IL-12 and IL-6 in IFN-DC. However, TLR7 triggering fails to significantly enhance the capacity of IFN-DC to induce antigen-specific cytotoxic T lymphocytes and to stimulate allogeneic CD4+ T cells. These data indicate that TLR engagement and IL-12 production do not represent indispensable prerequisites for optimal antigen-presenting cell function in IFN-DC, qualifying these cells as powerful cellular reagents of potential use in active specific immunotherapy.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Interferon-alpha/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Dendritic Cells/cytology , Humans , Interferon-alpha/immunology , Interleukin-4/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Phenotype , Toll-Like Receptors/metabolism
19.
Contemp Clin Trials ; 29(2): 165-81, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17707139

ABSTRACT

To the exception of early stages of disease, the morbidity and mortality of melanoma is considerable, with no acknowledged therapeutic options beyond surgery. Immunotherapy of melanoma has achieved some success, but further refinements are urgently needed in order to realize its potential. This paper describes a multi-centre phase I/II open labeled, controlled clinical trial investigating 2 innovative immunotherapeutic reagents. Two successive groups of 20 resected AJCC stages IIb-IV melanoma patients will be treated, first with melanoma epitopes included into Influenza virosomes (group 1), and second with a heterologous prime-boost protocol priming with a recombinant Vaccinia virus, and boosting with Influenza virosomes (group 2). Five melanoma epitopes from three different melanoma differentiation antigens were included into Influenza virosomes, that cross-stimulate CD4+ T cells and are endowed with high adjuvant capacity in the generation of CTL. The same five melanoma epitopes, two co-stimulatory molecules CD80 and CD86, and the CD40 ligand, a marker known to play a crucial role in CTL generation and memory maintenance were encoded in a recombinant Vaccinia virus. GM-CSF will be administered as a supporting cytokine. Both Influenza virosomes and octo-recombinant Vaccinia virus are innovative and original constructs assessed for the first time in human. Immunotherapy foresees 12 weekly immunizations for each group. Toxicity and adverse events will be monitored clinically. Immunological efficacy will be assessed dynamically by ex-vivo multimer analysis, Elispot, and quantitative real-time PCR for up to 3 months following completion of immunotherapy schedule. Disease free survival will be assessed by 4-monthly serial clinic visits, including physical and FDG-PET examinations, for a follow-up time of 2 years. Quality of life will be assessed with a dedicated FACT-BRM 4 questionnaire.


Subject(s)
Epitopes/immunology , Immunization, Secondary/methods , Immunotherapy, Active/methods , Melanoma/immunology , Melanoma/therapy , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Vaccines, Synthetic/administration & dosage , Vaccinia virus/immunology , Viral Vaccines/administration & dosage , Adolescent , CD4-Positive T-Lymphocytes/immunology , Disease-Free Survival , Humans , Immunization/methods , Immunization Schedule , Immunologic Memory/immunology , Melanoma/mortality , Orthomyxoviridae , Quality of Life , Skin Neoplasms/mortality , Virosomes
20.
J Invest Dermatol ; 127(3): 638-45, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17068481

ABSTRACT

Buruli ulcer (BU) caused by Mycobacterium ulcerans is a chronic necrotizing disease of the skin and the underlying soft tissue. Fat tissue necrosis accompanied by minimal inflammation is considered the most reliable histopathologic feature of BU. There may be a constant influx of inflammatory cells to the sites of active infection but these are thought to be killed by mycolactone, a polyketide toxin produced by M. ulcerans, through apoptosis and necrosis. Here we describe the spatial correlations between mycobacterial load and the expression of dendritic cell (DC) surface markers (cluster of differentiation (CD)83, CD11c, and CD123), the Toll-like receptor (TLR) 9 and pro- and anti-inflammatory cytokines (IL-8, IL-6, tumor necrosis factor-alpha (TNF-alpha), IFN-alpha, IL-12p40, IL-10, and IFN-gamma) within BU lesions. Although IL-8, IL-6, and TNF-alpha messenger RNA (mRNA) was detectable by real-time PCR in all lesions, the expression of the other cytokines was only found as small foci in some lesions. Correlations of the distribution of mRNA encoding the activation marker CD83 and the DC subset markers CD123 and CD11c indicate that both activated plasmacytoid and myeloid dendritic cells were present in the lesions. Results suggest that M. ulcerans specific immune responses may develop once therapeutic interventions have limited the production of mycolactone.


Subject(s)
Mycobacterium ulcerans/metabolism , Skin Diseases, Bacterial/immunology , Skin Ulcer/immunology , Skin Ulcer/pathology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Immune System/pathology , Immunohistochemistry , Inflammation , Necrosis , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...