Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell Death Dis ; 5: e1135, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24651438

ABSTRACT

Tumor microenvironment of solid tumors is characterized by a strikingly high concentration of adenosine and ATP. Physiological significance of this biochemical feature is unknown, but it has been suggested that it may affect infiltrating immune cell responses and tumor progression. There is increasing awareness that many of the effects of extracellular ATP on tumor and inflammatory cells are mediated by the P2X7 receptor (P2X7R). Aim of this study was to investigate whether: (i) extracellular ATP is a component of neuroblastoma (NB) microenvironment, (ii) myeloid-derived suppressor cells (MDSCs) express functional P2X7R and (iii) the ATP/P2X7R axis modulates MDSC functions. Our results show that extracellular ATP was detected in NB microenvironment in amounts that increased in parallel with tumor progression. The percentage of CD11b(+)/Gr-1(+) cells was higher in NB-bearing mice compared with healthy animals. Within the CD11b/Gr-1(+) population, monocytic MDSCs (M-MDSCs) produced higher levels of reactive oxygen species (ROS), arginase-1 (ARG-1), transforming growth factor-ß1 (TGF-ß1) and stimulated more potently in vivo tumor growth, as compared with granulocytic MDSCs (G-MDSCs). P2X7R of M-MDSCs was localized at the plasma membrane, coupled to increased functionality, upregulation of ARG-1, TGF-ß1 and ROS. Quite surprisingly, the P2X7R in primary MDSCs as well as in the MSC-1 and MSC-2 lines was uncoupled from cytotoxicity. This study describes a novel scenario in which MDSC immunosuppressive functions are modulated by the ATP-enriched tumor microenvironment.


Subject(s)
Adenosine Triphosphate/metabolism , Myeloid Cells/metabolism , Neuroblastoma/metabolism , Receptors, Purinergic P2X7/metabolism , Tumor Microenvironment , Animals , Arginase/metabolism , Biomarkers, Tumor/metabolism , CD11b Antigen/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , HEK293 Cells , Humans , Mice , Myeloid Cells/immunology , Myeloid Cells/pathology , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/pathology , Reactive Oxygen Species/metabolism , Receptors, Chemokine/metabolism , Receptors, Purinergic P2X7/genetics , Signal Transduction , Transfection , Transforming Growth Factor beta1/metabolism , Tumor Escape
2.
Scand J Rheumatol ; 40(2): 139-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20919945

ABSTRACT

OBJECTIVES: To compare the distribution of human leucocyte antigen (HLA) class I and II alleles in patients with erosive hand osteoarthritis (EHOA) to that of patients with non-erosive hand OA (non-EHOA) and in healthy Italian Bone Marrow Donors (IBMDs), in order to evaluate possible immunogenetic associations with EHOA. In the EHOA group we also sought possible associations between HLA alleles and disease severity. METHODS: Ninety-four patients with EHOA (82 women, 12 men; mean age 61.4 ± 8.45 years) and 37 with non-EHOA (28 women, nine men; mean age 59.21 ± 9.07 years) were studied. Disease severity was measured by the number of clinically active joints (NCAJ) and by the radiographic score (RS) using the Kallman scale. HLA typing was undertaken for A, B, C, and DRB1 loci; HLA-DRB1* genotyping was determined using polymerase chain reaction (PCR) with sequence-specific primers. Frequencies were compared with those of the healthy IBMDs. RESULTS: The alleles found more frequently in EHOA patients than in non-EHOA patients and healthy controls were: A23, A26, and A29; B38, B44, and HLA DRB1*01 and *07. The RS was more severe in the EHOA compared to the non-EHOA group (63.60 ± 23.14 vs. 34.34 ± 20.24, p < 0.001). Within the EHOA group, HLA-DRB1*07 was associated with a higher RS (67.36 ± 23 vs. 64.5 ± 18.5, p = 0.029). CONCLUSION: In this study of North Italian patients affected with EHOA, the HLA-DRB1*07 allele was found to be associated with both the development and greater severity of the disease.


Subject(s)
Hand Joints/physiopathology , Immunogenetic Phenomena/genetics , Immunogenetic Phenomena/physiology , Osteoarthritis/ethnology , Osteoarthritis/genetics , Aged , Case-Control Studies , Female , Gene Frequency/genetics , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans , Italy , Male , Middle Aged , Osteoarthritis/physiopathology , Severity of Illness Index
3.
Cell Transplant ; 20(6): 941-54, 2011.
Article in English | MEDLINE | ID: mdl-21054938

ABSTRACT

Natural CD4(+)Foxp3(+) T regulatory (Treg) cells can promote transplantation acceptance across major histocompatibility complex (MHC) barriers, while myeloid-derived suppressor cells (MDSCs) inhibit effector T-cell responses in tumor-bearing mice. One outstanding issue is whether combining the potent suppressive function of MDSCs with that of Treg cells might synergistically favor graft tolerance. In the present study, we evaluated the therapeutic potential of MDSCs and natural Treg cells in promoting allograft tolerance in mice by utilizing immunomodulatory agents to expand these cells in vivo. Upon administration of recombinant human granulocyte-colony stimulating factor (G-CSF; Neupogen), or interleukin-2 complex (IL-2C), Gr-1(+)CD11b(+) MDSCs or CD4(+)Foxp3(+) Treg cells were respectively induced at a high frequency in the peripheral lymphoid compartments of treated mice. Interestingly, induced MDSCs exhibited a more potent suppressive function in vitro when compared to MDSCs from naive mice. Furthermore, in vivo coadministration of Neupogen and IL-2C induced MDSCs at percentages that were higher than those seen when either agent was administered alone, suggesting an additive effect of the two drugs. Although treatment with either IL-2C or Neupogen led to a significant delay of MHC class II disparate allogeneic donor skin rejection, the combinatorial treatment was superior to either alone. Importantly, histological assessment of surviving grafts revealed intact morphology and minimal infiltrates at 60 days posttransplant. Collectively, our findings demonstrate that concurrent induction of MDSCs and Tregs is efficacious in downmodulating alloreactive T-cell responses in a synergistic manner and highlight the therapeutic potential of these naturally occurring suppressive leukocytes to promote transplantation tolerance.


Subject(s)
CD4 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Myeloid Cells/cytology , T-Lymphocytes, Regulatory/cytology , Animals , Filgrastim , Graft Survival , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Interleukin-2/pharmacology , Mice , Models, Animal , Phenotype , Recombinant Proteins/pharmacology , Skin/pathology , Skin Transplantation , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
4.
Cancer Res ; 61(3): 859-63, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11221870

ABSTRACT

To improve the immunogenicity of melanoma self-antigens, we used a novel strategy of nonviral genetic vaccination coupled with muscle electroporation. Electroporation-enhanced immunization with plasmids encoding either human gp100 or mouse TRP-2 antigens induced only partial rejection of B16 melanoma challenge. However, immunization with a combination of these two antigens caused tumor rejection in 100% of the immunized mice. Splenocytes from combination-immunized animals killed syngeneic targets loaded with peptides derived from both gp100 and TRP-2. Immune cell depletion experiments identified CD8+ T lymphocytes as the primary effectors of antitumor immunity. Most importantly, polyimmunization led to the generation of a therapeutic immune response that significantly improved the mean survival time of mice bearing established lung metastases. These results validated the usefulness of electroporation-enhanced, nonviral genetic immunization for the active immunotherapy of cancer and indicated that using a combination of different tumor antigens may be a decisive strategy for a successful therapeutic vaccination.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Animals , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Electroporation , Female , Intramolecular Oxidoreductases/genetics , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Melanoma, Experimental/prevention & control , Melanoma, Experimental/secondary , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Neoplasm Proteins/genetics , Neoplasm Transplantation , Plasmids/administration & dosage , Plasmids/genetics , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccines, Combined/genetics , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use , gp100 Melanoma Antigen
5.
Curr Gene Ther ; 1(1): 53-100, 2001 May.
Article in English | MEDLINE | ID: mdl-12109138

ABSTRACT

Molecular biology techniques have given novel impetus to the immunotherapy of cancer because they have catalyzed the identification of several potential tumor antigens, and permitted the generation of vectors for the delivery of genetic material encoding these antigens. Vaccines can be defined "genetic" when the antigen they enclose is present as DNA or RNA. Microrganisms used as vectors can deliver the genetic information, but naked nucleic acids have also been shown to be effective immunogens thanks to built-in adjuvants that activate professional antigen presenting cells. Although gene-based cancer vaccines have been tested in mouse models and selected for pilot clinical trials, enthusiasm has somewhat waned due to an apparently major drawback of cancer vaccination: tumor antigens are weak, and therefore fail to stimulate a sterilizing immune response in tumor-bearing patients. Mouse studies, however, have shown that cancer vaccines are extremely efficacious in establishing a state of active immunosurvellance against tumor growth. This review reconsiders the findings emerging from preclinical studies in the context of our current knowledge of the cellular and molecular bases of the immune responses to vaccines, in an attempt to approach critically the use of genetic vaccination for the treatment of cancer.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy, Active/methods , Neoplasms/therapy , Adenoviridae/genetics , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer/methods , Alphavirus/genetics , Animals , Bacteria/genetics , Bacteria/immunology , Genetic Vectors , Humans , Mice , Mice, Transgenic , Neoplasms/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Poxviridae/genetics , RNA/genetics , RNA/therapeutic use , Vaccines, DNA/genetics , Vaccines, DNA/therapeutic use
6.
J Immunother ; 24(6): 431-46, 2001.
Article in English | MEDLINE | ID: mdl-11759067

ABSTRACT

In the late 1970s, several findings suggested that accessory cells distinct from lymphocytes might suppress immune reactivity in tumor-bearing hosts. Studies in animal models and patients later confirmed that cells driven to act as dominant immune suppressors by growing cancers could subvert the immune system. These cells have also been termed natural suppressors, a functional definition connoting their ability to hamper various T- and B-lymphocyte responses without prior activation and independently from antigen and MHC restriction. These properties were attributed to distinct cell populations. The phenotypic discrepancies, together with the lack of antigen specificity, have generated serious restraints to research on tumor-induced suppression. Recent evidence indicates that suppressor cells are closely related to immature myeloid precursors and can be found in several situations that can exert adverse effects on the immunotherapy of cancer. The present review is an attempt to address the nature and properties of immature myeloid suppressors and their relationship to dendritic cells and macrophages, with the aim of clarifying the complex network of tumor-induced, negative regulators of the immune system.


Subject(s)
Immune Tolerance/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Animals , Humans , Receptors, Interleukin-2/immunology , Signal Transduction/immunology
7.
J Immunol ; 165(12): 6723-30, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11120790

ABSTRACT

We described a generalized suppression of CTL anamnestic responses that occurred in mice bearing large tumor nodules or immunized with powerful recombinant viral immunogens. Immune suppression entirely depended on GM-CSF-driven accumulation of CD11b(+)/Gr-1(+) myeloid suppressor cells (MSC) in secondary lymphoid organs. To further investigate the nature and properties of MSC, we immortalized CD11b(+)/Gr-1(+) cells isolated from the spleens of immunosuppressed mice, using a retrovirus encoding the v-myc and v-raf oncogenes. Immortalized cells expressed monocyte/macrophage markers (CD11b, F4/80, CD86, CD11c), but they differed from previously characterized macrophage lines in their capacities to inhibit T lymphocyte activation. Two MSC lines, MSC-1 and MSC-2, were selected based upon their abilities to inhibit Ag-specific proliferative and functional CTL responses. MSC-1 line was constitutively inhibitory, while suppressive functions of MSC-2 line were stimulated by exposure to the cytokine IL-4. Both MSC lines triggered the apoptotic cascade in Ag-activated T lymphocytes by a mechanism requiring cell-cell contact. Some well-known membrane molecules involved in the activation of apoptotic pathways (e.g., TNF-related apoptosis-inducing ligand, Fas ligand, TNF-alpha) were ruled out as candidate effectors for the suppression mechanism. The immortalized myeloid lines represent a novel, useful tool to shed light on the molecules involved in the differentiation of myeloid-related suppressors as well as in the inhibitory pathway they use to control T lymphocyte activation.


Subject(s)
Apoptosis/immunology , Cell Line, Transformed/immunology , Isoantigens/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Myeloid Cells/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , 3T3 Cells , Animals , Apoptosis Regulatory Proteins , Cell Communication/immunology , Cell Division/immunology , Cell Line, Transformed/metabolism , Cytotoxicity, Immunologic/immunology , Fas Ligand Protein , Female , Immune Tolerance , Immunophenotyping , Ligands , Lymphocyte Culture Test, Mixed , Macrophages/classification , Macrophages/metabolism , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloid Cells/classification , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/physiology , fas Receptor/metabolism
8.
Haematologica ; 85(11): 1172-206, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11074658

ABSTRACT

BACKGROUND AND OBJECTIVES: Vaccination is an effective medical procedure of preventive medicine based on the induction of a long-lasting immunologic memory characterized by mechanisms endowed with high destructive potential and specificity. In the last few years, identification of tumor-associated antigens (TAA) has prompted the development of different strategies for antitumor vaccination, aimed at inducing specific recognition of TAA in order to elicit a persistent immune memory that may eliminate residual tumor cells and protect recipients from relapses. In this review characterization of TAA, different potential means of vaccination in experimental models and preliminary data from clinical trials in humans have been examined by the Working Group on Hematopoietic Cells. EVIDENCE AND INFORMATION SOURCES: The method employed for preparing this review was that of informal consensus development. Members of the Working Group met four times and discussed the single points, previously assigned by the chairman, in order to achieve an agreement on different opinions and approve the final manuscript. Some of the authors of the present review have been working in the field of antitumor immunotherapy and have contributed original papers to peer-reviewed journals. In addition, the material examined in the present review includes articles and abstracts published in journals covered by the Science Citation Index and Medline. STATE OF THE ART: The cellular basis of antitumor immune memory consists in the generation and extended persistence of expanded populations of T- and B-lymphocytes that specifically recognize and react against TAA. The efficacy of the memory can be modulated by compounds, called "adjuvants", such as certain bacterial products and mineral oils, cytokines, chemokines, by monoclonal antibodies triggering co-stimulatory receptors. Strategies that have been shown in preclinical models to be efficient in protecting from tumor engraftment, or in preventing a tumor rechallenge, include vaccination by means of soluble proteins or peptides, recombinant viruses or bacteria as TAA genes vectors, DNA injection, tumor cells genetically modified to express co-stimulatory molecules and/or cytokines. The use of professional antigen-presenting cells, namely dendritic cells, either pulsed with TAA or transduced with tumor-specific genes, provides a useful alternative for inducing antitumor cytotoxic activity. Some of these approaches have been tested in phase I/II clinical trials in hematologic malignancies, such as lymphoproliferative diseases or chronic myeloid leukemia, and in solid tumors, such as melanoma, colon cancer, prostate cancer and renal cell carcinoma. Different types of vaccines, use of adjuvants, timing of vaccination as well as selection of patients eligible for this procedure are discussed in this review. PERSPECTIVES: Experimental models demonstrate the possibility of curing cancer through the active induction of a specific immune response to TAA. However, while pre-clinical research has identified several possible targets and strategies for tumor vaccination the clinical scenario is far more complex for a number of possible reasons. Since experimental data suggest that vaccination is more likely to be effective on small tumor burden, such as a minimal residual disease after conventional treatments, or tumors at an early stage of disease, better selection of patients will allow more reliable clinical results to be obtained. Moreover, a poor correlation is frequently observed between the ability of TAA to induce a T-cell response in vitro and clinical responses. Controversial findings may also be due to the techniques used for monitoring the immune status. Therefore, the development of reliable assays for efficient monitoring of the state of immunization of cancer patients against TAA is an important goal that will markedly improve the progress of antitumor vaccines. (ABSTRACT TRUNCATED)


Subject(s)
Cancer Vaccines , Adjuvants, Immunologic/therapeutic use , Animals , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/therapeutic use , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Clinical Trials as Topic , Humans
9.
Blood ; 96(12): 3838-46, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090068

ABSTRACT

Apoptotic death of CD8(+) T cells can be induced by a population of inhibitory myeloid cells that are double positive for the CD11b and Gr-1 markers. These cells are responsible for the immunosuppression observed in pathologies as dissimilar as tumor growth and overwhelming infections, or after immunization with viruses. The appearance of a CD11b(+)/Gr-1(+) population of inhibitory macrophages (iMacs) could be attributed to high levels of granulocyte-macrophage colony-stimulating factor (GM-CSF) in vivo. Deletion of iMacs in vitro or in vivo reversed the depression of CD8(+) T-cell function. We isolated iMacs from the spleens of immunocompromised mice and found that these cells were positive for CD31, ER-MP20 (Ly-6C), and ER-MP58, markers characteristic of granulocyte/monocyte precursors. Importantly, although iMacs retained their inhibitory properties when cultured in vitro in standard medium, suppressive functions could be modulated by cytokine exposure. Whereas culture with the cytokine interleukin 4 (IL-4) increased iMac inhibitory activity, these cells could be differentiated into a nonadherent population of fully mature and highly activated dendritic cells when cultured in the presence of IL-4 and GM-CSF. A common CD31(+)/CD11b(+)/Gr-1(+) progenitor can thus give rise to cells capable of either activating or inhibiting the function of CD8(+) T lymphocytes, depending on the cytokine milieu that prevails during antigen-presenting cell maturation. (Blood. 2000;96:3838-3846)


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Macrophage-1 Antigen/physiology , Macrophages/immunology , Mice, Inbred BALB C , Myeloid Progenitor Cells/immunology , Platelet Endothelial Cell Adhesion Molecule-1/physiology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Biomarkers/analysis , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cytokines/pharmacology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Female , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocytes , Immunosuppression Therapy , Interleukin-4/pharmacology , Isoantigens/immunology , Isoantigens/physiology , Lymphocyte Activation/drug effects , Macrophage-1 Antigen/immunology , Macrophages/cytology , Macrophages/drug effects , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Spleen/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured , Vaccinia virus
10.
Cancer Res ; 60(2): 253-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667570

ABSTRACT

"Self" melanocyte differentiation antigens are potential targets for specific melanoma immunotherapy. Vaccination against murine tyrosinase-related protein (TRP)-1/gp75 was shown recently to cause melanoma rejection, which was accompanied by autoimmune skin depigmentation (vitiligo). To further explore the linkage between immunotherapy and autoimmunity, we studied the response to vaccination with a related antigen, TRP-2. i.m. inoculation of plasmid DNA encoding murine trp-2 elicited antigen-specific CTLs that recognized the B16 mouse melanoma and protected the mice from challenge with tumor cells. Furthermore, mice bearing established s.c. B16 melanomas rejected the tumor upon vaccination with a recombinant vaccinia virus encoding trp-2. Depletion experiments showed that CD8+ lymphocytes and natural killer cells were crucial for the antitumor activity of the trp-2-encoding vaccines. Mice that rejected the tumor did not develop generalized vitiligo, indicating that protective immunity can be achieved in the absence of widespread autoimmune aggression.


Subject(s)
Cancer Vaccines/therapeutic use , Interferon Type I/immunology , Intramolecular Oxidoreductases/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Pregnancy Proteins/immunology , Vaccines, Synthetic/therapeutic use , Vitiligo/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/toxicity , Killer Cells, Natural/immunology , Lymphocyte Culture Test, Mixed , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptides/chemistry , Peptides/immunology , Vaccines, Synthetic/toxicity , Vitiligo/etiology
11.
J Immunother ; 22(6): 489-96, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10570747

ABSTRACT

Interleukin-10 (IL-10) has a wide range of in vivo biological activities and is a key regulatory cytokine of immune-mediated inflammation. The authors found that murine IL-10 given 12 hours after a recombinant vaccinia virus (rVV) containing the LacZ gene significantly enhanced the treatment of mice bearing 3-day-old pulmonary metastases expressing beta-galactosidase. Because IL-10 has been shown to inhibit the functions of key elements of both innate and acquired immune responses, the authors hypothesized that IL-10 might act by inhibiting clearance of the rVV, thus prolonging exposure to the experimental antigen. However, evidence that IL-10 was not acting primarily through such negative regulatory mechanisms included the following: (a) IL-10 also enhanced the therapeutic effectiveness of a recombinant fowlpox virus, which cannot replicate in mammalian cells; (b) Titers of rVV in immunized mice were lower, not higher; and (c) Although IL-10 did not alter levels of anti-vaccinia anti-bodies or natural killer cell activity, rVV-primed mice treated with IL-10 had enhanced vaccinia-specific cytotoxic T-lymphocyte activity. Thus, IL-10 enhanced the function of a recombinant poxvirus-based anti-cancer vaccine and may represent a potential adjuvant in the vaccination against human cancers using recombinant poxvirus-based vaccines.


Subject(s)
Cancer Vaccines/therapeutic use , Interleukin-10/administration & dosage , Neoplasms, Experimental/therapy , Vaccines, Synthetic/therapeutic use , Vaccinia virus/immunology , Adenocarcinoma/therapy , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Colonic Neoplasms/therapy , Female , Humans , Interleukin-10/therapeutic use , Kinetics , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Vaccinia virus/genetics , beta-Galactosidase/genetics , beta-Galactosidase/immunology
12.
J Immunol ; 162(10): 5728-37, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10229805

ABSTRACT

Tumor cells gene-modified to produce GM-CSF potently stimulate antitumor immune responses, in part, by causing the growth and differentiation of dendritic cells (DC). However, GM-CSF-modified tumor cells must be gamma-irradiated or they will grow progressively, killing the host. We observed that 23 of 75 (31%) human tumor lines and two commonly used mouse tumor lines spontaneously produced GM-CSF. In mice, chronic GM-CSF production by tumors suppressed Ag-specific CD8+ T cell responses. Interestingly, an inhibitory population of adherent CD11b(Mac-1)/Gr-1 double-positive cells caused the observed impairment of CD8+ T cell function upon direct cell-to-cell contact. The inhibitory cells were positive for some markers associated with Ag presenting cells, like F4/80, but were negative for markers associated with fully mature DC like DEC205, B7. 2, and MHC class II. We have previously reported that a similar or identical population of inhibitory "immature" APC was elicited after immunization with powerful recombinant immunogens. We show here that these inhibitory cells can be elicited by the administration of recombinant GM-CSF alone, and, furthermore, that they can be differentiated ex vivo into "mature" APC by the addition of IL-4 and GM-CSF. Thus, tumors may be able to escape from immune detection by producing "unopposed" GM-CSF, thereby disrupting the balance of cytokines needed for the maturation of fully functional DC. Further, CD11b/Gr-1 double-positive cells may function as "inhibitory" APC under the influence of GM-CSF alone.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Neoplasms, Experimental/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/drug effects , Antigens, Ly , Cell Communication , Cell Differentiation , Drug Interactions , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Histocompatibility Antigens Class II/isolation & purification , Humans , Interleukin-4/pharmacology , Lymphocyte Culture Test, Mixed , Macrophage-1 Antigen , Mice , Spleen/cytology , Spleen/immunology , Tumor Cells, Cultured/immunology
13.
J Immunol ; 161(10): 5313-20, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9820504

ABSTRACT

Following an infection or immunization, a primary CD8+ T cell response generally rises then falls rapidly before giving rise to a "memory" response. When we immunized mice with recombinant viral immunogens optimized to enhance the lytic capability of CD8+ T cells, we measured a profound depression in Ag-specific effector function after early restimulation. Indeed, a "mirror image" cytolytic capability was observed: the most powerful immunogens, as measured by cytolytic capacity 6 days after immunization, elicited the weakest secondary immune response when evaluated following an additional 6 days after restimulation. To understand the mechanism of this suppression, we examined the fate of splenocytes immunized with a vaccinia virus encoding Ag and IL-2 then restimulated ex vivo. We found that these splenocytes underwent an apoptotic cell death, upon early restimulation, that was not dependent on the engagement of the FasR (CD95). Unlike previously described mechanisms of "propriocidal cell death" and "clonal exhaustion," the cell death we observed was not an inherent property of the CD8+ T cells but rather was due to a population of splenocytes that stained positive for both the Mac-1 and Gr-1 surface markers. Deletion of these cells in vitro or in vivo completely abrogated the observed suppression of cytolytic reactivity of Ag-specific CD8+ T cells. These observations could account for the apparent absence of Ag-specific immune responses after some current vaccination regimens employing powerful immunogens. Finally, our results may shed new light on a mechanism for the suppression of CD8+ T cell responses and its effect on vaccine efficacy and on immune memory.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Macrophage-1 Antigen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Communication/immunology , Cell Membrane/immunology , Cytotoxicity, Immunologic , Female , HeLa Cells , Humans , Immune Tolerance , Immunization , Immunization, Secondary , Interleukin-2/genetics , Lymphocyte Count , Lymphocyte Depletion , Macrophage-1 Antigen/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Vaccinia virus/genetics , Vaccinia virus/immunology , beta-Galactosidase/genetics
14.
J Immunol ; 159(6): 2580-5, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9300676

ABSTRACT

Impaired immune responses are frequently observed in tumor-bearing hosts during progression of tumor growth, but the molecular basis of these functional defects remains unclear. To investigate tumor-induced immunosuppression, we first established that lymphocytes from mice bearing s.c. mammary adenocarcinoma (TS/A) tumors were severely impaired in their ability to generate cellular and humoral Ag-specific responses. Lymphocytes from these mice were then screened for abnormalities in the expression of signal transducing proteins known to be involved in the regulation of cellular immunity. Interestingly, purified T and B cells isolated from immunocompromised tumor-bearing mice displayed a marked decrease in the transcription activators STAT5a and -b at the protein level and to a lesser extent at the mRNA level. By contrast, no change in the expression of STAT1, -3, and -6 or of the TCR itself were detected. The correlation in the loss of T and B cell function with the selective decrease in STAT5a/b expression suggests that regulation of the STAT5 signaling pathway may provide a molecular mechanism for modulating the immune system.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/biosynthesis , Immunocompromised Host , Milk Proteins , Neoplasms, Experimental/immunology , T-Lymphocytes/metabolism , Trans-Activators/biosynthesis , Animals , B-Lymphocytes/immunology , DNA-Binding Proteins/immunology , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , STAT5 Transcription Factor , T-Lymphocytes/immunology , Trans-Activators/immunology
15.
Proc Natl Acad Sci U S A ; 94(7): 3183-8, 1997 Apr 01.
Article in English | MEDLINE | ID: mdl-9096367

ABSTRACT

Recombinant poxviruses encoding tumor-associated antigens (TAA) are attractive as candidate cancer vaccines. Their effectiveness, however, will depend upon expression of the TAA in appropriate antigen-presenting cells. We have used a murine model in which the TAA is beta-galactosidase (beta-gal) and a panel of recombinant vaccinia viruses (rVV) in which beta-gal was expressed under early or late promoters at levels that varied over 500-fold during productive infections in tissue culture cells. Remarkably, only those rVV employing early promoters were capable of prolonging the survival of mice bearing established tumors expressing the model TAA. Late promoters were ineffective regardless of their determined promoter strength. The best results were obtained when beta-gal was regulated by a strong early promoter coupled to a strong late promoter. When a variety of cell types were infected with the panel of viruses in vitro, dendritic cells were found to express beta-gal only under the control of the early promoters even though late promoters were intrinsically more active in other cell types. Furthermore, in a functional assay, dendritic cells infected in vitro with rVV encoding beta-gal regulated by an early promoter activated beta-gal-specific cytotoxic T lymphocytes, whereas similar rVV with a late promoter-regulated gene did not. These data indicate that promoter strength per se is not the most critical quality of a recombinant poxvirus-based tumor vaccine and that the use of promoters capable of driving the production of TAA in "professional" antigen presenting cells may be crucial.


Subject(s)
Antigens/biosynthesis , Cancer Vaccines/immunology , Dendritic Cells/immunology , Vaccines, Synthetic/immunology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Female , Humans , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Promoter Regions, Genetic , Vaccines, Synthetic/genetics , Vaccines, Synthetic/therapeutic use , Vaccinia virus/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
16.
Cancer Res ; 56(12): 2832-6, 1996 Jun 15.
Article in English | MEDLINE | ID: mdl-8665522

ABSTRACT

Activation of T lymphocytes in the absence of a costimulatory signal can result in anergy or apoptotic cell death. Two molecules capable of providing a costimulatory signal, B7-1 (CD80) and B7-2 (CD86), have been shown to augment the immunogenicity of whole-tumor cell vaccines. To explore a potential role for costimulation in the design of recombinant anticancer vaccines, we used lacZ-transduced CT26 as an experimental tumor and beta-galactosidase (beta-gal) as the model tumor antigen. Attempts to augment the function of a recombinant vaccinia virus (rVV) expressing beta-gal by admixture with rVV expressing murine B7-1 were unsuccessful. However, a double recombinant vaccinia virus engineered to express both B7-1 and the model antigen beta-gal was capable of significantly reducing the number of pulmonary metastases when administered to mice bearing tumors established for 3 or 6 days. Most important, the double recombinant vaccinia virus prolonged the survival of tumor-bearing mice. These effects were antigen specific. The related costimulatory molecule B7-2 was found to have a similar, although less impressive enhancing effect on the function of a rVV expressing beta-gal. Thus, the addition of B7-1 and, to a lesser extent, B7-2 to a rVV encoding a model antigen significantly enhanced the therapeutic antitumor effects of these poxvirus-based, therapeutic anticancer vaccines.


Subject(s)
Antigens, CD/immunology , B7-1 Antigen/immunology , Immunotherapy, Active , Lung Neoplasms/prevention & control , Membrane Glycoproteins/immunology , Vaccines, Synthetic/immunology , Vaccinia virus/metabolism , Viral Vaccines/immunology , beta-Galactosidase/immunology , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/immunology , B7-1 Antigen/metabolism , B7-2 Antigen , Female , Lung Neoplasms/secondary , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Survival Analysis , Vaccines, Synthetic/therapeutic use , Vaccinia virus/immunology
17.
J Immunol ; 156(9): 3357-65, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8617961

ABSTRACT

A number of cytokines and costimulatory molecules involved in immune activation have recently been identified including IL-12, a heterodimeric cytokine that supports the development of cell-mediated immunity, and B7-1, a costimulatory molecule involved in the activation of T lymphocytes. We explored the use of these immunomodulants as molecularly defined adjuvants in the function of recombinant anticancer vaccines using a murine model adenocarcinoma, CT26, transduced with a model Ag, beta-galactosidase (beta-gal). Although IL-12 given alone to mice bearing tumors established for 3 days did not have consistent antitumor activity, a profound therapeutic effect was observed when IL-12 administration was combined with a recombinant vaccinia virus (rVV) encoding beta-gal called VJS6. On the basis of the reported synergistic effects of IL-12 and the costimulatory molecule B7-1 (CD80) in vitro, we immunized mice with a double recombinant vaccinia encoding both the model tumor Ag and the costimulatory molecule B7-1, designated B7-1 beta-gal rVV. The adjuvant administration of IL-12 after immunization with this virus significantly enhanced survival in tumor-bearing animals. T cell subset depletions demonstrated that the in vivo activity of IL-12 was largely independent of CD4+ T lymphocytes, whereas the in vivo activity of a B7-1 rVV required both CD4+ and CD8+ T cells to elicit maximal therapeutic effect. To our knowledge, this is the first description of B7-1 and IL-12 cooperation in vivo and represents a novel strategy to enhance the efficacy of recombinant anticancer vaccines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , B7-1 Antigen/biosynthesis , B7-1 Antigen/immunology , Colonic Neoplasms/prevention & control , Interleukin-12/therapeutic use , Vaccines, Synthetic/immunology , Vaccinia virus/immunology , Adjuvants, Immunologic/antagonists & inhibitors , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/mortality , Female , Humans , Immunotherapy, Adoptive , Interleukin-12/antagonists & inhibitors , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Spleen/transplantation , Tumor Cells, Cultured , Vaccines, Synthetic/therapeutic use
18.
Int J Cancer ; 65(6): 847-51, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8631602

ABSTRACT

To examine the in vivo contribution of L-selectin in the sensitization of tumor-specific CTL, we investigated the effects of treatment with the anti-L-selectin monoclonal antibody (MAb) MEL-14 on the immune response to Moloney-murine sarcoma virus (M-MSV)-induced tumors, which exhibit spontaneous regression following generation of a strong virus-specific CTL response. Daily systemic administration of MEL-14 for 10 days to M-MSV-injected mice gave rise to larger sarcomas that persisted for a longer time, compared with those arising in control mice injected with virus only. The enhanced tumor growth could not be attributed to cytotoxic activity on leukocytes by MEL-14 since no reduction in the total cell number was detected in peripheral blood and spleen of MAb-treated mice. Evaluation of the immunological response in MAb-treated animals revealed a strong reduction in the generation of virus-specific CTL precursors (CTLp) in tumor-draining peripheral lymph nodes (PLN) 10 and 15 days after M-MSV injection, while in spleen, where lymphocyte localization is independent of L-selectin expression, CTLp generation was only delayed. By day 20, when tumors had begun to regress, the CTLp number showed a marked increase in both spleen and local PLN, where naive recirculating CTL could now enter because L-selectin was no longer down-regulated or blocked by the injected MAb. Our findings indicate that functional inactivation of L-selectin by MEL-14 treatment prevented migration of naive L-selectin+CTL through high endothelial venules (HEV) and their accumulation in PLN draining the tumor area, thereby precluding the initiation of a tumor-specific CTL response that takes place primarily at this site.


Subject(s)
Antibodies, Monoclonal/pharmacology , L-Selectin/immunology , Lymph Nodes/immunology , Sarcoma, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Neoplasm/immunology , Cell Division/drug effects , Cell Transformation, Viral , Lymph Nodes/drug effects , Mice , Mice, Inbred BALB C , Moloney murine sarcoma virus , Rats , Retroviridae Infections/virology , Sarcoma, Experimental/pathology , Sarcoma, Experimental/virology , T-Lymphocytes, Cytotoxic/drug effects , Tumor Virus Infections/virology
19.
J Immunol ; 156(1): 224-31, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8598466

ABSTRACT

Recombinant adenovirus (rAd), deleted of critical genes that enable viral replication and replaced with genes encoding heterologous proteins, has been shown to be a safe and effective vector in gene therapy studies. To evaluate a potential role for rAd as an immunogen, we used two different replication-defective type 2 rAds encoding the model Ag, beta-galactosidase (beta-gal). To determine whether rAd elicited the kind of immune responses therapeutic in an anti-tumor setting, the beta-gal-expressing adenocarcinoma, CT26.CL25, was used. Splenocytes from BALB/c mice immunized with 1 x 10(7) infectious units (iu) of rAd demonstrated anti-beta-gal activity after in vitro culture with the relevant L(d) beta-gal peptide. Adoptive transfer of these same splenocytes produced dramatic regression of established pulmonary metastases. However, when tumor-bearing mice were treated with 1 x 10(7) iu of rAd, no reduction in established disease was observed even when rAd was given with exogenous IL-2. To increase the viral dose delivered to each animal, we used an E1-E4-deleted rAd that could be grown to much higher titers. Significant reduction occurred with 10-fold more rAd (1 x10(8) iu) was administered. Exogenous IL-2 administration with 1 x 10(8) iu of rAd resulted in augmentation of this anti-tumor effect. These findings demonstrate that when using a nonreplicating virus, the viral dose is directly related to the immune response generated. These data constitute the first reported use of rAd in the treatment of an established experimental cancer and may have implication for the treatment of human cancer.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae/genetics , Antigens, Neoplasm/immunology , Colonic Neoplasms/therapy , Vaccines, Synthetic/immunology , Viral Vaccines/immunology , Adenocarcinoma/immunology , Adenoviridae/immunology , Amino Acid Sequence , Animals , Antigens, Neoplasm/genetics , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Dose-Response Relationship, Immunologic , Female , Immunization, Secondary , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Synthetic/administration & dosage , Viral Vaccines/administration & dosage
20.
Biochem Biophys Res Commun ; 218(1): 344-51, 1996 Jan 05.
Article in English | MEDLINE | ID: mdl-8573158

ABSTRACT

Extracellular ATP (ATPo) induces apoptosis and osmotic lysis in several cell lines. We investigated the role of protein tyrosine kinases (PTKs) and phosphatases (PTPases) in ATPo-induced apoptosis. The PTK inhibitor genistein prevented DNA fragmentation due to ATPo without affecting cell lysis. Comparison of western blot analysis and in vitro kinase assays of anti-phosphotyrosine immunoprecipitates indicated that ATPo activated PTKs whose activity was tightly regulated by PTPases. In fact, an early increase in tyrosine kinase activity was observed after ATPo-treatment and was prevented by specific PTPase inhibitors. In addition, a rapid dephosphorylation of phosphotyrosyl residues on several proteins was detected in ATPo-treated cells. Accordingly, inhibitors of PTPases, but not of serine/threonine phosphatases, were as effective as PTK-inhibitors in blocking ATPo-mediated DNA fragmentation. We describe the early events occurring in ATPo-induced apoptosis and suggest a role for PTPases in cell death.


Subject(s)
Adenosine Triphosphate/pharmacology , Apoptosis/physiology , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , DNA Damage , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genistein , Homeostasis , Isoflavones/pharmacology , Leukemia L1210 , Mast-Cell Sarcoma , Mice , Phosphotyrosine/analysis , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Vanadates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...