Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 12(1): 2198185, 2023.
Article in English | MEDLINE | ID: mdl-37066116

ABSTRACT

The tumor microenvironment (TME) in ovarian cancer (OC) is characterized by immune suppression, due to an abundance of suppressive immune cells populations. To effectively enhance the activity of immune checkpoint inhibition (ICI), there is a need to identify agents that target these immunosuppressive networks while promoting the recruitment of effector T cells into the TME. To this end, we sought to investigate the effect of the immunomodulatory cytokine IL12 alone or in combination with dual-ICI (anti-PD1 + anti-CTLA4) on anti-tumor activity and survival, using the immunocompetent ID8-VEGF murine OC model. Detailed immunophenotyping of peripheral blood, ascites, and tumors revealed that durable treatment responses were associated with reversal of myeloid cell-induced immune suppression, which resulted in enhanced anti-tumor activity by T cells. Single cell transcriptomic analysis further demonstrated striking differences in the phenotype of myeloid cells from mice treated with IL12 in combination with dual-ICI. We also identified marked differences in treated mice that were in remission compared to those whose tumors progressed, further confirming a pivotal role for the modulation of myeloid cell function to allow for response to immunotherapy. These findings provide the scientific basis for the combination of IL12 and ICI to improve clinical response in OC.


Subject(s)
Carcinoma, Ovarian Epithelial , Immunotherapy , Ovarian Neoplasms , Animals , Female , Humans , Mice , Carcinoma, Ovarian Epithelial/drug therapy , Immunosuppression Therapy , Immunotherapy/methods , Interleukin-12/pharmacology , Interleukin-12/therapeutic use , Myeloid Cells/pathology , Ovarian Neoplasms/drug therapy , Tumor Microenvironment
2.
Cancer Immunol Res ; 7(10): 1687-1699, 2019 10.
Article in English | MEDLINE | ID: mdl-31439615

ABSTRACT

Myeloid-derived suppressor cells (MDSC) are induced by and accumulate within many histologically distinct solid tumors, where they promote disease by secreting angiogenic and immunosuppressive molecules. Although IL1ß can drive the generation, accumulation, and functional capacity of MDSCs, the specific IL1ß-induced inflammatory mediators contributing to these activities remain incompletely defined. Here, we identified IL1ß-induced molecules that expand, mobilize, and modulate the accumulation and angiogenic and immunosuppressive potencies of polymorphonuclear (PMN)-MDSCs. Unlike parental CT26 tumors, which recruited primarily monocytic (M)-MDSCs by constitutively expressing GM-CSF- and CCR2-directed chemokines, IL1ß-transfected CT26 produced higher G-CSF, multiple CXC chemokines, and vascular adhesion molecules required for mediating infiltration of PMN-MDSCs with increased angiogenic and immunosuppressive properties. Conversely, CT26 tumors transfected with IL1ß-inducible molecules could mobilize PMN-MDSCs, but because they lacked the ability to upregulate IL1ß-inducible CXCR2-directed chemokines or vascular adhesion molecules, additional PMN-MDSCs could not infiltrate tumors. IL1ß-expressing CT26 increased angiogenic and immunosuppressive factors of tumor-infiltrating MDSCs, as did CT26 tumors individually transfected with G-CSF, Bv8, CXCL1, or CXCL5, demonstrating that mediators downstream of IL1ß could also modulate MDSC functional activity. Translational relevance was indicated by the finding that the same growth factors, cytokines, chemokines, and adhesion molecules responsible for the mobilization and recruitment of PMN-MDSCs into inflammatory CT26 murine tumors were also coordinately upregulated with increasing IL1ß expression in human renal cell carcinoma tumors. These studies demonstrated that IL1ß stimulated the components of a multifaceted inflammatory program that produces, mobilizes, chemoattracts, activates, and mediates the infiltration of PMN-MDSCs into inflammatory tumors to promote tumor progression.


Subject(s)
Carcinoma, Renal Cell/metabolism , Chemokine CXCL1/metabolism , Inflammation , Interleukin-1beta/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Receptors, Virus/metabolism , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Chemokines/immunology , Chemokines/metabolism , Disease Models, Animal , Female , Gene Expression Profiling/methods , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Survival Rate
3.
J Biol Chem ; 290(36): 21962-75, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26205817

ABSTRACT

Unlike other antiapoptotic Bcl-2 family members, Mcl-1 also mediates resistance to cancer therapy by uniquely inhibiting chemotherapy-induced senescence (CIS). In general, Bcl-2 family members regulate apoptosis at the level of the mitochondria through a common prosurvival binding groove. Through mutagenesis, we determined that Mcl-1 can inhibit CIS even in the absence of its apoptotically important mitochondrion-localizing domains. This finding prompted us to generate a series of Mcl-1 deletion mutants from both the N and C termini of the protein, including one that contained a deletion of all of the Bcl-2 homology domains, none of which impacted anti-CIS capabilities. Through subsequent structure-function analyses of Mcl-1, we identified a previously uncharacterized loop domain responsible for the anti-CIS activity of Mcl-1. The importance of the loop domain was confirmed in multiple tumor types, two in vivo models of senescence, and by demonstrating that a peptide mimetic of the loop domain can effectively inhibit the anti-CIS function of Mcl-1. The results from our studies appear to be highly translatable because we discerned an inverse relationship between the expression of Mcl-1 and of various senescence markers in cancerous human tissues. In summary, our findings regarding the unique structural properties of Mcl-1 provide new approaches for targeted cancer therapy.


Subject(s)
Aging/genetics , Cellular Senescence/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Doxorubicin/pharmacology , Female , HCT116 Cells , HT29 Cells , HeLa Cells , Humans , Immunoblotting , Immunohistochemistry , Mice, Nude , Microscopy, Confocal , Models, Molecular , Mutagenesis, Site-Directed , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , RNA Interference , Structure-Activity Relationship , Xenograft Model Antitumor Assays
4.
J Immunol ; 183(8): 5050-8, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19801523

ABSTRACT

Increased expression of gangliosides by different tumor types including renal cell carcinoma (RCC) is thought to contribute to the immune suppression observed in cancer patients. In this study, we report an increase in apoptotic T cells from RCC patients compared with T cells from normal donors that coincided with the detection of T cells staining positive for GM2 and that the apoptosis was predominantly observed in the GM2(+) but not the GM2(-) T cell population. Ganglioside shedding from tumor rather than endogenous production accounts for GM2(+) T cells since there was no detectable level of mRNA for GM2 synthase in RCC patient T cells and in T cells from normal healthy donors after incubation with either purified GM2 or supernatant from RCC cell lines despite their staining positive for GM2. Moreover, reactive oxygen species as well as activated caspase 3, 8, and 9 were predominantly elevated in GM2(+) but not GM2(-) T cells. Similarly, increased staining for GD2 and GD3 but not GD1a was detected with patient T cells with elevated levels of apoptosis in the GD2(+) and GD3(+) cells. These findings suggest that GM2, GD2, and GD3 play a significant role in immune dysfunction observed in RCC patient T cells.


Subject(s)
Carcinoma, Renal Cell/immunology , Gangliosides/immunology , Kidney Neoplasms/immunology , T-Lymphocytes/immunology , Apoptosis/immunology , Carcinoma, Renal Cell/metabolism , Caspases/immunology , Caspases/metabolism , Cell Line, Tumor , Gangliosides/metabolism , Gangliosidoses, GM2/immunology , Gangliosidoses, GM2/metabolism , Humans , Kidney Neoplasms/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism
5.
Cancer Res ; 69(7): 3095-104, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276353

ABSTRACT

We previously elucidated an important role for gangliosides in renal cell carcinoma-mediated T lymphocyte apoptosis, although the mechanism by which they mediated lymphocyte death remained unclear. Here, we show that when added in purified form, GD3 is internalized by activated T cells, initiating a series of proapoptotic events, including the induction of reactive oxygen species (ROS), an enhancement of p53 and Bax accumulation, an increase in mitochondrial permeability, cytochrome c release, and the activation of caspase-9. GD3-induced apoptosis of activated T cells was dose dependent and inhibitable by pretreating the lymphocytes with N-acetylcysteine, cyclosporin A, or bongkrekic acid, emphasizing the essential role of ROS and mitochondrial permeability to the process. Ganglioside-induced T-cell killing was associated with the caspase-dependent degradation of nuclear factor-kappaB-inducible, antiapoptotic proteins, including RelA; this suggests that their loss is initiated only after the cascade is activated and that their disappearance amplifies but not triggers GD3 susceptibility. Resting T cells did not internalize appreciable levels of GD3 and did not undergo any of the proapoptotic changes that characterize activated T lymphocytes exposed to the ganglioside. RelA overexpression endows Jurkat cells with resistance to GD3-mediated apoptosis, verifying the role of the intact transcription factor in mediating protection from the ganglioside.


Subject(s)
Apoptosis/immunology , Gangliosides/immunology , T-Lymphocytes/immunology , Acetylcysteine/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Caspase 8/metabolism , Caspase 9/metabolism , Caspase Inhibitors , Cell Line, Tumor , Cell Membrane Permeability , Cytochromes c/immunology , Cytochromes c/metabolism , Gangliosides/pharmacokinetics , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Jurkat Cells , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Lymphocyte Activation , Mitochondrial Membranes/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
6.
J Immunol ; 180(7): 4687-96, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354192

ABSTRACT

Tumors can promote their own progressive growth by inducing T cell apoptosis. Though previous studies suggested that tumor-mediated T cell killing is receptor dependent, we recently showed that tumor gangliosides also participate, a notion consistent with reports indicating that, in some cell types, gangliosides can activate the intrinsic apoptotic pathway by stimulating reactive oxygen species production, cytochrome c release, and caspase-9 activation. In this study, we used normal peripheral blood T cells, as well as caspase-8-, caspase-9-, and Fas-associated death domain protein-deficient Jurkat cells, to assess whether the death ligands and gangliosides overexpressed by the renal cell carcinoma (RCC) cell line SK-RC-45 can independently stimulate T cell apoptosis as a mechanism of immune escape. Anti-FasL Abs and the glycosylceramide synthase inhibitor 1-phenyl-2-hexadecanoylamino-3-pyrrolidino-1-propanol (PPPP) each partially inhibited the ability of SK-RC-45 to kill cocultured activated T cells; together, as purified molecules, RCC gangliosides and rFasL induced a more extensive mitochondrial permeability transition and greater levels of apoptosis than either agent alone, equivalent to that induced by the FasL- and ganglioside-expressing RCC line itself. rFasL-mediated apoptosis was completely inhibited in caspase-8- and Fas-associated death domain protein-negative Jurkat cells, though apoptosis induced by purified gangliosides remained intact, findings that correlate with the observed partial inhibition of SK-RC-45-induced apoptosis in the Jurkat lines with defective death receptor signaling. Western blot analysis performed on lysates made from wild-type and mutant Jurkat cells cocultured with SK-RC-45 revealed caspase activation patterns and other biochemical correlates which additionally supported the concept that tumor-associated gangliosides and FasL independently activate the caspase cascade in T cells through the intrinsic and extrinsic pathways, respectively.


Subject(s)
Apoptosis , Carcinoma, Renal Cell/metabolism , Receptors, Death Domain/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/metabolism , Cells, Cultured , Coculture Techniques , Enzyme Activation , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Gangliosides/metabolism , Humans , Mitochondria/metabolism , Signal Transduction , fas Receptor/metabolism
7.
J Immunol ; 178(10): 6642-52, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17475896

ABSTRACT

Previous studies from our laboratory demonstrated the role of tumor-derived gangliosides as important mediators of T cell apoptosis, and hence, as one mechanism by which tumors evade immune destruction. In this study, we report that TNF-alpha secreted by infiltrating inflammatory cells and/or genetically modified tumors augments tumor-associated GM2 levels, which leads to T cell death and immune dysfunction. The conversion of weakly apoptogenic renal cell carcinoma (RCC) clones to lines that can induce T cell death requires 3-5 days of TNF-alpha pretreatment, a time frame paralleling that needed for TNF-alpha to stimulate GM2 accumulation by SK-RC-45, SK-RC-54, and SK-RC-13. RCC tumor cell lines permanently transfected with the TNF-alpha transgene are similarly toxic for T lymphocytes, which correlates with their constitutively elevated levels of GM2. TNF-alpha increases GM2 ganglioside expression by enhancing the mRNA levels encoding its synthetic enzyme, GM2 synthase, as demonstrated by both RT-PCR and Southern analysis. The contribution of GM2 gangliosides to tumor-induced T cell death was supported by the finding that anti-GM2 Abs significantly blocked T cell apoptosis mediated by TNF-alpha-treated tumor cells, and by the observation that small interfering RNA directed against TNF-alpha abrogated GM2 synthase expression by TNF-transfected SK-RC-45, diminished its GM2 accumulation, and inhibited its apoptogenicity for T lymphocytes. Our results indicate that TNF-alpha signaling promotes RCC-induced killing of T cells by stimulating the acquisition of a distinct ganglioside assembly in RCC tumor cells.


Subject(s)
Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , G(M2) Ganglioside/biosynthesis , Gene Expression Regulation, Neoplastic/immunology , Kidney Neoplasms/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor-alpha/physiology , Adjuvants, Immunologic/physiology , Antibodies, Blocking/pharmacology , Apoptosis/immunology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , G(M2) Ganglioside/genetics , G(M2) Ganglioside/physiology , Glycosides/biosynthesis , Glycosides/physiology , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Resting Phase, Cell Cycle/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Transfection , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Up-Regulation/immunology
8.
J Immunol ; 178(4): 2278-86, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277133

ABSTRACT

The role of tumor-produced chemokines in the growth of malignancies remains poorly understood. We retrieved an in vivo growing MCA205 fibrosarcoma and isolated tumor cell clones that produce both CXCL9/monokine induced by IFN-gamma (Mig) and CXCL10/IFN-gamma-inducible protein 10 following stimulation with IFN-gamma and clones that produce IFN-gamma-inducible protein 10 but not Mig. The Mig-deficient variants grew more aggressively as cutaneous tumors in wild-type mice than the Mig-producing tumor cells. The growth of Mig-expressing, but not Mig-deficient, tumor cells was suppressed by NK and T cell activity. Transduction of Mig-negative variants to generate constitutive tumor cell production of Mig resulted in T cell-dependent rejection of the tumors and in induction of protective tumor-specific CD8(+) T cell responses to Mig-deficient tumors. The results indicate a critical role for tumor-derived Mig in T cell-mediated responses to cutaneous fibrosarcomas and suggest the loss of Mig expression as a mechanism used by tumor cells to evade these responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, CXC/immunology , Fibrosarcoma/immunology , Gene Expression Regulation, Neoplastic/immunology , Interferon-gamma/immunology , Monokines/immunology , Neoplasm Proteins/immunology , Skin Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chemokine CXCL9 , Chemokines, CXC/biosynthesis , Chemokines, CXC/deficiency , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic/genetics , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Knockout , Monokines/biosynthesis , Monokines/deficiency , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/deficiency , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Escape/genetics , Tumor Escape/immunology
9.
Cancer Res ; 67(1): 362-70, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17210719

ABSTRACT

Cancer patients often exhibit loss of proper cell-mediated immunity and reduced effector T-cell population in the circulation. Thymus is a major site of T-cell maturation, and tumors induce thymic atrophy to evade cellular immune response. Here, we report severe thymic hypocellularity along with decreased thymic integrity in tumor bearer. In an effort to delineate the mechanisms behind such thymic atrophy, we observed that tumor-induced oxidative stress played a critical role, as it perturbed nuclear factor-kappaB (NF-kappaB) activity. Tumor-induced oxidative stress increased cytosolic IkappaBalpha retention and inhibited NF-kappaB nuclear translocation in thymic T cells. These NF-kappaB-perturbed cells became vulnerable to tumor-secreted tumor necrosis factor (TNF)-alpha (TNF-alpha)-mediated apoptosis through the activation of TNF receptor-associated protein death domain-associated Fas-associated protein death domain and caspase-8. Interestingly, TNF-alpha-depleted tumor supernatants, either by antibody neutralization or by TNF-alpha-small interfering RNA transfection of tumor cells, were unable to kill T cell effectively. When T cells were overexpressed with NF-kappaB, the cells became resistant to tumor-induced apoptosis. In contrast, when degradation-defective IkappaBalpha (IkappaBalpha super-repressor) was introduced into T cells, the cells became more vulnerable, indicating that inhibition of NF-kappaB is the reason behind such tumor/TNF-alpha-mediated apoptosis. Curcumin could prevent tumor-induced thymic atrophy by restoring the activity of NF-kappaB. Further investigations suggest that neutralization of tumor-induced oxidative stress and restoration of NF-kappaB activity along with the reeducation of the TNF-alpha signaling pathway can be the mechanism behind curcumin-mediated thymic protection. Thus, our results suggest that unlike many other anticancer agents, curcumin is not only devoid of immunosuppressive effects but also acts as immunorestorer in tumor-bearing host.


Subject(s)
Curcumin/pharmacology , NF-kappa B/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Atrophy , Cell Death/immunology , Cell Line, Tumor , Humans , Mice , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxidative Stress/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , T-Lymphocytes/drug effects , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology
10.
Cancer Res ; 66(13): 6816-25, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16818659

ABSTRACT

Multiple mechanisms have been proposed to account for immune escape by tumors. Although gangliosides have long been known to suppress T-cell immunity, few studies have examined the effect of human tumor-derived gangliosides on immune responses. Here, we show that gangliosides isolated from renal cell carcinoma (RCC) cell lines and clear cell tumor tissue can induce apoptosis in peripheral blood T cells. The RCC tissue-derived gangliosides also suppressed IFN-gamma and, in many cases, interleukin-4 production by CD4+ T cells at concentrations (1 ng/mL-100 pg/mL) well below those that induce any detectable T-cell death (4-20 microg/mL). Additional findings show that GM2 expressed by RCC plays a significant role in promoting T-cell dysfunction. This is supported by the demonstration that all RCC cell lines examined (n = 5) expressed GM2 as did the majority of tumors (15 of 18) derived from patients with clear cell RCC. Furthermore, an antibody specific for GM2 (DMF10.167.4) partially blocked (50-60%) T-cell apoptosis induced by coculturing lymphocytes with RCC cell lines or with RCC tissue-derived gangliosides. DMF10.167.4 also partially blocked the suppression of IFN-gamma production induced by RCC tissue-derived gangliosides, suggesting that GM2 plays a role in down-regulating cytokine production by CD4+ T cells.


Subject(s)
Carcinoma, Renal Cell/immunology , G(M2) Ganglioside/immunology , Kidney Neoplasms/immunology , Apoptosis/immunology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Coculture Techniques , G(M2) Ganglioside/biosynthesis , Humans , Kidney Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Th1 Cells/immunology , Th2 Cells/immunology
11.
J Immunol ; 172(6): 3480-90, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15004148

ABSTRACT

T cells from cancer patients are often functionally impaired, which imposes a barrier to effective immunotherapy. Most pronounced are the alterations characterizing tumor-infiltrating T cells, which in renal cell carcinomas includes defective NF-kappaB activation and a heightened sensitivity to apoptosis. Coculture experiments revealed that renal tumor cell lines induced a time-dependent decrease in RelA(p65) and p50 protein levels within both Jurkat T cells and peripheral blood T lymphocytes that coincided with the onset of apoptosis. The degradation of RelA/p50 is critical for SK-RC-45-induced apoptosis because overexpression of RelA in Jurkat cells protects against cell death. The loss of RelA/p50 coincided with a decrease in expression of the NF-kappaB regulated antiapoptotic protein Bcl-xL at both the protein and mRNA level. The disappearance of RelA/p50 protein was mediated by a caspase-dependent pathway because pretreatment of T lymphocytes with a pan caspase inhibitor before coculture with SK-RC-45 blocked RelA and p50 degradation. SK-RC-45 gangliosides appear to mediate this degradative pathway, as blocking ganglioside synthesis in SK-RC-45 cells with the glucosylceramide synthase inhibitor, PPPP, protected T cells from tumor cell-induced RelA degradation and apoptosis. The ability of the Bcl-2 transgene to protect Jurkat cells from RelA degradation, caspase activation, and apoptosis implicates the mitochondria in these SK-RC-45 ganglioside-mediated effects.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gangliosides/physiology , Kidney Neoplasms/metabolism , NF-kappa B/metabolism , T-Lymphocyte Subsets/metabolism , Adjuvants, Immunologic/biosynthesis , Adjuvants, Immunologic/physiology , Apoptosis/drug effects , Apoptosis/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Caspases/physiology , Cell Line , Cell Line, Tumor , Coculture Techniques , Gangliosides/antagonists & inhibitors , Gangliosides/biosynthesis , Glucosyltransferases/antagonists & inhibitors , Growth Inhibitors/biosynthesis , Growth Inhibitors/physiology , Humans , Jurkat Cells , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , NF-kappa B/physiology , NF-kappa B p50 Subunit , Propanolamines/pharmacology , Pyrrolidines/pharmacology , Transcription Factor RelA
13.
Cancer Res ; 62(9): 2606-10, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11980656

ABSTRACT

The cytokine interleukin (IL)-10 has potent antitumor activity in many model systems when expressed locally at very high levels from the time of tumor transplantation. We now demonstrate that systemic administration of recombinant human IL-10 to animals bearing established highly malignant mammary tumors also leads to significant growth inhibition. We had shown previously that expression of the CXC chemokines Mig (monokine induced by IFN-gamma) and IP-10 (inducible protein 10) is observed in IL-10 transduced but not neo-vector control tumors. We now demonstrate that treatment of IL-10-tumor-bearing mice with antibodies to either chemokine partially reverses the therapeutic effect of IL-10. Tumor growth in animals treated with both antibodies is comparable with that of vector control tumors. Direct transduction of Mig cDNA into the parental tumor cell line before transplantation also results in smaller tumors. This tumor growth inhibition is associated with increased numbers of CD4+ cells, consistent with a T-cell chemoattractant activity for Mig. No change in vascularization, as indicated by CD31+ cells, was observed in either Mig or IL-10-transfected tumors. Thus, an antiangiogenic activity for either cytokine could not be confirmed. Mig and IP-10 are critical to the therapeutic response resulting from high levels of IL-10, and, furthermore, Mig as a single agent also has tumor-inhibitory activity in a model of breast cancer.


Subject(s)
Adenocarcinoma/immunology , Chemokines, CXC/immunology , Intercellular Signaling Peptides and Proteins , Interleukin-10/pharmacology , Mammary Neoplasms, Experimental/immunology , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Amino Acid Sequence , Animals , Chemokine CXCL10 , Chemokine CXCL9 , Chemokines, CXC/genetics , Immunotherapy , Interleukin-10/genetics , Interleukin-10/immunology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Molecular Sequence Data , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...